Method and pharmaceutical composition for treating cancer

Information

  • Patent Grant
  • 11149271
  • Patent Number
    11,149,271
  • Date Filed
    Tuesday, September 4, 2018
    5 years ago
  • Date Issued
    Tuesday, October 19, 2021
    2 years ago
Abstract
A method of treating a subject suffering from cancer includes administering an effective amount of a RNA molecule to the subject, wherein the RNA molecule is isolated or derived from a plant of the genus Taxus. A method of inhibiting growth or proliferation of cancer cells includes contacting cancer cells with the RNA molecule; and a pharmaceutical composition for treating cancer includes the RNA molecule and a pharmaceutically tolerable excipient. Also a double-stranded RNA molecule and a recombinant vector include the double-stranded RNA molecule.
Description
SEQUENCE LISTING

The Sequence Listing file entitled “sequencelisting” having a size of 43,663 bytes and a creation date of Sep. 4, 2018, that was filed with the patent application is incorporated herein by reference in its entirety


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. Nevertheless, there still remains a need to derive effective molecules from various sources such as plants for treatments.



Taxus chinensis (Pilger) Rehd. var. mairei, a species from the family of Taxaceae, is an ornamental evergreen shrub or tree widely distributed in high elevations of China. As an important medicinal plant, it has been exploited for production of small molecular anti-cancer drugs such as paclitaxel which is also called Taxol. However, patients have been found to develop resistance against commonly used drugs including Taxol and therefore there remains a continuing need for new and improved treatments for patients with cancer and for those having resistance against commonly used drugs and/or associated with different complications.


SUMMARY OF THE INVENTION

In a first aspect, the invention provides a method of treating a subject suffering from cancer, said method comprising the step of administering an effective amount of a RNA molecule said subject. The RNA molecule administered according to the invention is isolated or derived from a plant of the genus Taxus.


In an embodiment, the RNA molecule comprises a sequence selected from SEQ ID NO: 1 to SEQ ID NO: 100 or a functional variant or homologue thereof.


Preferably, the RNA molecule of the invention has a sequence length of from about 50 to 200 nucleotides or 10 to 30 base pairs.


In an embodiment, the RNA molecule is a non-coding molecule in particular a transfer RNA molecule.


In an alternative embodiment, the RNA molecule is a double-stranded RNA molecule comprising a sense sequence selected from SEQ ID NO: 1 to SEQ ID NO: 100 or a functional variant or homologue therefore, and a complementary antisense sequence.


In another aspect, the invention provides a method of inhibiting growth or proliferation of cancer cells comprising a step of contacting said cancer cells with an effective amount of a RNA molecule isolated or derived from a plant of the genus Taxus.


In an example embodiment, the cancer cells of the present invention are ovarian cancer cells, liver cancer cells, breast cancer cells, colorectal cancer cells, or lung cancer cells.


In a further aspect, the invention pertains to a pharmaceutical composition for treating cancer. The pharmaceutical composition comprises an RNA molecule and a pharmaceutically tolerable excipient, wherein said RNA molecule is isolated or derived from a plant of the genus Taxus.


Still further, the invention relates to a double-stranded RNA molecule comprising a sense sequence selected from SEQ ID NO: 1 to SEQ ID NO: 100 or a functional variant or homologue therefore, a complementary antisense sequence, and optionally a 3′ overhang.


In another aspect, the invention pertains to a recombinant vector comprising the double-stranded RNA molecule.


The 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 plant of the genus Taxus, or in particular a RNA molecule comprising a sequence selected from SEQ ID NO: 1 to 100. Administration of said RNA molecule is also suitable for inhibiting growth or proliferation of cancer cells. The inventors have found that non-coding RNA molecules isolated from a plant of the genus Taxus, particularly transfer RNA molecules, and RNA molecules derived from Taxus are particularly useful in treatment of cancer. The RNA molecules with a sequence length of about 10 to 200 nucleotides are 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 Taxol-resistant cell lines. Further, the pharmaceutical composition comprising the RNA molecule that is isolated or derived from a plant of the genus Taxus 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. 1 shows gel electrophoresis profiles of RNA molecules from Taxus Chinensis (Pilger) Rehd. var. mairei, including low range RNA markers (denoted as “Ladder”), small RNA molecules, and transfer RNATrP(CCA) in accordance with an example embodiment.



FIG. 2 is a bar chart showing read length distribution of transfer RNAs from Taxus chinensis (Pilger) Rehd. var. mairei in accordance with an example embodiment.



FIG. 3 is a bar chart showing the cytotoxicity of 25 nM RNA molecules tRNAHis(GUG), tRNAGlu(UUC), tRNATrp(CCA), tRNALeu(CAA), or tRNAArg(ACG) from Taxus chinensis (Pilger) Rehd. var. mairei on A2780 cell line, HepG2 cell line, and MCF-7 cell line compared to a control group and a RNAiMAX group where a transfection reagent was added to the cells, in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 4A is a bar chart showing the cell viability of A2780 cells after treatment with a RNA molecule tRNATrP(CCA) at different concentrations, i.e. 0.78 nM, 1.56 nM, 3.13 nM, 6.25 nM, 12.5 nM and 25 nM, compared to a control group and a RNAiMAX group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 4B is a bar chart showing the cell viability of A2780 cells after treatment with Taxol at different concentrations, i.e. 6.25 nM, 12.5 nM, 25 nM, 50 nM and 100 nM, compared to a control group and a RNAiMAX group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 5A is a bar chart showing the cell viability of A2780 cells after treatment with different RNA molecules derived from Taxus Chinensis (Pilger) Rehd. var. mairei with a sequence length of 22 bp at a dose of 50 nM, compared to a control group and a RNAiMAX group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 5B is a bar chart showing the cell viability of A2780 cells after treatment with different RNA molecules derived from Taxus Chinensis (Pilger) Rehd. var. mairei with a sequence length of 19 bp at a dose of 50 nM, compared to a control group and a RNAiMAX group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 5C is a bar chart showing the cell viability of Taxol-resistant A2780 cells (denoted as A2780T cells) after treatment with different RNA molecules derived from Taxus Chinensis (Pilger) Rehd. var. mairei with a sequence length of 22 bp at a dose of 50 nM, compared to a control group and a RNAiMAX group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 5D is a bar chart showing the cell viability of Taxol-resistant A2780T cells after treatment with different RNA molecules derived from Taxus Chinensis (Pilger) Rehd. var. mairei with a sequence length of 19 bp at a dose of 50 nM, compared to a control group and a RNAiMAX group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 5E is a bar chart showing cell viability of HCT-8 cells after treatment with different RNA molecules derived from Taxus Chinensis (Pilger) Rehd. var. mairei with a sequence length of 22 bp at a dose of 50 nM, compared to a control group and a RNAiMAX group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 5F is a bar chart showing cell viability of Taxol-resistant HCT-8 cells (denoted as HCT-8T cells) after treatment with different RNA molecules derived from Taxus Chinensis (Pilger) Rehd. var. mairei with a sequence length of 22 bp at a dose of 50 nM, compared to a control group and a RNAiMAX group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 6A is a bar chart showing the cell viability of A2780 cells after treatment with RNA molecule HC11 at different concentrations, i.e. 6.25 nM, 12.5 nM, 25 nM, 50 nM and 100 nM, compared to a control group and a RNAiMAX group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 6B is a bar chart showing the cell viability A2780 cells after treatment with Taxol at different concentrations, i.e. 6.25 nM, 12.5 nM, 25 nM, 50 nM, 100 nM and 200 nM, compared to a control group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 6C is a bar chart showing the cell viability of Taxol-resistant A2780T cells after treatment with RNA molecule HC11 at different concentrations, i.e. 6.25 nM, 12.5 nM, 25 nM, 50 nM, 100 nM and 200 nM, compared to a control group and a RNAiMAX group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 6D is a bar chart showing the cell viability of Taxol-resistant A2780T cells after treatment with Taxol at different concentrations, i.e. 0.16 μM, 0.8 μM, 4 μM, 20 μM and 100 μM, compared to a control group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 6E is a bar chart showing the cell viability of Taxol-resistant A549T cells after treatment with RNA molecule HC11 at different concentrations, i.e. 6.25 nM, 12.5 nM, 25 nM, 50 nM, 100 nM and 200 nM, compared to a control group and a RNAiMAX group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 6F is a bar chart showing the cell viability of Taxol-resistant A549T cells after treatment with Taxol at different concentrations, i.e. 0.032 μM, 0.16 μM, 0.8 μM, 4 μM, 20 μM and 100 μM, compared to a control group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 6G is a bar chart showing the cell viability of HCT-8 cells after treatment with RNA molecule HC36 at different concentrations, i.e. 6.25 nM, 12.5 nM, 25 nM, 50 nM and 100 nM, compared to a control group and a RNAiMAX group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 6H is a bar chart showing the cell viability of HCT-8 cells after treatment with RNA molecule HC37 at different concentrations, i.e. 6.25 nM, 12.5 nM, 25 nM, 50 nM and 100 nM, compared to a control group and a RNAiMAX group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 6I is a bar chart showing the cell viability of HCT-8 cells after treatment with Taxol at different concentrations, i.e. 50 nM, 100 nM, 200 nM, 300 nM, 400 nM and 500 nM, compared to a control group in accordance with an example embodiment (mean±SD n=3; *, p<0.05, **, p<0.001 vs. vehicle control).



FIG. 7A is a line graph showing the ratio of tumor volume of xenograft implanted A2780 cells in mice over time, in which the mice were treated with RNA molecule HC11 or HC30 with atelocollagen at a dose of 2.4 mg/kg once a week, compared to 1 mg/kg Taxol and a control group.



FIG. 7B is a line graph showing the ratio of weight changes of mice having xenograft implanted A2780 cells, in which the mice were treated with RNA molecule HC11 or HC30 with atelocollagen at a dose of 2.4 mg/kg once a week, compared to 1 mg/kg Taxol and a control group.





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, “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, the forms “a,” “an,” and “the,” are intended to include the singular and plural forms unless the context clearly indicates 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 plant of the genus Taxus. 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: 1 to SEQ ID NO: 100 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′-O-methyladenosine, N6-acetyladenosine, 1-methylinosine, pseudouridine, dihydrouridine, or 2-methylthio-N6-methyladenosine.


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.


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 RNA molecule of the invention comprises a sequence selected from SEQ ID NO: 201 to SEQ ID NO: 225 or a functional variant or homologue thereof; or the RNA molecule comprises SEQ ID NO: 201 to SEQ ID NO: 205 or a functional variant or homologue thereof; or the RNA molecule consists of a sequence selected from SEQ ID NO: 201 to SEQ ID NO: 225 or SEQ ID NO: 201 to SEQ ID NO: 205 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. The RNA molecule comprises a sequence selected from SEQ ID NO: 1 to SEQ ID NO: 100 or a functional variant or homologue thereof, in particular SEQ ID NO: 1 to SEQ ID NO: 36 or a functional variant or homologue thereof; or consists of a sequence selected from SEQ ID NO: 1 to SEQ ID NO: 100, in particular SEQ ID NO: 1 to SEQ ID NO: 36 or a functional variant or homologue thereof. Preferably, the RNA molecule is a double-stranded RNA molecule having a sense sequence selected from SEQ ID NO: 1 to SEQ ID NO: 100 or a functional variant or homologue thereof, and a complementary antisense sequence. The antisense sequence is complementary to the sense sequence and therefore the antisense sequence is preferably selected from SEQ ID NO: 101 to 200 or functional variant or homologue thereof. In a particular embodiment, the double-stranded RNA molecule of the present invention has a sense sequence selected from SEQ ID NO: 1 to SEQ ID NO: 36 or a functional variant or homologue thereof, and a complementary antisense sequence selected from SEQ ID NO: 101 to SEQ ID NO: 136 or a functional variant or homologue thereof. The inventors unexpectedly found that the double-stranded RNA molecules of the present invention are particularly useful in treatment of cancer such as Taxol-resistant cancer as described in detail below.


The RNA molecule of the present invention is preferably isolated or derived from the plant of the genus Taxus. The plant of the genus Taxus includes but is not limited to Taxus baccata, Taxus brevifolia, Taxus chinensis, Taxus chinensis (Pilger) Rehd. var. mairei, Taxus yunanensis, Taxus wallischiana, Taxus cuspidate, Taxus sumatrana, Taxus globasa, Taxus canadensis, and Taxus floridana. The plant of the genus Taxus may be the source of Taxol. In an embodiment, the RNA molecule is isolated or derived from Taxus chinensis.


In more detail, the preferred sequences of the RNA molecules of the present invention are listed in Tables 1 and 2 below. In an embodiment, RNA molecules of SEQ ID NO: 201 to 225 as shown in Table 1 are isolated from a plant of genus Taxus in particular from Taxus chinensis. These sequences are obtained by extraction, RNA isolation and purification of the plant. The inventors determined these RNA molecules are associated with chloroplasts. One possible approach to obtain the RNA molecules from a particular plant Taxus chinensis (Pilger) Rehd. var. mairei 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 Taxus chinensis (Pilger) Rehd. var. mairei


according to the present invention.










SEQ





ID


Length


NO.
tRNA(s)
Sequence (5′ to 3′)
(mer)





201
tRNAHis(GUG)
GCGGACGUAGCCAAGUGGUCCAAAGGC
78




AGUGGAUUGUGAAUCCACCACGCGCGG





GUUCAAUCCCCGUCGUUCGCCCCA






202
tRNAGlu(UUC)
GCCCCUAUCGUCUAGUGGCCCAGGACA
76




UCUCUCUUUCAAGGAGGCAACGGGGAU





UCGAUUUCCCCUAGGGGUACCA






203
tRNATrp(CCA)
GCGCUCUUAGUUCAGUGCGGUAGAACG
75




CAGGUCUCCAAAACCUGAUGCCGUAGG





UUCAAAUCCUACAGAGCGCCA






204
tRNALeu(CAA)
GCCUUGAUGGUGAAAUGGUAGACACGC
84




GAGACUCAAAAUCUCGUGCUAAACAGC





GUGGAGGUUCGAAUCCUCUUCAAGGCA





CCA






205
tRNAArg(ACG)
GGGCCUGUAGCUCAGAGGAUUAGAGCA
77




CGUGGUUGCGAACCACGGUGUCGGGGG





UUCGAAUCCCUCCUCGCCCACCA






206
tRNAAsp(GUC)
GGGAUUGUAGUUCAAUUGGUUAGAGUA
77




CCGCCCUGUCAAGACGGAAGUUGCGGG





UUCGAGCCCCGUCAGUCCCGCCA






207
tRNAAsn(GUU)
UCCUCAGUAGCUCAGUGGUAGAGCGGU
75




CGGCUGUUAACCGAUUGGUCGUAGGUU





CAAAUCCUAUUUGAGGAGCCA






208
tRNACys(GCA)
GGCGACAUAGCCAAGUGGUAAGGCAGG
74




GGACUGCAAAUCCCCCAUCCCCAGUUC





AAAUCCGGGUGUCGCCUCCA






209
tRNAGln(UUG)
GGGGCGUGGCCAAGCGGUAAGGCAACA
75




GGUUUUGGUCCUGUUAUUGCGAAGGUU





CGAAUCCUUUCGUCCCAGCCA






210
tRNAGly(GCC)
GGGUAUUGUUUAAUGGAUAAAAUUUAU
72




UCUUGCCAAGGAUAAGAUGCGGGUUCG





AUUCCCGCUACCCGCCCA






211
tRNAIle(UAU)
AGGGAUAUAACUCAGUAGUAGAGUGUC
75




ACCUUUAUGUGGUGAAAGUCAUCAGUU





CAAACCUGAUUAUCCCUACCA






212
tRNALeu(UAG)
GCCGCCAUGGUGAAAUUGGUAGACACG
83




CUGCUCUUAGGAAGCAGUGCUAGAGCA





UCUCGGUUCGAAUCCGAGUGGUGGCAC





CA






213
tRNALeu(UAA)
GGGGAUAUGGCGGAAUUGGUAGACGCU
90




ACGGACUUAAAAAAUCCGUUGGUUUUA





UAAACCGUGAGGGUUCAAGUCCCUCUA





UCCCCACCA






214
tRNALys(UUU)
GGGUUGUUAACUCAAUGGUAGAGUACU
75




CGGCUUUUAACCGAcGAGUUCCGGGUU





CAAGUCCCGGGCAACCCACCA






215
tRNAMet(CAU)
GCAUCCAUGGCUGAAUGGUCAAAGCAC
76




CCAACUCAUAAUUGGGAAGUCGCGGGU





UCAAUUCCUGCUGGAUGCACCA






216
tRNAMet(CAU)
CGCGGAGUAGAGCAGUUUGGUAGCUCG
77




CAAGGCUCAUAACCUUGAAGUCACGGG





UUCAAAUCCCGUCUCCGCAACCA






217
tRNAPhe(GAA)
GUCGGGAUAGCUCAGUUGGUAGAGCAG
76




AGGACUGAAAAUCCUCGUGUCACCAGU





UCAAAUCUGGUUCCUGGCACCA






218
tRNAPro(UGG)
AGGGAUGUAGCGCAGCUUGGUAGCGCG
77




UUUGUUUUGGGUACAAAAUGUCGCAGG





UUCAAAUCCUGUCAUCCCUACCA






219
tRNAPro(GGG)
CGGAGCAUAACGCAGUUUGGUAGCGUG
77




CCAUCUUGGGGUGAUGGAGGUCGCGGG





UUCAAAUCCUGUUGCUCCGACCA






220
tRNASer(UGA)
GGAGAGAUGGCCGAGUGGUUGAUGGCU
91




CCGGUCUUGAAAACCGGUAUAGUUUUA





AAAACUAUCGAGGGUUCGAAUCCCUCU





CUCUCCUCCA






221
tRNASer(GCU)
GGAGAGAUGGCUGAGCGGACUAAAGCG
91




GUGGAUUGCUAAUCCGUUGUACAGACU





AUCUGUACCGAGGGUUCGAAUCCCUCU





UUCUCCGCCA






222
tRNAThr(UGU)
GCCUGCUUAGCUCAGAGGUUAGAGCAU
76




CGCACUUGUAAUGCGACGGUCAUCGGU





UCGAUCCCGAUAGAAGGCUCCA






223
tRNAThr(GGU)
GCACUUUUAACUCAGUGGUAGAGUAAC
75




GCCAUGGUAAGGCGUAAGUCAUCGGUU





CAAGCCCGAUAAAGGGCUCCA






224
tRNATyr(GUA)
GGGUCGAUGCCCGAGUGGCUAAUGGGG
87




ACGGACUGUAAAUCCGUUGGCAAUAUG





CUUACGCUGGUUCAAAUCCAGCUCGGC





CCACCA






225
tRNAArg(CUC)
GCGUCCAUCGUCUAAUGGAUAGGACAG
75




AGGUCUUCUAAACCUUAGGUAUAGGUU





CAAAUCCUAUUGGACGUACCA









The sense sequences of SEQ ID NO: 1 to SEQ ID NO: 100 and the antisense sequences of SEQ ID NO: 101 to SEQ ID NO: 200 as shown in Table 2 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 Taxus chinensis (Pilger) Rehd. var. mairei. Each of the sense sequences together with the corresponding antisense sequence form a double-stranded RNA molecule. As shown in Table 2, the sense sequence of SEQ ID NO: 1 and the antisense sequence of SEQ ID NO: 101 form a double-stranded RNA molecule with a length of 22 base pairs, and the resultant RNA molecule is denoted as HC11 for easy reference. Similarly, the sense sequence of SEQ ID NO: 2 and the antisense sequence of SEQ ID NO: 102 form a double-stranded RNA molecule with a length of 19 base pairs, and the resultant RNA molecule is denoted as HC20. Other RNA molecules of the present invention are presented in the Table.


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 plant; and the 3′-t group RNA molecules contain a 3′ terminal portion of the corresponding full-length RNA molecules isolated from the plant. In another embodiment, RNA molecules may contain the anticodon loop portion of the corresponding full-length RNA molecules isolated from the plant and referred as anticodon group RNA molecules. The sense sequences of SEQ ID NO: 1 to SEQ ID NO: 100 can be generated by cleavage at different sites on the full-length RNA molecules SEQ ID NO: 201 to 225.


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 2







RNA molecules derived from the sequences in Table 1 through


artificial synthesis according to the present invention.
















SEQ
Sense
SEQ
Antisense






ID
sequence
ID
sequence
Length



Source
Code
NO.
(5′ to 3′)
NO.
(5′ to 3′)
(bp)
Group

















tRNAHis(GUG)
HC11
1
GCGGACGUA
101
UGGACCACU
22
5′-t





GCCAAGUGG

UGGCUACGU







UCCA

CCGC





HC20
2
GCGGACGUA
102
ACCACUUGG
19






GCCAAGUGG

CUACGUCCG







U

C





HC12
3
UCAAUCCCC
103
UGGGGCGAA
22
3′-t





GUCGUUCGC

CGACGGGGA







CCCA

UUGA





HC42
4
AUCCCCGUC
104
UGGGGCGAA
19






GUUCGCCCC

CGACGGGGA







A

U







tRNAGlu(UUC)
HC16
5
GCCCCUAUC
105
UGGGCCACU
22
5′-t





GUCUAGUGG

AGACGAUAG







CCCA

GGGC





HC25
6
GCCCCUAUC
106
GCCACUAGA
19






GUCUAGUGG

CGAUAGGGG







C

C





HC17
7
UCGAUUUCC
107
UGGUACCCC
22
3′-t





CCUAGGGGU

UAGGGGAAA







ACCA

UCGA





HC43
8
AUUUCCCCU
108
UGGUACCCC
19






AGGGGUACC

UAGGGGAAA







A

U







tRNATrp(CCA)
HC30
9
GCGCUCUUA
109
UACCGCACU
22
5′-t





GUUCAGUGC

GAACUAAGA







GGUA

GCGC





HC23
10
GCGCUCUUA
110
CGCACUGAA
19






GUUCAGUGC

CUAAGAGCG







G

C





HC31
11
GUUCAAAUC
111
UGGCGCUCU
22
3′-t





CUACAGAGC

GUAGGAUUU







GCCA

GAAC





HC46
12
CAAAUCCUA
112
UGGCGCUCU
19






CAGAGCGCC

GUAGGAUUU







A

G







tRNALeu(CAA)
HC18
13
GCCUUGAUG
113
UCUACCAUU
22
5′-t





GUGAAAUGG

UCACCAUCA







UAGA

AGGC





HC22
14
GCCUUGAUG
114
ACCAUUUCA
19






GUGAAAUGG

CCAUCAAGG







U

C





HC19
15
UCGAAUCCU
115
UGGUGCCUU
22
3′-t





CUUCAAGGC

GAAGAGGAU







ACCA

UCGA





HC44
16
AAUCCUCUU
116
UGGUGCCUU
19






CAAGGCACC

GAAGAGGAU







A

U







tRNAArg(ACG)
HC32
17
GGGCCUGUA
117
UAAUCCUCU
22
5′-t





GCUCAGAGG

GAGCUACAG







AUUA

GCCC





HC24
18
GGGCCUGUA
118
UCCUCUGAG
19






GCUCAGAGG

CUACAGGCC







A

C





HC33
19
UCGAAUCCC
119
UGGUGGGCG
22
3′-t





UCCUCGCCC

AGGAGGGAU







ACCA

UCGA





HC47
20
AAUCCCUCC
120
UGGUGGGCG
19






UCGCCCACC

AGGAGGGAU







A

U







tRNAAsp(GUC)
HC28
21
GGGAUUGUA
121
UAACCAAUU
22
5′-t





GUUCAAUUG

GAACUACAA







GUUA

UCCC





HC21
22
GGGAUUGUA
122
CCAAUUGAA
19






GUUCAAUUG

CUACAAUCC







G

C





HC29
23
UCGAGCCCC
123
UGGCGGGAC
22
3′-t





GUCAGUCCC

UGACGGGGC







GCCA

UCGA





HC45
24
AGCCCCGUC
124
UGGCGGGAC
19






AGUCCCGCC

UGACGGGGC







A

U







tRNACys(GCA)
HC34
25
GGCGACAUA
125
CUUACCACU
22
5′-t





GCCAAGUGG

UGGCUAUGU







UAAG

CGCC





HC26
26
GGCGACAUA
126
ACCACUUGG
19






GCCAAGUGG

CUAUGUCGC







U

C





HC35
27
UCAAAUCCG
127
UGGAGGCGA
22
3′-t





GGUGUCGCC

CACCCGGAU







UCCA

UUGA





HC48
28
AAUCCGGGU
128
UGGAGGCGA
19






GUCGCCUCC

CACCCGGAU







A

U







tRNAAsn(GUU)
HC36
29
CCUCAGUAG
129
CUCUACCAC
22
5′-t





CUCAGUGGU

UGAGCUACU







AGAG

GAGG





HC27
30
CCUCAGUAG
130
UACCACUGA
19






CUCAGUGGU

GCUACUGAG







A

G





HC37
31
GGUUCAAAU
131
CUCCUCAAA
22
3′-t





CCUAUUUGA

UAGGAUUUG







GGAG

AACC





HC49
32
UCAAAUCCU
132
CUCCUCAAA
19






AUUUGAGGA

UAGGAUUUG







G

A







tRNAMet(CAU)
HC38
33
CGCGGAGUA
133
UACCAAACU
22
5′-t





GAGCAGUUU

GCUCUACUC







GGUA

CGCG





HC40
34
CGCGGAGUA
134
CAAACUGCU
19






GAGCAGUUU

CUACUCCGC







G

G





HC39
35
GGUUCAAAU
135
UUGCGGAGA
22
3′-t





CCCGUCUCC

CGGGAUUUG







GCAA

AACC





HC41
36
UCAAAUCCC
136
UUGCGGAGA
19






GUCUCCGCA

CGGGAUUUG







A

A







tRNAThr(UGU)
HC50
37
GCCUGCUUA
137
CUAACCUCU
22
5′-t





GCUCAGAGG

GAGCUAAGC







UUAG

AGGC





HC52
38
GCCUGCUUA
138
ACCUCUGAG
19






GCUCAGAGG

CUAAGCAGG







U

C





HC51
39
UCGAUCCCG
139
UGGAGCCUU
22
3′-t





AUAGAAGGC

CUAUCGGGA







UCCA

UCGA





HC53
40
AUCCCGAUA
140
UGGAGCCUU
19






GAAGGCUCC

CUAUCGGGA







A

U







tRNAPro(UGG)
HC54
41
AGGGAUGUA
141
UACCAAGCU
22
5′-t





GCGCAGCUU

GCGCUACAU







GGUA

CCCU





HC56
42
AGGGAUGUA
142
CAAGCUGCG
19






GCGCAGCUU

CUACAUCCC







G

U





HC55
43
UCAAAUCCU
143
UGGUAGGGA
22
3′-t





GUCAUCCCU

UGACAGGAU







ACCA

UUGA





HC57
44
AAUCCUGUC
144
UGGUAGGGA
19






AUCCCUACC

UGACAGGAU







A

U







tRNAGly(GCC)
HC58
45
GGGUAUUGU
145
UUUUAUCCA
22
5′-t





UUAAUGGAU

UUAAACAAU







AAAA

ACCC





HC60
46
GGGUAUUGU
146
UAUCCAUUA
19






UUAAUGGAU

AACAAUACC







A

C





HC59
47
UUCGAUUCC
147
UGGGCGGGU
22
3′-t





CGCUACCCG

AGCGGGAAU







CCCA

CGAA





HC61
48
GAUUCCCGC
148
UGGGCGGGU
19






UACCCGCCC

AGCGGGAAU







A

C







tRNATyr(GUA)
HC62
49
GGGUCGAUG
149
UUAGCCACU
22
5′-t





CCCGAGUGG

CGGGCAUCG







CUAA

ACCC





HC64
50
GGGUCGAUG
150
GCCACUCGG
19






CCCGAGUGG

GCAUCGACC







C

C





HC63
51
UCAAAUCCA
151
UGGUGGGCC
22
3′-t





GCUCGGCCC

GAGCUGGAU







ACCA

UUGA





HC65
52
AAUCCAGCU
152
UGGUGGGCC
19






CGGCCCACC

GAGCUGGAU







A

U







tRNALeu(UAA)
HC66
53
GGGGAUAUG
153
CUACCAAUU
22
5′-t





GCGGAAUUG

CCGCCAUAU







GUAG

CCCC





HC68
54
GGGGAUAUG
154
CCAAUUCCG
19






GCGGAAUUG

CCAUAUCCC







G

C





HC67
55
UCAAGUCCC
155
UGGUGGGGA
22
3′-t





UCUAUCCCC

UAGAGGGAC







ACCA

UUGA





HC69
56
AGUCCCUCU
156
UGGUGGGGA
19






AUCCCCACC

UAGAGGGAC







A

U







tRNASer(UGA)
HC70
57
GGAGAGAUG
157
UCAACCACU
22
5′-t





GCCGAGUGG

CGGCCAUCU







UUGA

CUCC





HC72
58
GGAGAGAUG
158
ACCACUCGG
19






GCCGAGUGG

CCAUCUCUC







U

C





HC71
59
UCGAAUCCC
159
UGGAGGAGA
22
3′-t





UCUCUCUCC

GAGAGGGAU







UCCA

UCGA





HC73
60
AAUCCCUCU
160
UGGAGGAGA
19






CUCUCCUCC

GAGAGGGAU







A

U







tRNAGln(UUG)
HC74
61
GGGGCGUG
161
CCUUACCGC
22
5′-t





GCCAAGCG

UUGGCCACG







GUAAGG

CCCC





HC76
62
GGGGCGUG
162
UACCGCUUG
19






GCCAAGCG

GCCACGCCC







GUA

C





HC75
63
UCGAAUCCU
163
UGGCUGGGA
22
3′-t





UUCGUCCCA

CGAAAGGAU







GCCA

UCGA





HC77
64
AAUCCUUUC
164
UGGCUGGGA
19






GUCCCAGCC

CGAAAGGAU







A

U







tRNAArg(CUC)
HC78
65
GCGUCCAUC
165
CUAUCCAUU
22
5′-t





GUCUAAUGG

AGACGAUGG







AUAG

ACGC





HC80
66
GCGUCCAUC
166
UCCAUUAGA
19






GUCUAAUGG

CGAUGGACG







A

C





HC79
67
UCAAAUCCU
167
UGGUACGUC
22
3′-t





AUUGGACGU

CAAUAGGAU







ACCA

UUGA





HC81
68
AAUCCUAUU
168
UGGUACGUC
19






GGACGUACC

CAAUAGGAU







A

U







tRNAMet(CAU)
HC82
69
GCAUCCAUG
169
UUGACCAUU
22
5′-t





GCUGAAUGG

CAGCCAUGG







UCAA

AUGC





HC84
70
GCAUCCAUG
170
ACCAUUCAG
19






GCUGAAUGG

CCAUGGAUG







U

C





HC83
71
UCAAUUCCU
171
UGGUGCAUC
22
3′-t





GCUGGAUGC

CAGCAGGAA







ACCA

UUGA





HC85
72
AUUCCUGCU
172
UGGUGCAUC
19






GGAUGCACC

CAGCAGGAA







A

U







tRNALeu(UAG)
HC86
73
GCCGCCAUG
173
CUACCAAUU
22
5′-t





GUGAAAUUG

UCACCAUGG







GUAG

CGGC





HC88
74
GCCGCCAUG
174
CCAAUUUCA
19






GUGAAAUUG

CCAUGGCGG







G

C





HC87
75
UCGAAUCCG
175
UGGUGCCAC
22
3′-t





AGUGGUGGC

CACUCGGAU







ACCA

UCGA





HC89
76
AAUCCGAGU
176
UGGUGCCAC
19






GGUGGCACC

CACUCGGAU







A

U







tRNALys(UUU)
HC90
77
GGGUUGUUA
177
UCUACCAUU
22
5′-t





ACUCAAUGG

GAGUUAACA







UAGA

ACCC





HC92
78
GGGUUGUUA
178
ACCAUUGAG
19






ACUCAAUGG

UUAACAACC







U

C





HC91
79
UCAAGUCCC
179
UGGUGGGUU
22
3′-t





GGGCAACCC

GCCCGGGAC







ACCA

UUGA





HC93
80
AGUCCCGGG
180
UGGUGGGUU
19






CAACCCACC

GCCCGGGAC







A

U







tRNAPhe(GAA)
HC94
81
GUCGGGAUA
181
CUACCAACU
22
5′-t





GCUCAGUUG

GAGCUAUCC







GUAG

CGAC





HC96
82
GUCGGGAUA
182
CCAACUGAG
19






GCUCAGUUG

CUAUCCCGA







G

C





HC95
83
UCAAAUCUG
183
UGGUGCCAG
22
3′-t





GUUCCUGGC

GAACCAGAU







ACCA

UUGA





HC97
84
AAUCUGGUU
184
UGGUGCCAG
19






CCUGGCACC

GAACCAGAU







A

U







tRNAPro(GGG)
HC98
85
CGGAGCAUA
185
UACCAAACU
22
5′-t





ACGCAGUUU

GCGUUAUGC







GGUA

UCCG





HC100
86
CGGAGCAUA
186
CAAACUGCG
19






ACGCAGUUU

UUAUGCUCC







G

G





HC99
87
UCAAAUCCU
187
UGGUCGGAG
22
3′-t





GUUGCUCCG

CAACAGGAU







ACCA

UUGA





HC101
88
AAUCCUGUU
188
UGGUCGGAG
19






GCUCCGACC

CAACAGGAU







A

U







tRNASer(GCU)
HC102
89
GGAGAGAUG
189
UAGUCCGCU
22
5′-t





GCUGAGCGG

CAGCCAUCU







ACUA

CUCC





HC104
90
GGAGAGAUG
190
UCCGCUCAG
19






GCUGAGCGG

CCAUCUCUC







A

C





HC103
91
UCGAAUCCC
191
UGGCGGAGA
22
3′-t





UCUUUCUCC

AAGAGGGAU







GCCA

UCGA





HC105
92
AAUCCCUCU
192
UGGCGGAGA
19






UUCUCCGCC

AAGAGGGAU







A

U







tRNAThr(GGU)
HC106
93
GCACUUUUA
193
UCUACCACU
22
5′-t





ACUCAGUGG

GAGUUAAAA







UAGA

GUGC





HC108
94
GCACUUUUA
194
ACCACUGAG
19






ACUCAGUGG

UUAAAAGUG







U

C





HC107
95
UCAAGCCCG
195
UGGAGCCCU
22
3′-t





AUAAAGGGC

UUAUCGGGC







UCCA

UUGA





HC109
96
AGCCCGAUA
196
UGGAGCCCU
19






AAGGGCUCC

UUAUCGGGC







A

U







tRNAIle(UAU)
HC110
97
AGGGAUAUA
197
UCUACUACU
22
5′-t





ACUCAGUAG

GAGUUAUAU







UAGA

CCCU





HC112

AGGGAUAUA

ACUACUGAG
19





98
ACUCAGUAG
198
UUAUAUCCC







U

U





HC111

UCAAACCUG

UGGUAGGGA
22
3′-t




99
AUUAUCCCU
199
UAAUCAGGU







ACCA

UUGA





HC113

AACCUGAUU

UGGUAGGGA
19





100
AUCCCUACC
200
UAAUCAGGU







A

U









The inventors unexpectedly found that the RNA molecules isolated or derived from a plant of genus Taxus in particular Taxus chinensis (Pilger) Rehd. var. mairei 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 ovarian cancer, colorectal cancer or lung 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 Taxol, i.e. can be used to treat cancer which is resistant against Taxol. Specifically, the RNA molecules of the present invention can be used to treat Taxol-resistant lung cancer, Taxol-resistant colorectal cancer or Taxol-resistant ovarian 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. A dosage of the RNA molecule such as RNA molecule HC11 (formed by SEQ ID NO: 1 and SEQ ID NO: 101) or HC30 (formed by SEQ ID NO: 9 and SEQ ID NO: 109) may, for example, be at least about 0.1 mg/kg to 5 mg/kg, or about 2 mg/kg to 5 mg/kg, in particular 2.4 mg/kg.


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 Taxol. 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 LipofectamineRNAiMAX 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 plant of the genus Taxus as described above, in particular from Taxus chinensis.


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 plant of the genus Taxus or comprises a sequence selected from SEQ ID NO: 1 to SEQ ID NO: 100 or a functional variant or homologue thereof. The cancer cells are as defined above. Preferably, the cancer cells are ovarian cancer cells, liver cancer cells, breast cancer cells, colorectal cancer cells, or lung cancer cells. The cancer cells may be resistant against currently existing cancer drugs such as but are not limited to Taxol.


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 non-coding molecule preferably a transfer RNA molecule. Preferably, the RNA molecule comprises a sequence selected from SEQ ID NO: 201 to SEQ ID NO: 225 or a functional variant or homologue thereof; or the RNA molecule comprises SEQ ID NO: 201 to SEQ ID NO: 205 or a functional variant or homologue thereof; or the RNA molecule consists of a sequence selected from SEQ ID NO: 201 to SEQ ID NO: 225 or SEQ ID NO: 201 to SEQ ID NO: 205 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 a sense sequence selected from SEQ ID NO: 1 to SEQ ID NO: 100 or a functional variant or homologue thereof, in particular SEQ ID NO: 1 to SEQ ID NO: 36 or a functional variant or homologue thereof; or consists of a sequence selected from SEQ ID NO: 1 to SEQ ID NO: 100, in particular SEQ ID NO: 1 to SEQ ID NO: 36 or a functional variant or homologue thereof. The double-stranded RNA molecule comprises a complementary antisense sequence. 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.


In addition to the above, the present invention pertains to a double-stranded RNA molecule as described above, i.e. comprising a sense sequence selected from SEQ ID NO: 1 to SEQ ID NO: 100 or a functional variant or homologue thereof, and a complementary antisense sequence. In particular, the double-stranded RNA molecule consists of a sense sequence selected from SEQ ID NO: 1 to SEQ ID NO: 100 or a functional variant or homologue thereof, a complementary antisense sequence selected from SEQ ID NO: 101 to SEQ ID NO: 200, and optionally a 3′ overhang. Example embodiments of the double-stranded RNA molecule are presented in Table 2. 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 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: 100 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 plant of the genus Taxus, or in particular a RNA molecule comprising a sequence selected from SEQ ID NO: 1 to 100. 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 Taxol-resistant cell lines.


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


EXAMPLES
Chemicals and Materials

Fresh branches of Taxus chinensis (Pilger) Rehd. var. mairei were collected from Sanming City in the year of 2017 from Fujian Province, China. Cetrimonium bromide (CTAB) and sodium chloride were purchased from-Kingdin Industrial Co., Ltd. (Hong Kong, China). Water-saturated phenol was purchased from Leagene Co., Ltd. (Beijing, China). Chloroform and ethanol were purchased from Anaqua Chemicals Supply Inc. Ltd. (U.S.A.). Isopentanol and guanidinium thiocyanate were purchased from Tokyo Chemical Industry CO., Ltd. (Japan). Tris-HCl and ethylenediaminetetraacetic acid (EDTA) were purchased from Acros Organics (U.S.A), low range ssRNA ladder was purchased from New England Biolabs (Beverly, Mass., U.S.A.). mirVana™ miRNA isolation kit, SYBR gold nucleic acid gel stain and gel loading buffer II were purchased from Thermo Fisher Scientific (U.S.A.). 40% acrylamide/bis solution (19:1), tris/borate/EDTA (TBE), ammonium persulphate (APS) and tetramethylethylenediamine (TEMED) were purchased from Biorad Laboratories Inc. (U.S.A). Taxol-resistance adenocarcinomic human alveolar basal epithelial cell line (A549T) and human ovarian carcinoma cell line (A2780) were purchased from KeyGen Biotech Co. Ltd. (Nanjing, China), human hepatocellular carcinoma cell line (HepG2) and human breast cancer cell line (MCF-7) were purchased from ATCC (Manassas, Va., U.S.A.). Opti-MEM I Reduced Serum Media, Dulbecco's Modified Eagle Medium (DMEM), Minimum Essential Medium (MEM), RPMI Medium 1640, Fetal Bovine Serum (FBS), Penicillin-Streptomycin were purchased from Gibco, (Life Technologies, Auckland, New Zealand). 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) was purchased from Sigma (St. Louis, Mo., U.S.A.).


Example 1
Isolation of RNA Molecules from a Plant of Genus Taxus

Branches of Taxus chinensis (Pilger) Rehd. var. mairei were freshly collected and immediately stored in liquid nitrogen until use. RNAs having a length of 200 nucleotides or below, i.e. small RNAs species, were extracted from Taxus chinensis (Pilger) Rehd. var. mairei by using an optimized CTAB method combined with a commercial small RNA isolation kit, which method is described by Patel, R. S. et al. in Arch Oral Biol 2011, 56 (12), 1506-1513. Briefly, plant tissues were ground into a fine powder in liquid nitrogen and then homogenized in preheated (65° C.) CTAB extraction buffer using a digital dispersing device (IKA, Germany). After incubation for 2 min at 65° C., the tissue lysate was cooled down immediately in an ice bath for 10 min, followed by centrifugation at 12,000×g for 15 min at 4° C. The supernatant was collected and extracted with an equal volume of phenol:chloroform:isopentanol (50:48:1) by vortexing vigorously. Phases were separated at 4° C. by centrifugation at 12,000×g for 15 min and the supernatant was extracted again as described above with chloroform:isopentanol (24:1). The supernatant was collected and mixed with an equal volume of 6 M guanidinium thiocyanate, followed by adding 100% ethanol to a final concentration of 55%. The mixture was passed through a filter cartridge containing a silica membrane, which immobilizes the RNAs. The filter was then washed for several times with 80% (v/v) ethanol solution, and finally all RNAs were eluted with a low ionic-strength solution or RNase-free water. The small RNA species were isolated and enriched by using a mirVana™ miRNA isolation kit following the manufacturer's instruction.


Further, the total tRNAs in the isolated small RNA species were separated by electrophoresis in 6% polyacrylamide TBE gels containing 8 M urea prepared according to the manufacturer's protocol (Biorad, U.S.A.). After staining with SYBR Gold nucleic acid gel stain, polyacrylamide gels were examined using a UV lamp and the region of gels containing total tRNAs were cut off by using a clean and sharp scalpel. FIG. 1 shows gel electrophoresis profiles of small RNA species from Taxus Chinensis (Pilger) Rehd. var. mairei, including low range RNA markers (denoted as “Ladder”), small RNA species, and transfer RNATrP(CCA). The band was sliced into small pieces and the total tRNAs were recovered from the gel by electroelution in a 3 kD molecular weight cut-off dialysis tubing (Spectrum, C.A.) at 100 V for 50 min in 1×TAE buffer. The eluents in the dialysis tubing were recovered and the total tRNAs were desalted and concentrated by using the mirVana™ miRNA isolation kit. The quality and purity of the RNA products were then confirmed using a Nanodrop Spectrophotometer (Thermo Scientific, U.S.A.) and Agilent 2100 Bioanalyzer (Agilent, U.S.A.).


The inventors then constructed the total tRNAs library and performed sequencing. Sequencing libraries were generated by using TruSeq small RNA Library Preparation Kit (Illumina, U.S.A.), followed by a round of adaptor ligation, reverse transcription and PCR enrichment. PCR products were then purified and libraries were quantified on the Agilent Bioanalyzer 2100 system (Agilent Technologies, U.S.A.). The library preparations were sequenced at the Novogene Bioinformatics Institute (Beijing, China) on an Illumina HiSeq platform using the 150 bp paired-end (PE150) strategy to generate over 15 million raw paired reads. 1,729,438 clean reads were obtained by removing low quality regions and adaptor sequences. FIG. 2 is a bar chart showing read length distribution of tRNAs. The tRNA genes were identified by using the tRNAscan-SE 2.0 program (http://lowelab.ucsc.edu/tRNAscan-SE/) and annotated by searching the Nucleotide Collection (nr/nt) database using Basic Local Alignment Search Tool (BLAST) program (https://blast.ncbi.nlm.nih.gov/Blast.cgi). 25 tRNA sequences from Taxus chinensis (Pilger) Rehd. var. mairei were identified and listed in Table 1.


Each of the tRNAs was then isolated from a mixture of small RNAs (<200 mer) from Taxus chinensis (Pilger) Rehd. var. mairei by immobilization of the target tRNAs onto the streptavidin-coated magnetic beads with specific biotinylated capture DNA probes. To bind specific tRNA molecules, a corresponded single stranded DNA oligonucleotide (20 to 45-mer) were synthesized, which was designed based on the sequence information of Illumina sequencing and should be complementary to a unique segment of the target tRNA. Cognate DNA probes were incubated with small RNA mixture for about 1.5 h in annealing buffer and allowed to hybridize to the targeted tRNA molecules in solution at the proper annealing temperatures that were generally 5° C. lower than the melting temperature (Tm). Streptavidin-coated magnetic beads were then added to the mixture and incubated for 30 min at the annealing temperatures. After the hybridized sequences are immobilized onto the magnetic beads via the streptavidin-biotin bond, the biotinylated DNA/tRNA coated beads were separated with a magnet for 1-2 min and washed 3-4 times in washing buffer at 40° C. The magnetic beads were resuspended to a desired concentration in RNase-free water and thereby to release the immobilized tRNA molecules by incubation at 70° C. for 5 min. Accordingly, the isolated and purified tRNA molecules of SEQ ID NO: 201 to 225 were obtained.


Example 2
Synthesis of RNA Molecules

The inventors designed and synthesized RNA molecules having a length of about 19 to 22 bp based on the 25 isolated tRNA sequences in Example 1. 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: 1 to SEQ ID NO: 100 and a complementary antisense sequence selected from SEQ ID NO: 101 to SEQ ID NO: 200, as shown in Table 2, were designed and synthesized by cleavage at different sites on the tRNA sequences in Table 1.


Example 3
Cytotoxic Effect of RNA Molecules on Cancer Cells

A2780, Taxol-resistant A2780, HCT-8, Taxol-resistant HCT-8 and Taxol-resistant A549 cell lines were cultured in RPMI Medium 1640 medium containing 10% FBS and 1% penicillin/streptomycin. HepG2 and MCF-7 cell lines were cultured in Minimum Essential 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 and 2 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, Calif., U.S.A). Dose-response curves were obtained, and the IC50 values were calculated by GraphPad Prism 5 (GraphPad, La Jolla, Calif., USA). Each experiment was carried out for three times. IC50 results were expressed as means±standard deviation.


With reference to FIG. 3, A2780 cells, HepG2 cells and MCF-7 cells were treated with 25 nM RNA molecules of tRNAHis(GUG), tRNAGlu(UUC) tRNATrP(CCA), tRNALeu(CAA), tRNAArg(ACG), i.e. SEQ ID NO: 201 to 205, for 48 h before addition of MTT solution. The cell viability of these cells is compared to a control group and a RNAiMAX group where a transfection reagent was added to the cells. The results show that these RNA molecules are capable of inhibiting the growth and proliferation of ovarian cancer cells, liver cancer cells, and breast cancer cells, whereas the RNA molecules achieve more prominent effect on ovarian and liver cancer cells.



FIG. 4A shows the cytotoxic effect of tRNATrp(CCA), i.e. SEQ ID NO: 203, on A2780 cells. Different concentrations of tRNATrP(CCA) were used, i.e. 0.78 nM, 1.56 nM, 3.13 nM, 6.25 nM, 12.5 nM and 25 nM, and compared to a control group and a RNAiMAX group. It is shown that the IC50 value of tRNATrP(CCA) on ovarian cells in particular A2780 cells is about 14.3 nM. A comparative example using Taxol was conducted. FIG. 4B show the cytotoxic effect of Taxol on A2780 cells.



FIG. 5A and FIG. 5B show the cytotoxic effect of RNA molecules synthesized in Example 2 on A2780 cells, in particular those having sense sequence of SEQ ID NO: 1 to 36. The results show that the RNA molecules designed and synthesized based on the tRNA sequences identified in Example 1 are also effective in inhibiting the growth and proliferation of cancer cells in particular ovarian cancer cells in this example. Further, FIGS. 5C and 5D further demonstrated that the RNA molecules in Example 2 are also capable of inhibiting the growth and/or proliferation of Taxol-resistant A2780 cells. In other words, RNA molecules having sense sequence of SEQ ID NO: 1 to 36 and the complementary antisense sequence are useful in treating cancer which is resistant against Taxol, in particular Taxol-resistant ovarian cancer.



FIG. 5E show the cytotoxic effect of RNA molecules synthesized in Example 2 on HCT-8 cells, in particular those having a sense sequence of SEQ ID NO: 1 to 36 and a complementary antisense sequence. The results show that these RNA molecules are also effective in inhibiting the growth and proliferation of colorectal cancer cells. Further, FIG. 5F further demonstrated that the RNA molecules in Example 2 are also capable of inhibiting the growth and/or proliferation of Taxol-resistant HCT-8 cells. The results also show that the RNA molecules HC18, HC34, HC36, HC37 and HC39 are useful in treating cancer which is resistant against Taxol, in particular Taxol-resistant colorectal cancer.


The inventors then specifically determined the cytotoxic effect and IC50 of RNA molecule HC11 on A2780 cells, at different concentrations, i.e. 6.25 nM, 12.5 nM, 25 nM, 50 nM and 100 nM. As shown in FIG. 6A, the results are compared to a control group and a RNAiMAX group containing a transfecting agent. The results demonstrated that RNA molecule HC11 has a dose-dependent effect on inhibiting the growth and proliferation of ovarian cancer cells. The IC50 of it is 31 nM. A comparative example was conducted using Taxol with results presented in FIG. 6B.


Further, the inhibitory effect of HC11 against Taxol-resistant cancer cells was determined. FIG. 6C shows the cell viability of Taxol-resistant A2780T cells after treatment with HC11 at different concentrations, i.e. 6.25 nM, 12.5 nM, 25 nM, 50 nM, 100 nM and 200 nM, compared to a control group and a RNAiMAX group while FIG. 6D shows a comparative example using Taxol in the treatment. The results demonstrated that RNA molecule HC11 has a dose-dependent effect on inhibiting the growth and proliferation of Taxol-resistant ovarian cancer cells and its IC50 is 32.3 nM.


Meanwhile, FIG. 6E shows the cell viability of Taxol-resistant A549T cells after treatment with HC11 at different concentrations, and FIG. 6F shows the cell viability of Taxol-resistant A549T cells after treatment with Taxol at different concentrations. The results demonstrated that RNA molecule HC11 has a dose-dependent effect on inhibiting the growth and proliferation of Taxol-resistant lung cancer cells with IC50 being 87.3 nM.


Similarly, the inventors specifically determined the cytotoxic effect and IC50 of RNA molecules HC36 and 37 on HCT-8 cells, at different concentrations, i.e. 6.25 nM, 12.5 nM, 25 nM, 50 nM and 100 nM. As shown in FIG. 6G and FIG. 6H, the results are compared to a control group and a RNAiMAX group containing a transfecting agent. The results demonstrated that RNA molecules HC36 and HC37 have dose-dependent effect on inhibiting the growth and proliferation of colorectal cancer cells. The IC50 of HC36, 37 is 8.2 and 9.3 nM. A comparative example was conducted using Taxol with results presented in FIG. 6I.


Based on the above results, it is found that the small tRNA molecules isolated or derived from Taxus chinensis (Pilger) Rehd. var. mairei are highly effective at inhibiting growth and proliferation of cancer cells in vitro. The RNA molecules are also effective against Taxol-resistant cell lines.


Example 4
In Vivo Antitumor Effect of the RNA Molecules

Animal model having xenograft cancer was set. Female BALB/c nude mice (6-8-week old) were purchased from Shanghai SLAC Laboratory Animal Co., Ltd. and maintained at 25° C. with free access to food and water in a special pathogen-free laboratory of the animal environment facilities. The animal experiments were performed in compliance with institutional animal care guidelines and according to committee-approved protocol. To generate tumor xenografts, A2780 cells (4.0×106) were injected subcutaneously in 100 μL of 1640 medium through a 27-gauge needle into the armpit of 8-week-old BALB/c nude mice. After 4-5 weeks after tumors had reached 60-70 mm3, the tumor-bearing nude mice were treated with synthesized tRF with atelocollagen (Koken Co., Ltd., Tokyo, Japan). The concentration of atelocollagen was 1%, and tumor-adjacent injection was performed by one dose of HC11 or HC30 (RNA molecule of SEQ ID NO: 1 or SEQ ID NO: 9) (GenePharma Co., Ltd., Shanghai, China) at concentration of 2.4 mg/kg with atelocollagen once a week. A control group was set up in which vehicle was administered to the mice. A Taxol group for administering 1 mg/kg Taxol to the mice was also set as a comparison. The entire treatment lasted for 28 days.


Tumor diameters were measured at maximum length and maximum width with digital calipers. And the tumor volume was calculated by the formula: volume=(width)2×length/2. The data were statistically analyzed using GraphPad Prism 5 (GraphPad, La Jolla, Calif., USA). The results are presented in FIGS. 7A and 7B. According to the results, HC11 and HC30 are effective in inhibiting the growth of the tumor inside the mice, and maintaining a relative constant body weight. In other words, the RNA molecules of the present invention are effective in treating cancer cells both in vivo and in vitro.

Claims
  • 1. A method of treating a subject suffering from cancer comprising a step of administering an effective amount of a RNA molecule to the subject, wherein the RNA molecule comprises a sense sequence selected from SEQ ID NO: 1 to SEQ ID NO: 100 and a complementary antisense sequence.
  • 2. The method of claim 1, wherein the RNA molecule further comprises 2 mer 3′ overhangs.
  • 3. The method of claim 1, wherein the sense sequence is selected from SEQ ID NO: 1 to SEQ ID NO: 36.
  • 4. The method of claim 1, wherein the RNA molecule comprises at least one modified nucleoside selected from inosine, 1-methyladenosine, 2-methyladenosine, N6-m ethyladenosine, N6-isopentenyladenosine, 2′-O-methyladenosine, N6-acetyladenosine, 1-methylinosine, pseudouridine, dihydrouridine, or 2-methylthio-N6-methyladenosine.
  • 5. The method of claim 1, wherein the cancer is ovarian cancer, liver cancer, breast cancer, colorectal cancer, or lung cancer.
  • 6. The method of claim 1, wherein the cancer is resistant against Taxol.
  • 7. The method of claim 1, wherein the RNA molecule is isolated or derived from Taxus chinensis (Pilger) Rehd. var. mairei.
  • 8. The method of claim 1, wherein the step of administering the RNA molecule to the subject comprises contacting cancer cells of the subject with the RNA molecule.
  • 9. A method of treating a subject suffering from cancer comprising a step of administering an effective amount of a RNA molecule to the subject, wherein the RNA molecule comprises a sequence selected from SEQ ID NO: 201 to SEQ ID NO: 225.
  • 10. 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, wherein the RNA molecule comprises a sense sequence selected from SEQ ID NO: 1 to SEQ ID NO: 100 and a complementary antisense sequence.
  • 11. The method of claim 10, wherein the RNA molecule further comprises 2 mer 3′ overhangs.
  • 12. The method of claim 10, wherein the sense sequence is selected from SEQ ID NO: 1 to SEQ ID NO: 36.
  • 13. The method of claim 10, wherein the RNA molecule comprises at least one modified nucleoside selected from inosine, 1-methyladenosine, 2-methyladenosine, N6-methyladenosine, N6-isopentenyladenosine, 2′-O-methyladenosine, acetyladenosine, 1-methylinosine, pseudouridine, dihydrouridine, or 2-methylthio-N6-methyladenosine.
  • 14. The method of claim 10, wherein the cancer cells are ovarian cancer cells, liver cancer cells, breast cancer cells, colorectal cancer cells, or lung cancer cells.
  • 15. The method of claim 10, wherein the cancer cells are resistant against Taxol.
  • 16. The method of claim 10, wherein the RNA molecule is isolated or derived from Taxus chinensis (Pilger) Rehd. var. mairei.
  • 17. The method of claim 10, wherein the RNA molecule is provided in a composition comprising a gene delivery carrier.
  • 18. 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, wherein the RNA molecule comprises a sequence selected from SEQ ID NO: 201 to SEQ ID NO: 225.
US Referenced Citations (3)
Number Name Date Kind
20020142319 Gorlach Oct 2002 A1
20090239815 Litman Sep 2009 A1
20130137752 Brown May 2013 A1
Non-Patent Literature Citations (8)
Entry
Hao et al. Physiologia Plantarum 146: 388-403 (Year: 2012).
Rushi S. Patel et al, 2011, High resolution of micro RNA signatures in human whole saliva.
Rosemary Kanasty et al, 2013, Delivery materials for siRNA therapeutics.
Guilherme Loss-Morais et al, 2013 , Description of plant tRNA-derived RNA fragments (tRFs) associated with argonaute and identification of their putative targets.
Cell Research (2015) 25:521-524. doi:10.1038/cr.2015.25; published online Feb. 27, 2015, A novel chemopreventive strategy based on therapeutic microRNAs produced in plants.
Rani Goodarzi, et al., (2015), Endogenous tRNA-Derived Fragments Suppress Breast Cancer Progression via YBX1 Displacement.
Zhen Zhou, et a1, 2015, Honeysuckle-encoded atypical microRNA2911 directly targets influenza A viruses.
Veronica Balatti, et al, 2017, Role of the tRNA-Derived Small RNAs in Cancer—New Potential Biomarkers and Target for Therapy.
Related Publications (1)
Number Date Country
20200071695 A1 Mar 2020 US