WO2011067346A1 - Method for determining the efficiency of a cancer treatment - Google Patents

Method for determining the efficiency of a cancer treatment Download PDF

Info

Publication number
WO2011067346A1
WO2011067346A1 PCT/EP2010/068759 EP2010068759W WO2011067346A1 WO 2011067346 A1 WO2011067346 A1 WO 2011067346A1 EP 2010068759 W EP2010068759 W EP 2010068759W WO 2011067346 A1 WO2011067346 A1 WO 2011067346A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
mir
cells
compound
expression
Prior art date
Application number
PCT/EP2010/068759
Other languages
French (fr)
Inventor
Patrick Brest
Paul Hofman
Baharia Mograbi
Original Assignee
Institut National De La Sante Et De La Recherche Medicale (Inserm)
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Institut National De La Sante Et De La Recherche Medicale (Inserm) filed Critical Institut National De La Sante Et De La Recherche Medicale (Inserm)
Publication of WO2011067346A1 publication Critical patent/WO2011067346A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • C12N2310/141MicroRNAs, miRNAs
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2330/00Production
    • C12N2330/10Production naturally occurring
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/178Oligonucleotides characterized by their use miRNA, siRNA or ncRNA

Definitions

  • the present invention relates to a method for determining if a compound is efficient in the treatment of a cancer, in particular if a Histone DeACetylase inhibitor (HDACi) is efficient in the treatment of cancer.
  • HDACi Histone DeACetylase inhibitor
  • Cancer is a disease caused by both genetic and epigenetic reorganizations of the chromatin structure that lead to global alteration of gene expression and play a critical role in tumour initiation and progression (Hanahan et al. (2000) Cell 100(1 ):57-70, Jones et al. (2007) Ce// 128(4):683-92).
  • HDACs nuclear class I histone deacetylases
  • H2A, H2B, H3 and H4 nuclear class I histone deacetylases
  • Histone deacetylation can indicate epigenetic repression by inducing local chromatin condensation and thus plays an important role in transcriptional regulation, cell cycle progression and developmental events.
  • HDACs are generally over-expressed in tumors and promote tumor cell longevity by blocking the transcription of anti-tumoral genes (Bolden et al. (2006) Nat Rev Drug Discov 5(9):769-84).
  • HDACs have thus emerged as molecular targets for the development of enzymatic inhibitors to treat human cancer.
  • Many HDAC inhibitors (HDACi) are currently used in patients as monotherapy (Prince et al. (2009) Clin Cancer Res 15(12):3958-69) because of their activity against human malignancies with biological effects ranging from cell growth arrest, to apoptosis and induction of terminal differentiation.
  • HDAC inhibitors do not share the same efficiency against cancer. In particular, this efficiency may vary depending on the type of tumour to be treated and also from patient to patient.
  • this signature is likely to vary according to tumour type as well as duration of treatment, and inhibitor concentration. Further, these signatures are response signatures rather than prognostic signature of the drug effect.
  • MicroRNAs are a class of endogenously expressed small non-protein- coding RNAs that can post-transcriptionally modulate the expression of hundreds of genes by inhibiting the translation or promoting the degradation of targeted RNA, thereby controlling a wide range of biological functions such as cellular proliferation, differentiation, and apoptosis.
  • microRNAs may function as oncogenes or tumor suppressors, and alteration in microRNA expression may play a critical role in tumorigenesis and clinical outcome (Calin et al. (2006) Nature reviews 6(1 1 ):857-66, Esquela-Kerscher et al. (2006) 6(4):259-69).Thus, several microRNAs were reported to be associated with the clinical outcome of chronic lymphocytic leukaemia, lung and colon adenocarcinoma, and breast, pancreas and thyroid cancers.
  • the present invention arises from the unexpected finding, by the inventors, that in cancer cell lines treated with histone deacetylase inhibitors, the expression of miR-129-5p and miR-133b is specifically increased and that this increase is responsible for the induction of cell cycle arrest which is observed after treatment.
  • the present invention relates to a method, in particular an in vitro method, for determining the anti-cancer activity of a compound comprising:
  • the present invention also relates to at least one nucleic acid
  • ID NO: 2 is liable to induce cell cycle arrest in cancer cell
  • the present invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising as active ingredient substance at least one nucleic acid
  • the present invention also relates to a method for treating cancer in a patient, comprising administering the patient with a therapeutically effective quantity of at least one nucleic acid
  • Figure 1 represents modifications in microRNA miR-133b expression in thyroid treated cells (TPC1 , BcPAP, 8505c, CAL62) after TSA incubation (18h, 330 nmol/L) analyzed by RTQ-PCR. Results represent the fold increase of miR-133b in treated cells versus untreated used as reference (relative quantification, RQ). Stars indicate a significant increase versus reference.
  • Figure 2 represents modifications in microRNA miR-129-5p expression in thyroid treated cells (TPC1 , BcPAP, 8505c, CAL62) after TSA incubation (18h, 330 nmol/L) analyzed by RTQ-PCR. Results represent the fold increase of miR-129-5p in treated cells versus untreated used as reference (relative quantification, RQ). Stars indicate a significant increase versus reference.
  • Figure 3 represents the increase in expression of miR-129-5p and miR-133b in TPC1 cells treated with either TSA (18h, 330 nmol/L, gray bars) or SAHA (18h. 2.5 ⁇ , black bars) HDACi.
  • Results represent the fold increase of miR-129-5p and miR-133b in treated cells versus untreated used as reference (relative quantification, RQ). Stars indicate a significant increase versus reference.
  • Figure 4 represents the modification in expression of miR-129-5p and miR-133b in non-cancerous adenoma (black bars) and hyperplasic (gray bars) cells treated with TSA (18h, 330 nmol/L, gray bars) HDACi. Results represent the fold increase of miR-129-5p and miR-133b in treated cells versus untreated used as reference (relative quantification, RQ).
  • Figure 5 represents cell survival measured as the percentage of MTT viability (vertical axis) of TCP1 cells transfected with either antagomiR-control (amiR-CON) or antagomiR-129-5p (amiR-129-5p) for 48h and incubated without or with TSA (330 nmol/L) for the last 24h. Stars indicate a significant difference of MTT viability with or without TSA. n.s. not significant.
  • Figure 6 represents the percentage subG1 cells among TCP1 cells transfected with either antagomiR-control (amiR-CON) or antagomiR-129-5p (amiR-129-5p) for 48h and incubated without or with TSA (330 nmol/L) for the last 24h. Stars indicate a significant difference of subG1 cells with or without TSA. n.s. not significant.
  • Figure 7 represents GALNT1 expression quantification in cells after transfection of antagomiR and incubation without or with TSA (330nmol/L) for the last 24h.
  • Figure 8 represents cell survival measured as the percentage of viability (vertical axis) of TPC1 (A) or CAL62(B) cells transfected for 24h with either a control miRNA (miRNA-Neg) or miR-129-5p (lipofectamine alone was used as control) and incubated with HAMLET (0.3mg/ml) or etoposide (0.3mM) or staurosporine (0.3 ⁇ ) for 24h. Cell death was followed using a XTT viability assay.
  • the expression "compound” means any molecule or combination of molecules liable to be used in a therapeutic treatment of a patient.
  • a compound which has an "anti-cancer activity” relates to a compound according to the invention for use for treating a patient suffering from cancer.
  • the compound according to the invention is a compound able to alter chromatin structure. More preferably, the compound according to the invention is selected from a group consisting of a histone deacetylase (HDAC) inhibitor and a DNA methyltransferase inhibitor.
  • HDAC histone deacetylase
  • histone deacetylase inhibitors notably encompass hydroxamic acids, such as Trichostatin A (TSA), cyclic tetrapeptides (such as trapoxin B), depsipeptides, benzamides, electrophilic ketones, aliphatic acid compounds such as phenylbutyrate and valproic acid, SAHA (Vorinostat), CAY10433, HDAC1 , CAY10591 , CAY10398, CAY10603, CBHA, HDAC8, M344, Salermide, Sodium Butyrate, Belinostat/PXD101 , MS275, LAQ824/LBH589, CI994, and MGCD0103.
  • Other histone deacetylase inhibitors according to the invention are also described in Wang & Dymock (2009) Expert Opin. Ther. Pat. 19:1727-1757.
  • the HDAC inhibitor according to the invention is selected from the group consisting of Trichostatin A (TSA), SAHA (Vorinostat), CAY10433, HDAC1 , CAY10591 , CAY10398, CAY10603, CBHA, HDAC8, M344, Salermide and Sodium Butyrate. More preferably, the HDAC inhibitor according to the invention is selected from the group consisting of Trichostatin A and SAHA (Vorinostat).
  • Methyltransferases catalyze methylation of DNA at position 5 on cytosine.
  • Methyltransferase inhibitors inhibit the action of methyltransferases.
  • methyltransferase inhibitors As an example of methyltransferase inhibitors according to the invention, one can cite 5- Aza-2'-deoxycytidine (5-Azadc), 5-azacytidine (Vidaza), N-acetyl-S-farnesyl-L- cysteine, S-Farnesyl Thioacetic Acid, N-acetyl-S-geranygeranyl-L-Cysteine, Phenylethanolamine N-methyltransferase, Catechol-O-methyl transferase, Histamine dihydrochloride, Tryptamine hydrochloride, Hydralazine hydrochloride, Procainamide hydrochloride, azacytidine, decitabine, entecapone, chaetocin, SKF 91488 dihydrochloride, S-Far
  • cancer refers to or describe the pathological condition in mammals that is typically characterized by unregulated cell growth.
  • the cancer may be thyroid cancer, skin cancer like melanoma, breast cancer, bladder cancer, uterine/cervical cancer, oesophageal cancer, pancreatic cancer, colon carcinoma, colorectal cancer, kidney cancer, ovarian cancer, prostate cancer, head and neck cancer, lung cancer, non-small cell lung cancer and stomach cancer, B or T cell malignancy, leukaemia.
  • the cancer according to the invention is selected from the group consisting of thyroid cancer, skin cancer, for example melanoma, or colon carcinoma. More preferably, the cancer according to the invention is thyroid cancer or colon carcinoma.
  • the term "patient” or “individual” to be treated is preferably intended for a human or non-human mammal (such as a rodent (mouse, rat), a feline, a canine, or a primate) affected or likely to be affected with cancer.
  • a human or non-human mammal such as a rodent (mouse, rat), a feline, a canine, or a primate
  • the patient is a human.
  • treating means reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition.
  • the treatment of the disorder may consist in destroying or depleting cancer cells. Most preferably, such treatment leads to the complete depletion of cancer cells.
  • the depletion of the cancer cells is obtained by inducing an arrest of the cellular cycle of said cells.
  • the expression "cellular cycle” relates to the series of events that take place in a cell leading to its division and duplication (replication). As is well known, the cycle is divided in four successive phases G1 , S, G2 and M. The expression “an arrest of the cellular cycle” means that this cycle is totally stopped in either one of the phases. Such an arrest leads to cellular death.
  • the "cells from cancer”, “cancer cells” or “cancerous cells” according to the invention can originate from a cancer cell line.
  • the cancer test cells may be the papillary thyroid cell lines CAL62, 8505c, BcPAP and TCP1 , notably described by Schweppe et al. (2008) The Journal of clinical endocrinology and metabolism 93(1 1 ):4331 -41 .
  • the methods and techniques for growing these cells depend on the types of cells which are being tested and are well known by the skilled person.
  • the "cells from cancer” “cancer cells” or “cancerous cells” according to the invention can be cells from a patient affected with cancer, preferably a cancer according to the invention.
  • the patient can optionally be under anti-cancer treatment, preferably under treatment with the compound according to the invention.
  • the cancer cells according to the invention are cancer cells from a patient treated with the compound according to the invention.
  • cancer cells which have been in contact with said compound preferably means that the cancer cells have been contacted with the compound which anti-cancer activity is to be determined under conditions and concentrations which allow the compound to have an effect on the cells. These conditions and concentrations can be easily be determined by the person skilled in the art.
  • the contact between the cancer cells and the compound which anti-cancer activity is to be determined can be an in vitro contact and/or an in vivo contact between the cancer cells and the compound.
  • An in vivo contact according to the invention notably occurs when the cancer cells are cells from a patient treated with said compound.
  • the level of expression of a mi-RNA selected from miR-1 29-5p and/or miR-133b and/or precursors thereof can be either directly detected in the cells or be determined upon a further in vitro contact with compound.
  • cells from a cancer can be contacted with trichostatin A (TSA) at a concentration of about 100ng/ml for about 10 hours.
  • TSA trichostatin A
  • mi-RNA or "microRNA” is used to qualify a class of RNAs generally 10 to 30, in particular 15 to 25 nucleotides long, and play a role in the post-transcriptional regulation of specific genes, by degrading or blocking the translation of the mRNAs resulting from the transcription of these genes.
  • mi-RNAs according to the invention are mi-RNAs which expression is increased after administration of an anti-cancer medicament to a patient, which increase in expression can further be involved in the anticancer effect of this medicament.
  • the mi-RNAs according to the invention are miR-129-5p, in particular Homo sapiens (hsa) miR-129-5p, more particularly represented by SEQ ID NO: 1 , and miR-133b, in particular Homo sapiens (hsa) miR-133b, more particularly represented by SEQ ID NO: 2.
  • miR-129-5p and “miR-133b” notably encompass all the various allelic or polymorphic variants of miR-129-5p, in particular of hsa miR-129-5p, and of miR-133b, in particular of hsa miR-133b, as well as the various orthologous sequences of miR-129-5p and miR-133b, i.e. the species-specific sequences.
  • miR-129-5p and miR-133b are notably described in Griffiths-Jones (2004)
  • a "precursor" of a mi-RNA according to the invention refers to any intermediate in the mi-RNA maturation pathway starting from the transcript obtained from the transcription of a chromosome and ending with the obtaining of a mi-RNA.
  • a precursor according to the invention can be a mi-RNA primary transcripts (pri-miRNA) or a pre-miRNA, as well as a long non-coding RNA, a snoRNA or a transposon which encodes a mi-RNA before processing via the enzyme Dicer.
  • the most preferred precursor of miR-129-5p according to the invention is miR-129-2, in particular represented by SEQ ID NO: 3.
  • the most preferred precursor of miR-133b according to the invention is miR-133b hairpin, in particular represented by SEQ ID NO: 4.
  • miR-129-2 and miR-133b hairpin are notably described in Griffiths-Jones (2004) Nucleic Acids Res 32:D109-D1 1 1 ; Griffiths-Jones et al. (2008) Nucleic Acids Res 36:D154-D158; and the miRBase database available at http://microrna.sanger.ac.uk.
  • Particularly preferred variants of miR-129-5p and of miR-133b may lack one or two nucleotides on the 3' end with respect to their respective consensus sequences, in particular represented by SEQ ID NO: 1 and 2.
  • Other particularly preferred variants of miR-129-5p and of miR-133b may comprise one or two additional nucleotides with respect to their respective consensus sequences, in particular represented by SEQ ID NO: 1 and 2, said additional one or two additional nucleotides being those immediately on the 3' end of the sequence of miR-129-5p and miR-133b within the sequence of their respective precursors, miR-129-2 and miR-133b hairpin, which are themselves notably represented by SEQ ID NO: 3 and 4 respectively.
  • the level of expression of the mi-RNAs in the cells can be measured by any techniques known by the person skilled in the art. Numerous methods are available which allow quantifying a target RNA, for example, methods based on PCR after reverse transcription (RT-PCR) using oligonucleotides which are specific of the target RNA sequences, or alternatively, methods allowing the hybridization of the target RNA, of duplicates or triplicates of the target RNA with probes under stringent conditions.
  • the probes according to the invention are preferably laid down on microarrays.
  • the expression "hybridization under stringent conditions” indicates that the target RNA or the duplicates thereof can specifically bind pairwise, essentially by forming Watson- Crick-type pairs ⁇ e.g.
  • Adequate stringent conditions according to the invention can be easily determined by one of skill in the art.
  • Preferred stringent conditions according to the invention comprise a hybridization step of 10 to 20 hours, preferably 16 hours, at about 40 to 55 °C, preferably 50 °C, under an ionic strength equivalent to that provided by 500 mM to 2 M NaCI, preferably 1 M NaCI.
  • Additional compounds well known to one skilled in the art can also be added such as pH buffers (e.g. Tris or MES), EDTA, Tween, Bovine Serum Albumin, and herring sperm DNA.
  • the "reference value" according to the invention can be a unique value such as a given level of expression of a mi-RNA or of a precursor thereof, alternatively, it can an average between expression levels of a mi-RNA or of a precursor thereof.
  • the reference value can be the level of expression of miR- 129-5p and/or miR-133b and/or precursors thereof in cells which are non-cancer cells, which are preferably of the same type as the cancer cells which have been contacted with the compound which anti-cancer activity is determined according to the invention.
  • the non-cancer cells will be non-cancer papillary thyroid cells.
  • the reference value according to the invention can be the level of expression of miR-129-5p and/or miR-133b and/or precursors thereof in cancer cells which have not been in contact with the compound which anti-cancer activity is determined according to the invention.
  • the expression "comparing the levels of expression” means that the value of the level of expression of miR-129-5p and/or mi-R133b and/or precursors thereof in the cancer cells after contact with the compound which anti-cancer activity is determined according to the invention is compared to the reference value for, respectively, miR-129-5p and/or mi-R133b and/or precursors thereof.
  • determining the anti-cancer activity relates to the identification of compounds likely or unlikely to have an effect in the treatment of cancer.
  • the expression “increase in the level of expression of mi-RNA selected from miR-129-5p and/or mi-R133b and/or precursors thereof” indicatess that the level of expression is higher in the cancer cells after treatment with the compound according to the invention, compared with the reference value.
  • an increase of the level of expression indicates that the compound presents an anti-cancer activity.
  • the level of expression of miR-129-5p or mi-R133b or precursor thereof is increased in the cancer cells contacted with compound when the level of expression is at least 1 .2 fold, more preferably at least 1 .5 fold, and most preferably at least 2 fold over the reference value.
  • the method of the invention may further comprise monitoring the level of expression of a mi-RNA selected from miR-129-5p, miR-133b and precursors thereof over a period of time in cancer cells from a patient treated with the compound which anti-cancer activity is determined according to the invention.
  • the anti-cancer activity of a compound used for treating cancer in a patient treated with the compound is determined according to the invention at consecutive times during the course of the treatment in order to establish a therapeutic follow-up of the patient.
  • the follow-up is typically performed by determining the level of expression of miR-129-5p, miR-133b and precursors thereof according to the invention, at various time intervals, for instance every 2 weeks, 1 month, 2 months, 3 months, 5 months, etc.
  • the reference value according to the invention can also be, in addition to the previous definitions of the term, a previous level of expression of a mi-RNA selected from miR-129-5p and/or miR-133b and/or precursors thereof in cancer cells form the same patient.
  • the therapeutic follow-up according to the invention is started at the same time or upon onset of the treatment of the patient with an anti-cancer compound according to the invention.
  • a decrease in the level of expression of the mi-RNA selected from miR-129-5p and/or miR-133b and/or precursors thereof during the therapeutic follow-up is preferably indicative of a decrease of the anti-cancer activity of the compound thereby indicating a loss of sensitivity of cancer cells the patient to the compound.
  • the nucleic acid for use as a medicament, comprised in a pharmaceutical composition, or administered to a patient can be of any type, it can notably be natural or synthetic, DNA or RNA, single or double stranded.
  • the nucleic acid can comprise non-natural modifications of the bases or bonds, in particular for increasing the resistance to degradation.
  • the nucleic acid is RNA
  • the modifications notably encompass capping its ends or modifying the 2' position of the ribose backbone so as to decrease the reactivity of the hydroxyl moiety, for instance by suppressing the hydroxyl moiety (to yield a 2'-deoxyribose or a 2'-deoxyribose-2'-fluororibose), or substituting the hydroxyl moiety with an alkyl group, such as a methyl group (to yield a 2'-0-methyl-ribose).
  • nucleic acid of the invention comprises or consists of SEQ ID NO: 1 or SEQ ID NO: 2, or of the sequences derived therefrom
  • the nucleic acid of the invention is intended to directly exert its effect on its cellular targets.
  • the nucleic acid is preferably a RNA molecule.
  • the nucleic acid of the invention encodes a nucleic acid comprising or consisting of SEQ ID NO: 1 or SEQ ID NO: 2, or of the sequences derived therefrom
  • the nucleic acid of the invention is intended to be expressed within cells where the nucleic acid it encodes, in particular a RNA molecule, will exert its effect on its cellular targets.
  • the nucleic acid of the invention is preferably a DNA molecule, more preferably a double stranded DNA molecule.
  • the nucleic acid according to the invention preferably also comprises genetic elements ensuring expression of the encoded nucleic acid, in particular a promoter sequence of RNA polymerase II or III.
  • nucleic acid of the invention comprises SEQ ID NO: 1 or SEQ ID NO: 2 or a sequence derived therefrom
  • it is less than 1000 nucleotides long, more preferably less than 100 nucleotides long, and most preferably less than 50 nucleotides long.
  • sequence derived from SEQ ID NO: 1 or SEQ ID NO: 2 by substitution deletion or insertion of at least one nucleotide presents at least 85%, more preferably at least 90%, and most preferably at least 95% identity with the sequence from which it is derived.
  • the percentage of identity between two sequences is obtained by aligning the two sequences so as to maximize the number of positions of each sequence for which the nucleotides are identical and dividing the number of positions of each sequence for which the nucleotides are identical by the number of nucleotides of the longer of the two sequences.
  • the medicament or the pharmaceutical composition according to the invention comprises, or the method of treatment of the invention comprises administering, a RNA molecule consisting of SEQ ID NO: 1 and/or a RNA molecule consisting of SEQ ID NO: 2.
  • the medicament or the pharmaceutical composition according to the invention comprises, or the method of treatment of the invention comprises administering, miR-129-5p or a precursor thereof, such as miR-129-2, as defined above, and/or miR-133b or a precursor thereof, such as miR-133b hairpin, as defined above.
  • the nucleic acid can be associated to one or more pharmaceutically acceptable carriers.
  • the pharmaceutically acceptable carrier be suitable for delivering nucleic acid into cells.
  • Carriers suitable for delivering nucleic acid into cells are well known to one of skill in the art and notably comprise cationic lipids or peptides, nanoparticles and liposomes, optionally linked to moieties, such as antibodies or antibody fragments, having a specificity towards a specific receptor of the target cells, notably cancer cells.
  • Either local or systemic routes can be used for administering the nucleic acid of the invention to a patient.
  • Examples of administration procedures for nucleic acids are notably described in Nguyen et al. ⁇ op. cit.) and Dykxhoorn et al. ⁇ op. cit.)
  • RNA molecule consisting of SEQ ID NO: 1 and/or a RNA molecule consisting of SEQ ID NO: 2 according to the invention are administered to a patient in need thereof in combination or are present together in a same medicament or pharmaceutical composition.
  • the at least one nucleic acid according to the invention is combined in a medicament or a pharmaceutical composition, or administered in combination, with at least one other anti-cancer compound.
  • the other anti-cancer compound can be a medicament useful to treat thyroid cancer or skin cancer like melanoma cancer.
  • medicaments include cytotoxic agent such as gemcitabine, paclitaxel, cisplatin, etoposide and doxorubicin and mechanism-based agents such as HSP90 inhibitor17-AAG and the proteasome inhibitor bortezomib.
  • HAMLET was produced from native purified human milk alpha-lactalbumin on an oleic acid- conditioned ion exchange matrix.
  • Thyroid cancer cell lines BcPAP [papillary, BRAF-V 600 E], TPC1 [papillary, RET- PTC1 ], 8505c [anaplasic, BRAF-V 600 E] and CAL62 [anaplasic, KRAS-G 12 R] have been previously characterized (Ito et al. (1993) Cancer research 53:2940-3; Fabien et al. (1994) Cancer 73:2206-12; and Gioanni et al. (1991 ) Bulletin du cancer 78:1053-62).
  • DMEM Dulbecco's modified Eagle's medium
  • penicillin 100 units/ml
  • thyrocytes were derived from adenoma or hyperplasia thyroid tissue immediately after thyroidectomy at the University of Nice hospital. Patient consent was received and the institutional review board approved the project. Cell dissociation procedures, were similar to those used previously. Briefly, thyroid tissue was cut into small pieces (less than 1 mm diameter), washed in PBS and digested for 3x20 min with a solution consisting of 10 U/ml collagenase, 1 mg/ml dispase (Invitrogen) in PBS. After filtration on 300 ⁇ gaze, cells were seeded at a density of 10 5 per 25cm 2 Flask. Cells were fed with F12/Coon's medium supplemented with 5%FCS twice a week.
  • RNAIMAX Lipofectamin RNAIMAX
  • 5 pmol/L of the specific miRNA 5 pmol/L of the specific miRNA
  • 20 pmol/L of the specific antagomiR Serum was added after 6 h.
  • HDACi were added in fresh complete medium for 24h.
  • cells were lysed for protein or RNA analysis as described in the following parts. Rates of transfection were checked by RT-PCR of the specific transfected miRNA on 7500 thermal cycler (Applied Biosystem).
  • Cell cycle analysis was used in order to analyze both growth arrest (with S phase decrease), and cell death (by increase in subG1 cells) as two endpoints for HDACi activity (Brest et al. (2007) Cancer research 67:1 1327-34).
  • Cells were harvested and fixed in 75% ethanol for 2h. After washes, fixed cells were stained with propidium iodide (2.5 ⁇ 9/ ⁇ , Sigma-Aldrich) and RNase A (250 ⁇ 9/ ⁇ ) at 4°C overnight prior to measurement.
  • Cellular fluorescence was measured using the Facscalibur flow cytometry (Becton Dickinson, San Jose, CA). The single-cell populations were determined on the basis of their fluorescence-intensity values FL2-A and FL2-W.
  • S-Phase percent was calculated as a ratio between (S-phase cells)/(G1 +S+G2-phase cells).
  • Cell death quantification was calculated as a ratio between (SubG1 -phase cells)/(subG1 +G1 +S+G2-phase cells).
  • RNAeasy extraction kit Qiagen, Courtaboeuf, France, according to the manufacturer's instructions. The quality was assessed by measuring both the optical density (Nanodrop, Wilmington, DE), and the electrophoretic mobility using a Bioanalyzer (Agilent Technologies, Santa Clara, CA).
  • the transcriptional profile induced by HDACi was characterized by oligonucleotide microarray analysis of cells treated with HDACi (1 6 h) vs. control cells, by using a microarray containing 2054 mature miRNAs (409 homo sapiens) found in the miRNA registry, as described by Potier et al. (2009) PloS One 4:e671 8. Fragments of tRNAs, snoRNAs, 5S and 5.8S RNA were also printed on the microchip, providing internal positive controls for specific hybridization. The oligonucleotide sequences are available on http://www.microarray.fr/microRNA/.
  • Each oligonucleotide was spotted four times on each slide (2 distinct pairs of spots), in order to reduce positional bias of the fluorescence readout.
  • This miRNA platform has been registered on the Gene Expression Omnibus public data repository (GEO) under the reference GPL471 5.
  • RNA were labeled with Cy3 or Cy5 fluorescent dye using the Ulysis Alexa fluor nucleic acid labeling kit (Amersham Bioscience, Pittsburgh, PA), then miRNA were isolated using the mirVana miRNA isolation Kit (Applied Biosystem). Three independent experiments were made to identify miRNA up regulated or down regulated in cells. For each oligonucleotide microarray, TI F images containing data from each fluorescence channel were quantified with GenePix Pro 6.1 program (Axon Instruments). Normalization was obtained with the software limma from Bioconductor according to the vsn approach described by Huber et al. (2002) Bioinformatics 18 Suppl 1 :S96-1 04.
  • the transcriptional profile induced by miRNA transfection was characterized by oligonucleotide microarray analysis of cells treated with miR-1 33b or miR-1 29-5p vs. control miRNA transfected cells, by using oligonucleotide microarrays containing 25.484 distinct oligonucleotide probes covering most of the known human transcripts (Le Brigand et al. (2006) Nucleic acids research 34:e87). The list of the probes (length 51 bp) is available online htjp:/ www.irilcroarray.jr. Microarrays were printed with a ChipWriter Pro (Bio-Rad) on commercial hydrogel slides (Schott) and processed according to the manufacturer's instructions.
  • ChipWriter Pro Bio-Rad
  • RNA (2 ⁇ g) were amplified with the Amino Allyl MessageAmp aRNA kit (Ambion) according to the manufacturer's instructions. Cy3- and Cy5-labeled aRNA were hybridized on array for 17 h at 62 °C (a dye-swap method has been used). Arrays were then washed with expression wash buffer kit (Agilent) and were scanned with a genePix4000B microarray scanner (Axon instrument).
  • 16-bit TIF images were quantified with the corresponding software (GenePix Pro 6.1 program (Axon Instruments) for the GenePix and Quantarray for the ScanArray. Intra and inter slide normalization of three independent experiments were performed using Global Loess and the quantile methods respectively. Means of ratios from all comparisons were calculated and B test analysis was performed using the Limma package available from Bioconductor (Gentleman et al. (2004) Genome Biol 5:R80). Differentially expressed genes were selected using a Benjamini-Hochberg correction of the p-value for multiple tests, with a positive B value.
  • the sylamer algorithm was used in order to characterize the specificity of miR-133b and miR-129-5p-transfection on global mRNA gene expression. As expected, mRNA harboring the seed sequences of miR-133b and miR-129-5p were specifically downregulated.
  • the human targets of the differentially expressed miRNAs were predicted using public Web-based prediction tools, such as PicTar (http/./pictar.bio.nyu.edu) (Krek et al. (2005) Nature genetics 37:495-500), TargetScan (http://cjenes.rnit.edu/tarqetscan/index.htmj) (Lewis et al. (2005) Cell 120:15-20), and miRBase Targets
  • mRNA from HDACi treated- or miR-133b and miR-129-5p transfected- cells were subjected to reverse transcription-polymerase chain reaction followed by specific Taqman assays (Applied Biosystems). Relative fold changes of expression in induced samples against control were calculated using the comparative Ct (2- ⁇ ) method. Real-time quantization was carried out on the 7500 Fast (Applied Biosystem) thermal cycler, and reagents provided by Applied Biosystem under conditions suggested by the manufacturer.
  • HDACi increase histone acetylation by preventing HDAC activity (Gorisch et al. (2005) J Cell Sci 118:5825-34).
  • the inventors showed that all thyroid cell-lines used were sensitive to TSA (330 nmol/L), as evidenced by the increased histone H4 acetylation.
  • P21 WAF1 (CDKN1 A), a gene down-regulated in cancers (Kastan & Bartek (2004) Nature 432:316-23; Massague (2004) Nature 432:298-306), and previously shown to be restored by HDACi treatment (Brest et al. (2007) Cancer research 67:1 1327-34; Blagosklonny et al. (2002) Molecular cancer therapeutics 1 :937-41 ) was upregulated in HDACi-treated thyroid cells, in agreement with previous studies (Mitsiades et al. (2005) Clin Cancer Res 11 :3958- 65; Luong et al. (2006) Clin Cancer Res 12:5570-7).
  • P21 WAF1 acts as a potent cyclin-dependent kinase inhibitor at the G1 checkpoint causing a rapid G1 -S arrest. Consistently, the inventors found a decrease in S phase in cycling cells (5.8% in TSA-treated cells vs 12.8% unteated cells, p ⁇ 0.05) and an increase of apoptotic cells with fragmented DNA (66% in TSA treated cells vs 2% in control, p ⁇ 0.001 ). Furthermore, DNA damage and apoptotic cell death were confirmed by phosphorylation of histone H2AX and by cleavage of PARP with a higher sensitivity of papillary thyroid cells. Altogether these findings indicate that TSA was able to induce cell cycle arrest and apoptotic cell death in thyroid cancer cells, regardless of the oncogenic status.
  • HDAC inhibitors induce miR-133b and miR-129-5p overexpression
  • miRNAs are candidates that can be considered as putative tumor suppressor. So far, the relationship between miRNA induction and HDACi sensibility of thyroid tumor cells is unknown.
  • miRNA profile was assessed by microarray analyzes after HDACi treatment. BCPAP and 8505c cell lines were treated with TSA overnight (330 nmol/L), miRNA were isolated and their expression level was analyzed on microarray (Table 1 ).
  • the inventors chose to focus their attention on the specific increase in miR-133b and miR-129-5p since their increase was confirmed by RT-PCR after TSA treatment in all the thyroid cell lines used.
  • TSA treatment induced an increase in miR-133b expression ranging from 3-fold (8505c, CAL62) to 5.6-fold (TCP1 ) ( Figure 1 ) and an increase in miR- 129-5p expression ranging from 7-fold (CAL62) to 18-fold (TCP1 ) ( Figure 2) in comparison with untreated cells used as reference.
  • SAHA another HDACi
  • TCP1 TCP1 cells that showed similar fold of increase as TSA
  • Figure 3 the highest induction was found in papillary cell lines and correlated to the higher sensitivity in cell death. At that stage it was of interest to ascertain the specificity of HDACi-induced responses (viability, miRNA expression) on normal healthy thyrocytes.
  • miR-133b and miR-129-5p overexpressions are sufficient for cell growth arrest
  • these miRNAs were transfected into TCP1 cells. After 48 h, transfected cells were analyzed for cell cycle distribution. Introduction of miR-133b and/or miR-129-5p caused a significant reductions of population of cells entering in S-Phase (5.1 ⁇ 0.5% for miR-133b, 3.3 ⁇ 0.4% for miR-129-5p, p ⁇ 0.05), similarly to HDACi treatment (TSA, 330nM, 24h). Cells transfected with negative control miR-CON1 showed the same number of cells in S-Phase (9.7 ⁇ 0.8%) than non-transfected cells. These data underscore that expression of miR-133b and miR-129-5p are sufficient alone for cell growth arrest in an HDACi-treatment similar manner.
  • miR-133b and miR-129-5p overexpressions induce decrease expression of cyclin B1 and cyclin F
  • cyclin B1 CCNB1
  • cyclin F CCNF
  • miR-133b and miR-129-5p are not necessary for cell growth arrest
  • HDACi-induced overexpression of miR-129-5p and miR-133b participates to cell cycle arrest via a specific decrease in cyclin B1 and F expression in thyroid cancer cells.
  • miR-129-5p is sufficient for HDACi induced cell death
  • HDACi has been shown to induce cell cycle arrest followed by cell death in thyroid cancer cells.
  • the role of both miR-133b and miR-129-5p was investigated in this context.
  • miR-129-5p-transfected TCP1 cells showed an increased cell death, suggesting that this miRNA is cytotoxic for cancer cells.
  • the inventors then analyzed pan-genomic microarray of miR-129-5p-transfected TPC1 cells on cell death-involved genes expression. Data collected from three independent biological experiments, revealed that a total of 224 transcripts were significantly modulated (102 up and 122 down-regulated, p ⁇ 0.01 ) following pre-miR-129-5p transfection when compared to the control condition. Analysis of this signature with Ingenuity PathwayTM software indicated a significant enrichment for "Molecular functions" with terms such as “Cellular Compromise", “Cellular Growth and Proliferation", “Cell Cycle” or "Cell Death".
  • the inventors then looked for potential over representation of miR-129-5p-predicted direct targets among the downregulated transcripts (cut-offs equal to 8.0 for the log 2 (signal), to -1 for the log 2 (ratio), and to 0.01 for the adjusted p-value) and isolated 42 transcripts corresponding to putative direct targets (Table 6).
  • GALNT1 GALNT1
  • BCL6 B- cell CLL/lymphoma 6, 2 "2 4 ⁇ -5.3 fold
  • PDCD2 pro- apoptotic protein
  • TCP1 cells were transfected 24h either with antagomiR-CON1 (control) or antagomiR-129-5p and then incubated with TSA (330 nmol/L, 24h). Moreover, antagomiR-133b alone or combined with antagomiR-129-5p were also used to block TSA-induced cell death.
  • miR-129-5p by blocking expression of miR-129-5p or miR-133b in cells, the inventors showed a prevention of HDACi-induced cell death, thereby showing that miR-129-5p and miR-133b are mandatory for HDACi killing effect on tumor cells. 2.8. miR-129-5p promotes drug-induced cell death
  • HDACi have previously been shown to induce cell death in response to other cancer drugs like etoposide, cisplatin or HAMLET.
  • miR-129-5p is a shared target
  • Combination with miR-129-5p increased the sensitivity to HAMLET, staurosporine or etoposide ( Figure 8).
  • the loss of viability in response to HAMLET (0.3 mg/ml, 24h) was 40% in miR-CTL transfected control cells, but increased to 70% in tumor cells transfected with miR-129-5p.
  • a similar increase in cell death was obtained in cells treated with staurosporine or etoposide.
  • miR-129-5p affects tumor cell viability per se, it also improves the sensitivity to different cancer drugs through effects on HDAC function.
  • Table 1 Profile of altered miRNA in TSA-treated BCPAP and 8505C cell lines (Fold change > ⁇ 0.75, Adjusted p Value ⁇ 5 10 "3 )
  • Table 2 Putative targeted genes by miR-129-5p found downregulated pangenomic microarray: GO Pathway, gene list
  • Table 3 Putative targeted genes by miR-133b found downregulated in pangenomic microarray: GO Pathway, gene list
  • CDK6 1021 9.8 -2.2

Abstract

The present invention relates to an in vitro method for determining the anti-cancer activity of a compound comprising: i) measuring the level of expression of a mi-RNA selected from miR-129-5p, miR-133b and precursors thereof in cancer cells which have been in contact with said compound; ii) comparing the level of expression with a reference value; iii) determining therefrom the anti-cancer activity of the compound.

Description

Method for determining the efficiency of a cancer treatment
Field of the invention
The present invention relates to a method for determining if a compound is efficient in the treatment of a cancer, in particular if a Histone DeACetylase inhibitor (HDACi) is efficient in the treatment of cancer.
Background of the invention
Cancer is a disease caused by both genetic and epigenetic reorganizations of the chromatin structure that lead to global alteration of gene expression and play a critical role in tumour initiation and progression (Hanahan et al. (2000) Cell 100(1 ):57-70, Jones et al. (2007) Ce// 128(4):683-92).
As a central actor of epigenetic remodeling, nuclear class I histone deacetylases (HDACs) are responsible for the deacetylation of lysine residues on the N-terminal part of the core histones (H2A, H2B, H3 and H4). Histone deacetylation can indicate epigenetic repression by inducing local chromatin condensation and thus plays an important role in transcriptional regulation, cell cycle progression and developmental events. Interestingly, HDACs are generally over-expressed in tumors and promote tumor cell longevity by blocking the transcription of anti-tumoral genes (Bolden et al. (2006) Nat Rev Drug Discov 5(9):769-84). HDACs have thus emerged as molecular targets for the development of enzymatic inhibitors to treat human cancer. Many HDAC inhibitors (HDACi) are currently used in patients as monotherapy (Prince et al. (2009) Clin Cancer Res 15(12):3958-69) because of their activity against human malignancies with biological effects ranging from cell growth arrest, to apoptosis and induction of terminal differentiation.
However, all HDAC inhibitors do not share the same efficiency against cancer. In particular, this efficiency may vary depending on the type of tumour to be treated and also from patient to patient.
It is therefore highly desirable to implement methods enabling to evaluate the efficiency of HDAC inhibitors in order to classify patients and tumours into groups that are responsive to the drug. In various studies biomarkers have been assessed to elucidate how HDAC inhibitors exert their effect. Some sets of genes were identified in cancer cells from patient, the expression of which being indicative of response to HDACi treatment (Stimson et al. (2009) Cancer Letters 280:177-183).
However, this signature is likely to vary according to tumour type as well as duration of treatment, and inhibitor concentration. Further, these signatures are response signatures rather than prognostic signature of the drug effect.
Thus, to date, no biomarker has been found that could help predict clearly the tumour response to a HDACi (Stimson et al. (2009) Cancer Letters 280:177- 183).
Accordingly, there is a need for such a method.
MicroRNAs are a class of endogenously expressed small non-protein- coding RNAs that can post-transcriptionally modulate the expression of hundreds of genes by inhibiting the translation or promoting the degradation of targeted RNA, thereby controlling a wide range of biological functions such as cellular proliferation, differentiation, and apoptosis. Recent evidence indicates that microRNAs may function as oncogenes or tumor suppressors, and alteration in microRNA expression may play a critical role in tumorigenesis and clinical outcome (Calin et al. (2006) Nature reviews 6(1 1 ):857-66, Esquela-Kerscher et al. (2006) 6(4):259-69).Thus, several microRNAs were reported to be associated with the clinical outcome of chronic lymphocytic leukaemia, lung and colon adenocarcinoma, and breast, pancreas and thyroid cancers.
Summary of the invention
The present invention arises from the unexpected finding, by the inventors, that in cancer cell lines treated with histone deacetylase inhibitors, the expression of miR-129-5p and miR-133b is specifically increased and that this increase is responsible for the induction of cell cycle arrest which is observed after treatment.
Thus, the present invention relates to a method, in particular an in vitro method, for determining the anti-cancer activity of a compound comprising:
i) measuring the level of expression of a mi-RNA selected from miR-129-5p and/or miR-133b and/or precursors thereof in cancer cells which have been in contact with said compound; ii) comparing the level of expression with a reference value;
iii) determining therefrom the anti-cancer activity of the compound.
The present invention also relates to at least one nucleic acid
(i) comprising or consisting of, or
(ii) encoding a nucleic acid comprising or consisting of,
a sequence selected from the group consisting of:
1 ) SEQ ID NO: 1 and SEQ ID NO: 2 and
2) a sequence derived from SEQ ID NO: 1 and SEQ ID NO: 2 by substitution, deletion or insertion of at least one nucleotide, provided that a nucleic acid consisting of the sequence derived from SEQ ID NO: 1 and SEQ
ID NO: 2 is liable to induce cell cycle arrest in cancer cell,
for use as a medicament, in particular for treating cancers.
The present invention also relates to a pharmaceutical composition comprising as active ingredient substance at least one nucleic acid
(i) comprising or consisting of, or
(ii) encoding a nucleic acid comprising or consisting of,
a sequence selected from the group consisting of:
1 ) SEQ ID NO: 1 and SEQ ID NO: 2 and
2) a sequence derived from SEQ ID NO: 1 and SEQ ID NO: 2 by substitution, deletion or insertion of at least one nucleotide, provided that a nucleic acid consisting of the sequence derived from SEQ ID NO: 1 and SEQ ID NO: 2 is liable to induce cell cycle arrest in cancer cell,
optionally in association with a pharmaceutically acceptable carrier.
The present invention also relates to a method for treating cancer in a patient, comprising administering the patient with a therapeutically effective quantity of at least one nucleic acid
(i) comprising or consisting of, or
(ii) encoding a nucleic acid comprising or consisting of,
a sequence selected from the group consisting of:
1 ) SEQ ID NO: 1 and SEQ ID NO: 2 and
2) a sequence derived from SEQ ID NO: 1 and SEQ ID NO: 2 by substitution, deletion or insertion of at least one nucleotide, provided that a nucleic acid consisting of the sequence derived from SEQ ID NO: 1 and SEQ ID NO: 2 is liable to induce cell cycle arrest in cancer cell.
Brief description of the drawings
Figure 1 represents modifications in microRNA miR-133b expression in thyroid treated cells (TPC1 , BcPAP, 8505c, CAL62) after TSA incubation (18h, 330 nmol/L) analyzed by RTQ-PCR. Results represent the fold increase of miR-133b in treated cells versus untreated used as reference (relative quantification, RQ). Stars indicate a significant increase versus reference.
Figure 2 represents modifications in microRNA miR-129-5p expression in thyroid treated cells (TPC1 , BcPAP, 8505c, CAL62) after TSA incubation (18h, 330 nmol/L) analyzed by RTQ-PCR. Results represent the fold increase of miR-129-5p in treated cells versus untreated used as reference (relative quantification, RQ). Stars indicate a significant increase versus reference.
Figure 3 represents the increase in expression of miR-129-5p and miR-133b in TPC1 cells treated with either TSA (18h, 330 nmol/L, gray bars) or SAHA (18h. 2.5 μΜ, black bars) HDACi. Results represent the fold increase of miR-129-5p and miR-133b in treated cells versus untreated used as reference (relative quantification, RQ). Stars indicate a significant increase versus reference.
Figure 4 represents the modification in expression of miR-129-5p and miR-133b in non-cancerous adenoma (black bars) and hyperplasic (gray bars) cells treated with TSA (18h, 330 nmol/L, gray bars) HDACi. Results represent the fold increase of miR-129-5p and miR-133b in treated cells versus untreated used as reference (relative quantification, RQ).
Figure 5 represents cell survival measured as the percentage of MTT viability (vertical axis) of TCP1 cells transfected with either antagomiR-control (amiR-CON) or antagomiR-129-5p (amiR-129-5p) for 48h and incubated without or with TSA (330 nmol/L) for the last 24h. Stars indicate a significant difference of MTT viability with or without TSA. n.s. not significant.
Figure 6 represents the percentage subG1 cells among TCP1 cells transfected with either antagomiR-control (amiR-CON) or antagomiR-129-5p (amiR-129-5p) for 48h and incubated without or with TSA (330 nmol/L) for the last 24h. Stars indicate a significant difference of subG1 cells with or without TSA. n.s. not significant. Figure 7 represents GALNT1 expression quantification in cells after transfection of antagomiR and incubation without or with TSA (330nmol/L) for the last 24h.
Figure 8 represents cell survival measured as the percentage of viability (vertical axis) of TPC1 (A) or CAL62(B) cells transfected for 24h with either a control miRNA (miRNA-Neg) or miR-129-5p (lipofectamine alone was used as control) and incubated with HAMLET (0.3mg/ml) or etoposide (0.3mM) or staurosporine (0.3μΜ) for 24h. Cell death was followed using a XTT viability assay.
Detailed description of the invention
As intended herein, the expression "compound" means any molecule or combination of molecules liable to be used in a therapeutic treatment of a patient. A compound which has an "anti-cancer activity" relates to a compound according to the invention for use for treating a patient suffering from cancer. Preferably, the compound according to the invention is a compound able to alter chromatin structure. More preferably, the compound according to the invention is selected from a group consisting of a histone deacetylase (HDAC) inhibitor and a DNA methyltransferase inhibitor.
Examples of histone deacetylase inhibitors according to the invention notably encompass hydroxamic acids, such as Trichostatin A (TSA), cyclic tetrapeptides (such as trapoxin B), depsipeptides, benzamides, electrophilic ketones, aliphatic acid compounds such as phenylbutyrate and valproic acid, SAHA (Vorinostat), CAY10433, HDAC1 , CAY10591 , CAY10398, CAY10603, CBHA, HDAC8, M344, Salermide, Sodium Butyrate, Belinostat/PXD101 , MS275, LAQ824/LBH589, CI994, and MGCD0103. Other histone deacetylase inhibitors according to the invention are also described in Wang & Dymock (2009) Expert Opin. Ther. Pat. 19:1727-1757.
Preferably, the HDAC inhibitor according to the invention is selected from the group consisting of Trichostatin A (TSA), SAHA (Vorinostat), CAY10433, HDAC1 , CAY10591 , CAY10398, CAY10603, CBHA, HDAC8, M344, Salermide and Sodium Butyrate. More preferably, the HDAC inhibitor according to the invention is selected from the group consisting of Trichostatin A and SAHA (Vorinostat).
Methyltransferases catalyze methylation of DNA at position 5 on cytosine.
Aberrations in methylation play a causal role in a variety of diseases, including cancer. Methyltransferase inhibitors inhibit the action of methyltransferases. As an example of methyltransferase inhibitors according to the invention, one can cite 5- Aza-2'-deoxycytidine (5-Azadc), 5-azacytidine (Vidaza), N-acetyl-S-farnesyl-L- cysteine, S-Farnesyl Thioacetic Acid, N-acetyl-S-geranygeranyl-L-Cysteine, Phenylethanolamine N-methyltransferase, Catechol-O-methyl transferase, Histamine dihydrochloride, Tryptamine hydrochloride, Hydralazine hydrochloride, Procainamide hydrochloride, azacytidine, decitabine, entecapone, chaetocin, SKF 91488 dihydrochloride, S-Farnesylthiacetic acid, 5-AzadCyd, RG108, Zebularine, 2'-deoxycytidine, arabinosylcytosine (Ara-C), arabinosyl-5-azacytosine (fazarabine), dihydro-5-azacytidine (DHAC).
The terms "cancer" and "malignancy" refer to or describe the pathological condition in mammals that is typically characterized by unregulated cell growth. In particular, the cancer may be thyroid cancer, skin cancer like melanoma, breast cancer, bladder cancer, uterine/cervical cancer, oesophageal cancer, pancreatic cancer, colon carcinoma, colorectal cancer, kidney cancer, ovarian cancer, prostate cancer, head and neck cancer, lung cancer, non-small cell lung cancer and stomach cancer, B or T cell malignancy, leukaemia.. Preferably, the cancer according to the invention is selected from the group consisting of thyroid cancer, skin cancer, for example melanoma, or colon carcinoma. More preferably, the cancer according to the invention is thyroid cancer or colon carcinoma.
According to the invention, the term "patient" or "individual" to be treated is preferably intended for a human or non-human mammal (such as a rodent (mouse, rat), a feline, a canine, or a primate) affected or likely to be affected with cancer. Preferably, the patient is a human.
The term "treating" or "treatment" means reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition. In particular, the treatment of the disorder may consist in destroying or depleting cancer cells. Most preferably, such treatment leads to the complete depletion of cancer cells. Preferably, the depletion of the cancer cells is obtained by inducing an arrest of the cellular cycle of said cells.
As intended herein, the expression "cellular cycle" relates to the series of events that take place in a cell leading to its division and duplication (replication). As is well known, the cycle is divided in four successive phases G1 , S, G2 and M. The expression "an arrest of the cellular cycle" means that this cycle is totally stopped in either one of the phases. Such an arrest leads to cellular death.
The "cells from cancer", "cancer cells" or "cancerous cells" according to the invention can originate from a cancer cell line. For example, when the efficiency of a compound for the treatment of thyroid cancer is tested, the cancer test cells may be the papillary thyroid cell lines CAL62, 8505c, BcPAP and TCP1 , notably described by Schweppe et al. (2008) The Journal of clinical endocrinology and metabolism 93(1 1 ):4331 -41 . The methods and techniques for growing these cells depend on the types of cells which are being tested and are well known by the skilled person.
Alternatively, the "cells from cancer" "cancer cells" or "cancerous cells" according to the invention can be cells from a patient affected with cancer, preferably a cancer according to the invention. The patient can optionally be under anti-cancer treatment, preferably under treatment with the compound according to the invention. More preferably, the cancer cells according to the invention are cancer cells from a patient treated with the compound according to the invention.
As intended herein, the expression "cancer cells which have been in contact with said compound" preferably means that the cancer cells have been contacted with the compound which anti-cancer activity is to be determined under conditions and concentrations which allow the compound to have an effect on the cells. These conditions and concentrations can be easily be determined by the person skilled in the art.
The contact between the cancer cells and the compound which anti-cancer activity is to be determined can be an in vitro contact and/or an in vivo contact between the cancer cells and the compound.
An in vivo contact according to the invention notably occurs when the cancer cells are cells from a patient treated with said compound. In this case, the level of expression of a mi-RNA selected from miR-1 29-5p and/or miR-133b and/or precursors thereof can be either directly detected in the cells or be determined upon a further in vitro contact with compound.
By way of example of an in vitro contact, cells from a cancer can be contacted with trichostatin A (TSA) at a concentration of about 100ng/ml for about 10 hours.
The term "mi-RNA" or "microRNA" is used to qualify a class of RNAs generally 10 to 30, in particular 15 to 25 nucleotides long, and play a role in the post-transcriptional regulation of specific genes, by degrading or blocking the translation of the mRNAs resulting from the transcription of these genes. In particular, it is preferred that mi-RNAs according to the invention are mi-RNAs which expression is increased after administration of an anti-cancer medicament to a patient, which increase in expression can further be involved in the anticancer effect of this medicament. Preferably, the mi-RNAs according to the invention are miR-129-5p, in particular Homo sapiens (hsa) miR-129-5p, more particularly represented by SEQ ID NO: 1 , and miR-133b, in particular Homo sapiens (hsa) miR-133b, more particularly represented by SEQ ID NO: 2.
As intended herein, the terms "miR-129-5p" and "miR-133b" notably encompass all the various allelic or polymorphic variants of miR-129-5p, in particular of hsa miR-129-5p, and of miR-133b, in particular of hsa miR-133b, as well as the various orthologous sequences of miR-129-5p and miR-133b, i.e. the species-specific sequences.
miR-129-5p and miR-133b are notably described in Griffiths-Jones (2004)
Nucleic Acids Res 32:D109-D1 1 1 ; Griffiths-Jones et al. (2008) Nucleic Acids Res 36:D154-D158; and the miRBase database available at http://microrna.sanger.ac.uk. A "precursor" of a mi-RNA according to the invention refers to any intermediate in the mi-RNA maturation pathway starting from the transcript obtained from the transcription of a chromosome and ending with the obtaining of a mi-RNA. Thus, a precursor according to the invention can be a mi-RNA primary transcripts (pri-miRNA) or a pre-miRNA, as well as a long non-coding RNA, a snoRNA or a transposon which encodes a mi-RNA before processing via the enzyme Dicer. The most preferred precursor of miR-129-5p according to the invention is miR-129-2, in particular represented by SEQ ID NO: 3. The most preferred precursor of miR-133b according to the invention is miR-133b hairpin, in particular represented by SEQ ID NO: 4. miR-129-2 and miR-133b hairpin are notably described in Griffiths-Jones (2004) Nucleic Acids Res 32:D109-D1 1 1 ; Griffiths-Jones et al. (2008) Nucleic Acids Res 36:D154-D158; and the miRBase database available at http://microrna.sanger.ac.uk.
Particularly preferred variants of miR-129-5p and of miR-133b may lack one or two nucleotides on the 3' end with respect to their respective consensus sequences, in particular represented by SEQ ID NO: 1 and 2. Other particularly preferred variants of miR-129-5p and of miR-133b may comprise one or two additional nucleotides with respect to their respective consensus sequences, in particular represented by SEQ ID NO: 1 and 2, said additional one or two additional nucleotides being those immediately on the 3' end of the sequence of miR-129-5p and miR-133b within the sequence of their respective precursors, miR-129-2 and miR-133b hairpin, which are themselves notably represented by SEQ ID NO: 3 and 4 respectively.
The level of expression of the mi-RNAs in the cells can be measured by any techniques known by the person skilled in the art. Numerous methods are available which allow quantifying a target RNA, for example, methods based on PCR after reverse transcription (RT-PCR) using oligonucleotides which are specific of the target RNA sequences, or alternatively, methods allowing the hybridization of the target RNA, of duplicates or triplicates of the target RNA with probes under stringent conditions. The probes according to the invention are preferably laid down on microarrays. As intended herein, the expression "hybridization under stringent conditions" indicates that the target RNA or the duplicates thereof can specifically bind pairwise, essentially by forming Watson- Crick-type pairs {e.g. G-C pairs or U-A pairs), with probes having sequences complementary thereto. Adequate stringent conditions according to the invention can be easily determined by one of skill in the art. Preferred stringent conditions according to the invention comprise a hybridization step of 10 to 20 hours, preferably 16 hours, at about 40 to 55 °C, preferably 50 °C, under an ionic strength equivalent to that provided by 500 mM to 2 M NaCI, preferably 1 M NaCI. Additional compounds well known to one skilled in the art can also be added such as pH buffers (e.g. Tris or MES), EDTA, Tween, Bovine Serum Albumin, and herring sperm DNA.
The "reference value" according to the invention can be a unique value such as a given level of expression of a mi-RNA or of a precursor thereof, alternatively, it can an average between expression levels of a mi-RNA or of a precursor thereof.
For example, the reference value can be the level of expression of miR- 129-5p and/or miR-133b and/or precursors thereof in cells which are non-cancer cells, which are preferably of the same type as the cancer cells which have been contacted with the compound which anti-cancer activity is determined according to the invention. For example, if the cells from cancer originate from a papillary thyroid cancer, then the non-cancer cells will be non-cancer papillary thyroid cells.
Alternatively, the reference value according to the invention can be the level of expression of miR-129-5p and/or miR-133b and/or precursors thereof in cancer cells which have not been in contact with the compound which anti-cancer activity is determined according to the invention.
As intended herein, the expression "comparing the levels of expression" means that the value of the level of expression of miR-129-5p and/or mi-R133b and/or precursors thereof in the cancer cells after contact with the compound which anti-cancer activity is determined according to the invention is compared to the reference value for, respectively, miR-129-5p and/or mi-R133b and/or precursors thereof.
The expression "determining the anti-cancer activity" as used herein relates to the identification of compounds likely or unlikely to have an effect in the treatment of cancer. As intended herein, the expression "increase in the level of expression of mi-RNA selected from miR-129-5p and/or mi-R133b and/or precursors thereof" indicatess that the level of expression is higher in the cancer cells after treatment with the compound according to the invention, compared with the reference value.
Preferably, an increase of the level of expression indicates that the compound presents an anti-cancer activity. Preferably, as intended herein, the level of expression of miR-129-5p or mi-R133b or precursor thereof is increased in the cancer cells contacted with compound when the level of expression is at least 1 .2 fold, more preferably at least 1 .5 fold, and most preferably at least 2 fold over the reference value.
The method of the invention may further comprise monitoring the level of expression of a mi-RNA selected from miR-129-5p, miR-133b and precursors thereof over a period of time in cancer cells from a patient treated with the compound which anti-cancer activity is determined according to the invention. Preferably, the anti-cancer activity of a compound used for treating cancer in a patient treated with the compound is determined according to the invention at consecutive times during the course of the treatment in order to establish a therapeutic follow-up of the patient. The follow-up is typically performed by determining the level of expression of miR-129-5p, miR-133b and precursors thereof according to the invention, at various time intervals, for instance every 2 weeks, 1 month, 2 months, 3 months, 5 months, etc.
Thus, when a therapeutic follow-up is performed, the reference value according to the invention can also be, in addition to the previous definitions of the term, a previous level of expression of a mi-RNA selected from miR-129-5p and/or miR-133b and/or precursors thereof in cancer cells form the same patient.
Preferably, the therapeutic follow-up according to the invention is started at the same time or upon onset of the treatment of the patient with an anti-cancer compound according to the invention. Advantageously, a decrease in the level of expression of the mi-RNA selected from miR-129-5p and/or miR-133b and/or precursors thereof during the therapeutic follow-up is preferably indicative of a decrease of the anti-cancer activity of the compound thereby indicating a loss of sensitivity of cancer cells the patient to the compound. As intended herein the nucleic acid for use as a medicament, comprised in a pharmaceutical composition, or administered to a patient, can be of any type, it can notably be natural or synthetic, DNA or RNA, single or double stranded. In particular, where the nucleic acid is synthetic, it can comprise non-natural modifications of the bases or bonds, in particular for increasing the resistance to degradation. Where the nucleic acid is RNA, the modifications notably encompass capping its ends or modifying the 2' position of the ribose backbone so as to decrease the reactivity of the hydroxyl moiety, for instance by suppressing the hydroxyl moiety (to yield a 2'-deoxyribose or a 2'-deoxyribose-2'-fluororibose), or substituting the hydroxyl moiety with an alkyl group, such as a methyl group (to yield a 2'-0-methyl-ribose).
In the frame of the above-defined medicament, pharmaceutical composition and method of treatment, where the nucleic acid of the invention comprises or consists of SEQ ID NO: 1 or SEQ ID NO: 2, or of the sequences derived therefrom, the nucleic acid of the invention is intended to directly exert its effect on its cellular targets. In this case, the nucleic acid is preferably a RNA molecule. In contrast, where the nucleic acid of the invention encodes a nucleic acid comprising or consisting of SEQ ID NO: 1 or SEQ ID NO: 2, or of the sequences derived therefrom, the nucleic acid of the invention is intended to be expressed within cells where the nucleic acid it encodes, in particular a RNA molecule, will exert its effect on its cellular targets. In this case the nucleic acid of the invention is preferably a DNA molecule, more preferably a double stranded DNA molecule. Besides, as will be clear to one of skill in the art, the nucleic acid according to the invention preferably also comprises genetic elements ensuring expression of the encoded nucleic acid, in particular a promoter sequence of RNA polymerase II or III.
Methods for delivering nucleic acids into cells in vitro or in vivo are well known to one of skill in the art and are notably described in Nguyen et al. (2008) Curr Opin Mol Ther 10:158-67 and Dykxhoorn et al. (2006) Gene Therapy 13:541 - 552, which are incorporated herein by reference.
Preferably, in the frame of the above-defined medicament, pharmaceutical composition and method of treatment, where the nucleic acid of the invention comprises SEQ ID NO: 1 or SEQ ID NO: 2 or a sequence derived therefrom, it is less than 1000 nucleotides long, more preferably less than 100 nucleotides long, and most preferably less than 50 nucleotides long. Also preferably, the sequence derived from SEQ ID NO: 1 or SEQ ID NO: 2 by substitution deletion or insertion of at least one nucleotide presents at least 85%, more preferably at least 90%, and most preferably at least 95% identity with the sequence from which it is derived. As intended herein, the percentage of identity between two sequences is obtained by aligning the two sequences so as to maximize the number of positions of each sequence for which the nucleotides are identical and dividing the number of positions of each sequence for which the nucleotides are identical by the number of nucleotides of the longer of the two sequences.
More preferably, the medicament or the pharmaceutical composition according to the invention comprises, or the method of treatment of the invention comprises administering, a RNA molecule consisting of SEQ ID NO: 1 and/or a RNA molecule consisting of SEQ ID NO: 2.
Most preferably, the medicament or the pharmaceutical composition according to the invention comprises, or the method of treatment of the invention comprises administering, miR-129-5p or a precursor thereof, such as miR-129-2, as defined above, and/or miR-133b or a precursor thereof, such as miR-133b hairpin, as defined above.
Where a medicament, a pharmaceutical composition, or a method of treatment of the invention is contemplated, the nucleic acid can be associated to one or more pharmaceutically acceptable carriers. In particular, it is preferred that the pharmaceutically acceptable carrier be suitable for delivering nucleic acid into cells. Carriers suitable for delivering nucleic acid into cells are well known to one of skill in the art and notably comprise cationic lipids or peptides, nanoparticles and liposomes, optionally linked to moieties, such as antibodies or antibody fragments, having a specificity towards a specific receptor of the target cells, notably cancer cells.
Either local or systemic routes can be used for administering the nucleic acid of the invention to a patient. Examples of administration procedures for nucleic acids are notably described in Nguyen et al. {op. cit.) and Dykxhoorn et al. {op. cit.)
Preferably, a RNA molecule consisting of SEQ ID NO: 1 and/or a RNA molecule consisting of SEQ ID NO: 2 according to the invention are administered to a patient in need thereof in combination or are present together in a same medicament or pharmaceutical composition.
Preferably also, the at least one nucleic acid according to the invention is combined in a medicament or a pharmaceutical composition, or administered in combination, with at least one other anti-cancer compound. In particular, the other anti-cancer compound can be a medicament useful to treat thyroid cancer or skin cancer like melanoma cancer. Examples of such medicaments include cytotoxic agent such as gemcitabine, paclitaxel, cisplatin, etoposide and doxorubicin and mechanism-based agents such as HSP90 inhibitor17-AAG and the proteasome inhibitor bortezomib.
EXAMPLE
1. Materials and Methods 1.1. Reagents and HDACi
Etoposide, Staurosporine and the most widely used HDAC inhibitor Trichostatin A (TSA) were provided by Sigma-Aldrich (Paris, France) and the clinically-relevant HDACi Vorinostat (SAHA) by Cayman Chemical (Ann Arbor, Ml). HAMLET was produced from native purified human milk alpha-lactalbumin on an oleic acid- conditioned ion exchange matrix.
miRNAs (Pre-miR™ miRNA Precursor Molecules, Ambion®) and antagomiRs (Anti-miR™ miRNA Inhibitors, Ambion®) were purchased from Applied Biosystem (Courtaboeuf, France). 1.2. Cell lines and treatment
Thyroid cancer cell lines BcPAP [papillary, BRAF-V600E], TPC1 [papillary, RET- PTC1 ], 8505c [anaplasic, BRAF-V600E] and CAL62 [anaplasic, KRAS-G12R] have been previously characterized (Ito et al. (1993) Cancer research 53:2940-3; Fabien et al. (1994) Cancer 73:2206-12; and Gioanni et al. (1991 ) Bulletin du cancer 78:1053-62).
Cells were grown in Dulbecco's modified Eagle's medium (DMEM) supplemented with glutamax, penicillin (100 units/ml)-streptomycin (100 μ9/ιηΙ), sodium pyruvate (1 mM) (Invitrogen, Cergy Pontoise, France), 10% fetal calf serum. The absence of cross-contaminations between the cell-lines was checked by amplification and sequencing of RET/PTC 1 , BRAF-V600E, and KRAS-G12R after each defreezing (every 2 months) (Schweppe ei al. (2008) Journal of clinical endocrinology and metabolism 93:4331 -41 ).
To test the toxicity sensitivity of HDACi treatments on healthy normal thyroid cells, primary cultures of thyrocytes were derived from adenoma or hyperplasia thyroid tissue immediately after thyroidectomy at the University of Nice hospital. Patient consent was received and the institutional review board approved the project. Cell dissociation procedures, were similar to those used previously. Briefly, thyroid tissue was cut into small pieces (less than 1 mm diameter), washed in PBS and digested for 3x20 min with a solution consisting of 10 U/ml collagenase, 1 mg/ml dispase (Invitrogen) in PBS. After filtration on 300 μιη gaze, cells were seeded at a density of 105 per 25cm2 Flask. Cells were fed with F12/Coon's medium supplemented with 5%FCS twice a week.
For all experiments, cells were grown to 70% confluence, and TSA (330nM) or Vorinostat (2.5 μΜ) were added for 16h in fresh complete medium. As controls, cells were left untreated. The effects of HDACi were assessed on cell viability (cell cycle, MTT and morphology) by biochemical assays as well as miRNA and mRNA profiling by microarray analyses.
1.3. miRNA Transfection
Cells were plated at 100 000 cells/well in a 6 wells plate, after 24 h, cells were washed and incubated in 1 ml antibiotic free medium (7 vol: 3 vol DMEM/Optimem) containing 2.5 μΙ of Lipofectamin RNAIMAX, 5 pmol/L of the specific miRNA, or 20 pmol/L of the specific antagomiR. Serum was added after 6 h. When indicated, the following day, HDACi were added in fresh complete medium for 24h. 48h after transfection, cells were lysed for protein or RNA analysis as described in the following parts. Rates of transfection were checked by RT-PCR of the specific transfected miRNA on 7500 thermal cycler (Applied Biosystem).
1.4. Cell cycle analysis
Cell cycle analysis was used in order to analyze both growth arrest (with S phase decrease), and cell death (by increase in subG1 cells) as two endpoints for HDACi activity (Brest et al. (2007) Cancer research 67:1 1327-34). Cells were harvested and fixed in 75% ethanol for 2h. After washes, fixed cells were stained with propidium iodide (2.5 μ9/ιτιΙ, Sigma-Aldrich) and RNase A (250 μ9/ιτιΙ) at 4°C overnight prior to measurement. Cellular fluorescence was measured using the Facscalibur flow cytometry (Becton Dickinson, San Jose, CA). The single-cell populations were determined on the basis of their fluorescence-intensity values FL2-A and FL2-W. S-Phase percent was calculated as a ratio between (S-phase cells)/(G1 +S+G2-phase cells). Cell death quantification was calculated as a ratio between (SubG1 -phase cells)/(subG1 +G1 +S+G2-phase cells). 1.5. Western blot
Cells were lysed in Laemmli buffer (12.5 mM Na2HP04, 15% glycerol, 3% SDS). 50 μg of protein extracts were separated by SDS-PAGE, electrotransferred onto a PVDF membrane, and incubated with anti-acetyl histone H4 (1 /3000, Millipore, Molsheim, France), anti-p21 WAF1 (1 /1000, Santa Cruz Biotechnologies, Heidelberg, Germany), anti-phospho-ser139-H2AX (clone JBW, 1 /3000, Millipore), or anti-PARP (1 /1000, Millipore), in TN-Milk buffer (10 mM Tris HCI, pH 7.4, 0.15 M NaCI, 1 mM EDTA, 0.1 % Tween-20, 5% non fat milk) overnight at 4°C. Appropriate HRP-conjugated secondary antibodies were then applied for 1 h at room temperature. Immunoreactive bands were revealed by enhanced chemiluminescence (ECL, PerkinElmer, Courtaboeuf, France). 1.6. MTT Colorimetric Viability Assay
Cell survival was examined using the MTT colorimetric assay as described (Xiao et al. (2009) Molecular cancer therapeutics 8:350-6). After miRNA transfection and/or HDACi treatment, cells were detached, plated in 96-well plates at 2 104 cells/well in 4-6 replicates, and incubated with 150 μΙ_ MTT (0.5 mg/ml) for 4 h at 37 °C. The accumulated formazan in viable cells was dissolved in 100 μΙ_ DMSO, and then quantified by optical density at 570 nm (Dynex, France). Cell viability was estimated as a percentage of the value of untreated controls (100%). All experiments were repeated at least three times, and each experimental condition was repeated at least in quadruplicate wells in each experiment. Data reported are average value ± SD of representative experiments.
1.7. Total RNA Extraction
After incubations, cells were lysed in Trizol (Invitrogen). After chloroform, water fraction containing total RNA was collected. After addition of absolute ethanol (1 .5vol/vol), total RNA was purified with the RNAeasy extraction kit (Qiagen, Courtaboeuf, France), according to the manufacturer's instructions. The quality was assessed by measuring both the optical density (Nanodrop, Wilmington, DE), and the electrophoretic mobility using a Bioanalyzer (Agilent Technologies, Santa Clara, CA).
1.8. Global miRNA Expression profiling induced by HDACi
The transcriptional profile induced by HDACi was characterized by oligonucleotide microarray analysis of cells treated with HDACi (1 6 h) vs. control cells, by using a microarray containing 2054 mature miRNAs (409 homo sapiens) found in the miRNA registry, as described by Potier et al. (2009) PloS One 4:e671 8. Fragments of tRNAs, snoRNAs, 5S and 5.8S RNA were also printed on the microchip, providing internal positive controls for specific hybridization. The oligonucleotide sequences are available on http://www.microarray.fr/microRNA/. Each oligonucleotide was spotted four times on each slide (2 distinct pairs of spots), in order to reduce positional bias of the fluorescence readout. This miRNA platform has been registered on the Gene Expression Omnibus public data repository (GEO) under the reference GPL471 5.
Total RNA were labeled with Cy3 or Cy5 fluorescent dye using the Ulysis Alexa fluor nucleic acid labeling kit (Amersham Bioscience, Pittsburgh, PA), then miRNA were isolated using the mirVana miRNA isolation Kit (Applied Biosystem). Three independent experiments were made to identify miRNA up regulated or down regulated in cells. For each oligonucleotide microarray, TI F images containing data from each fluorescence channel were quantified with GenePix Pro 6.1 program (Axon Instruments). Normalization was obtained with the software limma from Bioconductor according to the vsn approach described by Huber et al. (2002) Bioinformatics 18 Suppl 1 :S96-1 04.
1.9. Microarray Analysis of Gene Expression profile induced by miRNA transfection
The transcriptional profile induced by miRNA transfection was characterized by oligonucleotide microarray analysis of cells treated with miR-1 33b or miR-1 29-5p vs. control miRNA transfected cells, by using oligonucleotide microarrays containing 25.484 distinct oligonucleotide probes covering most of the known human transcripts (Le Brigand et al. (2006) Nucleic acids research 34:e87). The list of the probes (length 51 bp) is available online htjp:/ www.irilcroarray.jr. Microarrays were printed with a ChipWriter Pro (Bio-Rad) on commercial hydrogel slides (Schott) and processed according to the manufacturer's instructions.
Total RNA (2 μg) were amplified with the Amino Allyl MessageAmp aRNA kit (Ambion) according to the manufacturer's instructions. Cy3- and Cy5-labeled aRNA were hybridized on array for 17 h at 62 °C (a dye-swap method has been used). Arrays were then washed with expression wash buffer kit (Agilent) and were scanned with a genePix4000B microarray scanner (Axon instrument).
16-bit TIF images were quantified with the corresponding software (GenePix Pro 6.1 program (Axon Instruments) for the GenePix and Quantarray for the ScanArray. Intra and inter slide normalization of three independent experiments were performed using Global Loess and the quantile methods respectively. Means of ratios from all comparisons were calculated and B test analysis was performed using the Limma package available from Bioconductor (Gentleman et al. (2004) Genome Biol 5:R80). Differentially expressed genes were selected using a Benjamini-Hochberg correction of the p-value for multiple tests, with a positive B value. Before analysis, the sylamer algorithm was used in order to characterize the specificity of miR-133b and miR-129-5p-transfection on global mRNA gene expression. As expected, mRNA harboring the seed sequences of miR-133b and miR-129-5p were specifically downregulated. The human targets of the differentially expressed miRNAs were predicted using public Web-based prediction tools, such as PicTar (http/./pictar.bio.nyu.edu) (Krek et al. (2005) Nature genetics 37:495-500), TargetScan (http://cjenes.rnit.edu/tarqetscan/index.htmj) (Lewis et al. (2005) Cell 120:15-20), and miRBase Targets
(ht;p: microrna.sarKjer ac. jk-';arqet$/v3/) (Griffiths-Jones et al. (2006) Nucleic acids research 34(Database issue):D140-4).
1.10. Validation of miRNA and pangenomic arrays
For validation of mRNA putative target identified by microarray, mRNA from HDACi treated- or miR-133b and miR-129-5p transfected- cells were subjected to reverse transcription-polymerase chain reaction followed by specific Taqman assays (Applied Biosystems). Relative fold changes of expression in induced samples against control were calculated using the comparative Ct (2-ΔΔΟί) method. Real-time quantization was carried out on the 7500 Fast (Applied Biosystem) thermal cycler, and reagents provided by Applied Biosystem under conditions suggested by the manufacturer.
1.11. Statistical analysis
For statistical analysis, GraphPad Instat3 program was used. The results were evaluated for statistical significance by student t-test or ANOVA test. Error bars were marked as the standard deviation (S.D.) of the mean, p values less than 0.05 were regarded as significant. 1.12. Viability assay
Cell survival was examined using the Cell Proliferation Kit II (XTT) from Roche and quantification of DNA fragmented-subG1 population by cell cycle analysis as previously described (19). All experiments were repeated at least three times, and each XTT experimental sample (5000 cells/well) was repeated at least in quadruplicate wells for each experiment. The data are average values ± SD of representative experiments.
2. Results 2.1. HDACi induce cell cycle arrest and cell death
HDACi increase histone acetylation by preventing HDAC activity (Gorisch et al. (2005) J Cell Sci 118:5825-34). The activity of HDACi on a panel of thyroid cancer cell lines carrying different oncogenes RET/PTC 1 [TPC1 ], BRAF V600E [BcPAP, 8505c] and KRAS G12R [CAL62], was therefore analyzed by western blotting using specific antibodies for acetylated histone H4. The inventors showed that all thyroid cell-lines used were sensitive to TSA (330 nmol/L), as evidenced by the increased histone H4 acetylation. Moreover, P21 WAF1 (CDKN1 A), a gene down-regulated in cancers (Kastan & Bartek (2004) Nature 432:316-23; Massague (2004) Nature 432:298-306), and previously shown to be restored by HDACi treatment (Brest et al. (2007) Cancer research 67:1 1327-34; Blagosklonny et al. (2002) Molecular cancer therapeutics 1 :937-41 ) was upregulated in HDACi-treated thyroid cells, in agreement with previous studies (Mitsiades et al. (2005) Clin Cancer Res 11 :3958- 65; Luong et al. (2006) Clin Cancer Res 12:5570-7). P21 WAF1 acts as a potent cyclin-dependent kinase inhibitor at the G1 checkpoint causing a rapid G1 -S arrest. Consistently, the inventors found a decrease in S phase in cycling cells (5.8% in TSA-treated cells vs 12.8% unteated cells, p<0.05) and an increase of apoptotic cells with fragmented DNA (66% in TSA treated cells vs 2% in control, p<0.001 ). Furthermore, DNA damage and apoptotic cell death were confirmed by phosphorylation of histone H2AX and by cleavage of PARP with a higher sensitivity of papillary thyroid cells. Altogether these findings indicate that TSA was able to induce cell cycle arrest and apoptotic cell death in thyroid cancer cells, regardless of the oncogenic status.
2.2. HDAC inhibitors induce miR-133b and miR-129-5p overexpression
The molecular mechanism responsible for HDACi antitumoral activity remains elusive. miRNAs are candidates that can be considered as putative tumor suppressor. So far, the relationship between miRNA induction and HDACi sensibility of thyroid tumor cells is unknown. To gain insight into their possible involvement, miRNA profile was assessed by microarray analyzes after HDACi treatment. BCPAP and 8505c cell lines were treated with TSA overnight (330 nmol/L), miRNA were isolated and their expression level was analyzed on microarray (Table 1 ). Among the HDACi-induced miRNAs, the inventors chose to focus their attention on the specific increase in miR-133b and miR-129-5p since their increase was confirmed by RT-PCR after TSA treatment in all the thyroid cell lines used. TSA treatment induced an increase in miR-133b expression ranging from 3-fold (8505c, CAL62) to 5.6-fold (TCP1 ) (Figure 1 ) and an increase in miR- 129-5p expression ranging from 7-fold (CAL62) to 18-fold (TCP1 ) (Figure 2) in comparison with untreated cells used as reference. These results were confirmed by using SAHA, another HDACi, on TCP1 cells that showed similar fold of increase as TSA (Figure 3). Interestingly, the highest induction was found in papillary cell lines and correlated to the higher sensitivity in cell death. At that stage it was of interest to ascertain the specificity of HDACi-induced responses (viability, miRNA expression) on normal healthy thyrocytes. As shown in Figure 4, the increase of miR-133b and miR-129-5p under HDACi treatment was restricted to cancer cells since no significant modification of the level of expression was observed in hyperplasia or adenoma primary thyroid culture treated with TSA. Taken together, these findings clearly indicate that HDACi anti-tumoral activity was closely correlated with their ability to upregulate the expression of miRNA miR- 133b and miR-129-5p: both miRNAs were dramatically induced in the HDACi- hypersensitive TCP1 cells while unchanged in HDACi-resistant healthy cells.
2.3. miR-133b and miR-129-5p overexpressions are sufficient for cell growth arrest
To gain insight into the possible involvement of the miR-133b and miR-129-5p in the mediation of the HDACi-antitumoral effect, these miRNAs were transfected into TCP1 cells. After 48 h, transfected cells were analyzed for cell cycle distribution. Introduction of miR-133b and/or miR-129-5p caused a significant reductions of population of cells entering in S-Phase (5.1 ±0.5% for miR-133b, 3.3±0.4% for miR-129-5p, p<0.05), similarly to HDACi treatment (TSA, 330nM, 24h). Cells transfected with negative control miR-CON1 showed the same number of cells in S-Phase (9.7±0.8%) than non-transfected cells. These data underscore that expression of miR-133b and miR-129-5p are sufficient alone for cell growth arrest in an HDACi-treatment similar manner.
2.4. miR-133b and miR-129-5p overexpressions induce decrease expression of cyclin B1 and cyclin F
This conclusion was strengthened by the fact that genes involved in cell cycle were downregulated by overexpression of these miRNAs. To delineate the putative targets of these miRNA, the mRNA profile of miRNA-transfected TPC1 cells was analyzed by a pan-genomic microarray. The inventors focused on genes containing a seed match for miR-133b and miR-129-5p that were downregulated with a logarithm fold change higher than 0.8. As defined by the Gene Ontology Consortium (www.geneontology.org) (Ashburner et al. (2000) Nature genetics 25:25-9), the miRNA transfection associated-meta-signature mostly shed light on genes involved in the cell division, mitosis, and cell cycle (Tables 2, 3, 4) underlying the involvement of these miRNAs in cell cycle arrest.
In order to confirm putative downregulation of cyclin targeted by miR-133b and miR-129-5p, the expression of cyclin B1 (CCNB1 ) and cyclin F (CCNF) was analyzed by qPCR. Consistently, transfection of either miR-133b or miR-129-5p induced a decrease in cyclin B1 and in cyclin F expression (5.6-fold and 7-fold respectively) similarly than TSA-treated cells. In conclusion, the inventors found that miR-129-5p, miR-133b and HDACi shared similar targets like cyclins B1 and F to induce cell cycle arrest.
2.5. miR-133b and miR-129-5p are not necessary for cell growth arrest
Moreover, the role of miR-133b and 129-5p in HDACi-induced cell cycle arrest was then evaluated by transfection of miRNAs or antagomiRs. Interestingly, an additional effect on S-phase decrease was observed when HDACi and miR-133b or miR-129-5p were combined, showing that both miRNA improved HDACi treatment. Unexpectedly, knocking down miR-133b and/or miR-129-5p expression with antagomiRs did not affect HDACi-induced cell growth arrest, showing a potential compensatory regulation of miR-129-5p and miR-133b knockdown.
In conclusion, HDACi-induced overexpression of miR-129-5p and miR-133b participates to cell cycle arrest via a specific decrease in cyclin B1 and F expression in thyroid cancer cells.
2.6. miR-129-5p is sufficient for HDACi induced cell death
HDACi has been shown to induce cell cycle arrest followed by cell death in thyroid cancer cells. The role of both miR-133b and miR-129-5p was investigated in this context.
Interestingly, miR-129-5p-transfected TCP1 cells showed an increased cell death, suggesting that this miRNA is cytotoxic for cancer cells. The inventors then analyzed pan-genomic microarray of miR-129-5p-transfected TPC1 cells on cell death-involved genes expression. Data collected from three independent biological experiments, revealed that a total of 224 transcripts were significantly modulated (102 up and 122 down-regulated, p<0.01 ) following pre-miR-129-5p transfection when compared to the control condition. Analysis of this signature with Ingenuity Pathway™ software indicated a significant enrichment for "Molecular functions" with terms such as "Cellular Compromise", "Cellular Growth and Proliferation", "Cell Cycle" or "Cell Death". Using the bioinformatic tool "MicroToptable", the inventors then looked for potential over representation of miR-129-5p-predicted direct targets among the downregulated transcripts (cut-offs equal to 8.0 for the log2 (signal), to -1 for the log2 (ratio), and to 0.01 for the adjusted p-value) and isolated 42 transcripts corresponding to putative direct targets (Table 6).
Interestingly, the inventors found a decreased expression of GALNT1 (2"2 0~-4 fold), a gene previously described to be regulated by miR-129-5p, and BCL6 (B- cell CLL/lymphoma 6, 2"2 4~-5.3 fold), a repressor of the expression of the pro- apoptotic protein PDCD2 (programmed cell death 2) which was increased in the inventor's model (21 7~3.2 fold) (Table 5). Of particular interest, BCL6 was previously described to be repressed by HDACi both transcriptionally and post- translationally (Saito et al. (2006) Cancer cell 9:435-43; Bereshchenko et al. (2002) Nature genetics 32:606-13).
These results showed that the BCL6/PDCD2 couple is regulated by miR-129-5p which may be involved in HDACi induced cell death.
2.7. miR-129-5p is necessary for HDACi induced cell death
To examine if miR-129-5p is mandatory for HDACi-induced cell death, TCP1 cells were transfected 24h either with antagomiR-CON1 (control) or antagomiR-129-5p and then incubated with TSA (330 nmol/L, 24h). Moreover, antagomiR-133b alone or combined with antagomiR-129-5p were also used to block TSA-induced cell death.
As shown by microscopy and MTT viability (Figure 5), knockdown of miR-129-5p and/or miR-133b expression blocked HDACi-induced cell death in comparison with antagomiR-CON1 transfected cells treated by HDACi. These results were confirmed by the absence of subG1 population in comparison with TSA-treated cells (Figure 6).
Furthermore, HDACi-dependent repression of GALNT1 expression was reverted by antagomiR-129-5p (Figure 7). Similar results were found in BcPAP, Cal62 and 8505c cells transfected with antagomiR-129-5p and treated with TSA.
In conclusion, by blocking expression of miR-129-5p or miR-133b in cells, the inventors showed a prevention of HDACi-induced cell death, thereby showing that miR-129-5p and miR-133b are mandatory for HDACi killing effect on tumor cells. 2.8. miR-129-5p promotes drug-induced cell death
HDACi have previously been shown to induce cell death in response to other cancer drugs like etoposide, cisplatin or HAMLET. To examine if miR-129-5p is a shared target, we transfected TPC1 cells with either miR-129-5p or miR-CTL for 24h before the addition of increasing concentrations of etoposide or HAMLET. Combination with miR-129-5p increased the sensitivity to HAMLET, staurosporine or etoposide (Figure 8). The loss of viability in response to HAMLET (0.3 mg/ml, 24h) was 40% in miR-CTL transfected control cells, but increased to 70% in tumor cells transfected with miR-129-5p. A similar increase in cell death was obtained in cells treated with staurosporine or etoposide. Thus, while miR-129-5p affects tumor cell viability per se, it also improves the sensitivity to different cancer drugs through effects on HDAC function.
Table 1 : Profile of altered miRNA in TSA-treated BCPAP and 8505C cell lines (Fold change > ±0.75, Adjusted p Value <5 10"3)
Figure imgf000027_0002
Table 2: Putative targeted genes by miR-129-5p found downregulated pangenomic microarray: GO Pathway, gene list
Figure imgf000027_0003
Figure imgf000027_0001
Table 3: Putative targeted genes by miR-133b found downregulated in pangenomic microarray: GO Pathway, gene list
Figure imgf000028_0001
Name ID AveExpr t Fold
BRCA1 672 10.1 -1.0
CCNB1 891 12.9 -1.2
CCNF 899 9.9 -0.9
CD2AP 23607 9.3 -1.6
CDCA8 55143 9.8 -1.1
CENPE 1062 10.3 -1.0
CENPF 1063 11.6 -0.8
CEP55 55165 10.7 -0.9
CETN3 1070 10.0 -1.4
CIT 11113 11.1 -0.9
DSN1 79980 10.2 -1.2
FBX05 26271 9.7 -0.9
HELLS 3070 9.6 -0.9
MAEA 10296 11.9 -1.3
MCM3 4172 10.1 -0.8
MDC1 9656 9.1 -1.1
MKI67 4288 10.1 -1.0
NCAPG 64151 11.3 -1.3
NEK2 4751 9.9 -1.0
NUSAP1 51203 10.8 -1.2
RFC1 5981 10.3 -0.8
RFC5 5985 10.0 -0.9
SMC1A 8243 10.1 -1.1
TOP2A 7153 11.5 -1.4
TPD52L1 7164 12.0 -2.1
TPX2 22974 12.3 -0.9
WTAP 9589 10.7 -0.9 Table 4: Alteration of cell cycle related protein expression by transfection of miR-
129-5 or miR-133b
Figure imgf000029_0001
Table 5: Apoptotic related genes regulated by miR-129-5p
Log Entrez Gene RT-PCR
Symbol RefSeq p-value
Ratio ID Log
BCL6 NM 001706 -2.4 1.03E-02 604 -2.2
TNFSF10 NM 003810 -2.0 1.05E-02 8743
GALNT1 NM 020474 -1.7 8.40E-03 2589 -2.0
RUNX1 NM 001754 -1.6 2.63E-02 861
CTSB NM 147780 -1.6 1.05E-02 1508
NFKBIA NM 020529 -1.6 1.97E-02 4792
NUPR1 NM 012385 -1.5 1.63E-02 26471
FOX01 NM 002015 -1.4 2.30E-02 2308
BMF NM 001003942 -1.4 2.08E-02 90427
EGR1 NM 001964 -1.1 1.92E-02 1958
BAD NM 004322 -1.1 1.60E-02 572
TNFRSF25 NM 003790 -1.1 2.08E-02 8718
TNFRSF10B NM 147187 0.9 2.52E-02 8795
HDAC1 NM 004964 0.9 2.44E-02 3065
TBPL1 NM 004865 1.0 2.53E-02 9519
GADD45A NM 001924 1.0 2.40E-02 1647
BECN1 NM 003766 1.1 1.64E-02 8678
DAPK1 NM 004938 1.2 2.70E-02 1612
PPARG NM 138712 1.3 1.29E-02 5468
EAF2 NM 018456 1.3 2.97E-02 55840
HOXA3 NM 153632 1.5 2.14E-02 3200
ABCB1 NM 000927 1.5 1.05E-02 5243
PDCD2 NM 144781 1.7 2.79E-02 5134 +1.8
CYCS NM 018947 1.7 2.79E-02 54205
CYR61 NM 001554 2.6 2.08E-02 3491 Table 6: List of the mir-129-5p-predicted direct targets down-regulated following miR-129-5p overexpression in TCP1 cells
Symbol Genbank ID Av. Expr. LogFC
CDKN1C 1028 8.8 -1.3
ACTN1 87 12.9 -1.2
GALNT1 2589 9.7 -1.6
TRIP13 9319 9.9 -1.7
DNAJA3 9093 9.6 -1.3
RUNX1 861 10.1 -1.5
ARPC5 10092 12.8 -1.8
ZNF706 51123 11.8 -1.4
CKAP4 10970 13.1 -1.4
DPY19L1 23333 10.4 -1.8
PTMA 5757 11.6 -2.5
AZGP1 563 9.7 -2.2
ITGAE 3682 11.3 -1.1
C11orf54 28970 9.8 -1.3
NPEPPS 9520 11.8 -2.6
CA12 771 10.8 -2.2
VPS26A 9559 12 -2.3
AK2 204 10.4 -1.5
ZMAT2 153527 11.2 -2.1
CIT 11113 10.8 -1.3
PDXDC1 23042 10.8 -1.2
CCNF 899 9.7 -1.1
DNAH11 8701 10.7 -1.2
CDK6 1021 9.8 -2.2
CACYBP 27101 10.4 -1.7
MATR3 9782 12.6 -1.3
RASSF5 83593 9.5 -1.3
LYRM1 57149 10.4 -1.5
VAMP2 6844 10.3 -1.2
C20orf108 116151 8.4 -1.3
ALDH3A2 224 9.6 -1.2
F AM 129 A 116496 13.2 -2
BPTF 2186 10.3 -1.1
THUMPD2 80745 9.9 -1.1
TNFSF10 8743 9 -1.5
EMP1 2012 12.2 -3.8
LBR 3930 11.9 -1.8
KLHL24 54800 9.3 -1.7
CTBP2 1488 9.6 -1.9
GINS3 64785 10.1 -1.3
DTL 51514 10.2 -1.7
SH3KBP1 30011 11.1 -1

Claims

1. An in vitro method for determining the anti-cancer activity of a compound able to alter chromatin structure comprising:
i) measuring the level of expression of a mi-RNA selected from miR-129-5p and/or miR-133b and/or precursors thereof in cancer cells which have been in contact with said compound able to alter chromatin structure;
ii) comparing the level of expression with a reference value;
iii) determining therefrom the anti-cancer activity of the compound able to alter chromatin structure.
2. The method according to claim 1 , wherein the cancer cells are from a patient treated with the compound able to alter chromatin structure.
3. The method according to claim 1 or 2, wherein the cancer cells are contacted with the compound able to alter chromatin structure in vitro.
4. The method according to any of claims 1 to 3, wherein an increase in the level of expression of a mi-RNA selected from miR-129-5p and/or miR-133b and/or precursors thereof compared to the reference value is indicative of the efficiency of the compound able to alter chromatin structure on said cancer.
5. The method according to any of claims 1 to 4, wherein the reference value is the level of expression of miR-129-5p and/or miR-133b and/or precursors thereof in cancer cells which have not been in contact with the compound able to alter chromatin structure.
6. The method according to any of claims 1 to 4, wherein the reference value is the level of expression of miR-129-5p and/or miR-133b and/or precursors thereof in a non-cancer cell.
7. The method according to any of claims 1 to 6, wherein the cancer cells are from a patient treated with the compound able to alter chromatin structure and wherein the anti-cancer activity of the compound able to alter chromatin structure is determined at consecutive times during the course of the treatment in order to establish a therapeutic follow-up of the patient.
8. The method according to claim 7, wherein a decrease of the anti-cancer activity of the compound able to alter chromatin structure during the follow-up is indicative of lost of the sensitivity of the patient cancer cells to the compound able to alter chromatin structure.
9. The method according to claim 9, wherein the compound able to alter chromatin structure is selected from the group consisting of a histone deacetylase inhibitor and a DNA methyltransferase inhibitor.
10. The method according to any of claims 1 to 9, wherein the cancer is selected from the group consisting of the thyroid cancer, skin cancer and colon carcinoma.
11. At least one nucleic acid
(i) comprising or consisting of, or
(ii) encoding a nucleic acid comprising or consisting of,
a sequence selected from the group consisting of:
1 ) SEQ ID NO: 1 and SEQ ID NO: 2; and
2) a sequence derived from SEQ ID NO: 1 and SEQ ID NO: 2 by substitution, deletion or insertion of at least one nucleotide, provided that a nucleic acid consisting of the sequence derived from SEQ ID NO: 1 and SEQ ID NO: 2 is liable to induce cell cycle arrest in cells from cancer,
for use as a medicament.
12. The at least one nucleic acid according to claim 12, wherein the medicament comprises a RNA molecule consisting of SEQ ID NO: 1 and/or a RNA molecule consisting of SEQ ID NO: 2.
13. The at least one nucleic acid according to any one of claims 12 to 13, wherein the medicament is used for treating a cancer.
14. The at least one nucleic acid according to claim 14, wherein the cancer is selected in the group consisting of the thyroid or cancer, skin cancer and colon carcinoma.
PCT/EP2010/068759 2009-12-04 2010-12-02 Method for determining the efficiency of a cancer treatment WO2011067346A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP09306181.0 2009-12-04
EP09306181 2009-12-04

Publications (1)

Publication Number Publication Date
WO2011067346A1 true WO2011067346A1 (en) 2011-06-09

Family

ID=41647101

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2010/068759 WO2011067346A1 (en) 2009-12-04 2010-12-02 Method for determining the efficiency of a cancer treatment

Country Status (1)

Country Link
WO (1) WO2011067346A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060105360A1 (en) * 2004-02-09 2006-05-18 Croce Carlo M Diagnosis and treatment of cancers with microRNA located in or near cancer associated chromosomal features
WO2007081680A2 (en) * 2006-01-05 2007-07-19 The Ohio State University Research Foundation Microrna expression abnormalities in pancreatic endocrine and acinar tumors
WO2009080437A1 (en) * 2007-12-21 2009-07-02 Exiqon A/S Micro-rna based drug resistance analysis method
EP2088208A1 (en) * 2008-01-23 2009-08-12 Fujifilm Corporation Method for detecting carcinoma and agent for suppressing carcinoma
WO2009147525A1 (en) * 2008-06-04 2009-12-10 Karolinska Institutet Innovations Ab Skin cancer associated micrornas

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060105360A1 (en) * 2004-02-09 2006-05-18 Croce Carlo M Diagnosis and treatment of cancers with microRNA located in or near cancer associated chromosomal features
WO2007081680A2 (en) * 2006-01-05 2007-07-19 The Ohio State University Research Foundation Microrna expression abnormalities in pancreatic endocrine and acinar tumors
WO2009080437A1 (en) * 2007-12-21 2009-07-02 Exiqon A/S Micro-rna based drug resistance analysis method
EP2088208A1 (en) * 2008-01-23 2009-08-12 Fujifilm Corporation Method for detecting carcinoma and agent for suppressing carcinoma
WO2009147525A1 (en) * 2008-06-04 2009-12-10 Karolinska Institutet Innovations Ab Skin cancer associated micrornas

Non-Patent Citations (37)

* Cited by examiner, † Cited by third party
Title
ASHBURNER ET AL., NATURE GENETICS, vol. 25, 2000, pages 25 - 9
BERESHCHENKO ET AL., NATURE GENETICS, vol. 32, 2002, pages 606 - 13
BLAGOSKLONNY ET AL., MOLECULAR CANCER THERAPEUTICS, vol. 1, 2002, pages 937 - 41
BOLDEN ET AL., NAT REV DRUG DISCOV, vol. 5, no. 9, 2006, pages 769 - 84
BREST ET AL., CANCER RESEARCH, vol. 67, 2007, pages 11327 - 34
CALIN ET AL., NATURE REVIEWS, vol. 6, no. 11, 2006, pages 857 - 66
CRAWFORD MELISSA ET AL: "MicroRNA 133B targets pro-survival molecules MCL-1 and BCL2L2 in lung cancer.", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 23 OCT 2009, vol. 388, no. 3, 23 October 2009 (2009-10-23), pages 483 - 489, XP002568539, ISSN: 1090-2104 *
DYKXHOORN ET AL., GENE THERAPY, vol. 13, 2006, pages 541 - 552
DYRSKJOT LARS ET AL: "Genomic profiling of microRNAs in bladder cancer: miR-129 is associated with poor outcome and promotes cell death in vitro.", CANCER RESEARCH 1 JUN 2009, vol. 69, no. 11, 1 June 2009 (2009-06-01), pages 4851 - 4860, XP002568538, ISSN: 1538-7445 *
FABIEN ET AL., CANCER, vol. 73, 1994, pages 2206 - 12
GENTLEMAN ET AL., GENOME BIOL, vol. 5, 2004, pages 80
GIOANNI ET AL., BULLETIN DU CANCER, vol. 78, 1991, pages 1053 - 62
GORISCH ET AL., J CELL SCI, vol. 118, 2005, pages 5825 - 34
GRIFFITHS-JONES ET AL., NUCLEIC ACIDS RES, vol. 36, 2008, pages D154 - D158
GRIFFITHS-JONES ET AL., NUCLEIC ACIDS RESEARCH, vol. 34, 2006, pages 140 - 4
GRIFFITHS-JONES, NUCLEIC ACIDS RES, vol. 32, 2004, pages D109 - D111
HANAHAN ET AL., CELL, vol. 100, no. 1, 2000, pages 57 - 70
HUBER ET AL., BIOINFORMATICS, vol. 18, no. 1, 2002, pages 96 - 104
ITO ET AL., CANCER RESEARCH, vol. 53, 1993, pages 2940 - 3
JONES ET AL., CELL, vol. 128, no. 4, 2007, pages 683 - 92
KASTAN; BARTEK, NATURE, vol. 432, 2004, pages 316 - 23
KREK ET AL., NATURE GENETICS, vol. 37, 2005, pages 495 - 500
LE BRIGAND ET AL., NUCLEIC ACIDS RESEARCH, vol. 34, 2006, pages E87
LEWIS ET AL., CELL, vol. 120, 2005, pages 15 - 20
LUONG ET AL., CLIN CANCER RES, vol. 12, 2006, pages 5570 - 7
MASSAGUE, NATURE, vol. 432, 2004, pages 298 - 306
MITSIADES ET AL., CLIN CANCER RES, vol. 11, 2005, pages 3958 - 65
NGUYEN ET AL., CURR OPIN MOL THER, vol. 10, 2008, pages 158 - 67
POTIER ET AL., PLOS ONE, vol. 4, 2009, pages E6718
PRINCE ET AL., CLIN CANCER RES, vol. 15, no. 12, 2009, pages 3958 - 69
RABBANI AZRA ET AL: "The anthracycline antibiotics: antitumor drugs that alter chromatin structure.", BIOESSAYS : NEWS AND REVIEWS IN MOLECULAR, CELLULAR AND DEVELOPMENTAL BIOLOGY JAN 2005 LNKD- PUBMED:15612030, vol. 27, no. 1, January 2005 (2005-01-01), pages 50 - 56, XP002616101, ISSN: 0265-9247 *
SAITO ET AL., CANCER CELL, vol. 9, 2006, pages 435 - 43
SCHWEPPE ET AL., JOURNAL OF CLINICAL ENDOCRINOLOGY AND METABOLISM, vol. 93, 2008, pages 4331 - 41
SCHWEPPE ET AL., THE JOURNAL OF CLINICAL ENDOCRINOLOGY AND METABOLISM, vol. 93, no. 11, 2008, pages 4331 - 41
STIMSON ET AL., CANCER LETTERS, vol. 280, 2009, pages 177 - 183
WANG; DYMOCK, EXPERT OPIN. THER. PAT., vol. 19, 2009, pages 1727 - 1757
XIAO ET AL., MOLECULAR CANCER THERAPEUTICS, vol. 8, 2009, pages 350 - 6

Similar Documents

Publication Publication Date Title
Yu et al. miR-93 suppresses proliferation and colony formation of human colon cancer stem cells
Zhang et al. Co-suppression of miR-221/222 cluster suppresses human glioma cell growth by targeting p27kip1 in vitro and in vivo
Zhang et al. The tumor suppressive role of miRNA-370 by targeting FoxM1 in acute myeloid leukemia
Roberts et al. Expression analysis in multiple muscle groups and serum reveals complexity in the microRNA transcriptome of the mdx mouse with implications for therapy
Koval et al. Method for widespread microRNA-155 inhibition prolongs survival in ALS-model mice
Diao et al. miR-203, a tumor suppressor frequently down-regulated by promoter hypermethylation in rhabdomyosarcoma
Saito et al. Epigenetic alterations and microRNA misexpression in cancer and autoimmune diseases: a critical review
Li et al. MicroRNA-409-3p regulates cell proliferation and apoptosis by targeting PHF10 in gastric cancer
Li et al. MiR‐383 is downregulated in medulloblastoma and targets peroxiredoxin 3 (PRDX3)
Yungang et al. miR-370 targeted FoxM1 functions as a tumor suppressor in laryngeal squamous cell carcinoma (LSCC)
Jin et al. MiR-424 functions as a tumor suppressor in glioma cells and is down-regulated by DNA methylation
Liu et al. Mir-126 inhibits growth of SGC-7901 cells by synergistically targeting the oncogenes PI3KR2 and Crk, and the tumor suppressor PLK2
Liu et al. Identification of miRNomes in human stomach and gastric carcinoma reveals miR-133b/a-3p as therapeutic target for gastric cancer
D’Adamo et al. MicroRNAs and autophagy: fine players in the control of chondrocyte homeostatic activities in osteoarthritis
US20110190383A1 (en) Diagnostic, Prognostic and Therapeutic Uses of MIRs in Adaptive Pathways and/or Disease Pathways
AU2009228393B2 (en) Compositions and methods for diagnosing and treating melanoma
Ediriweera et al. Targeting miRNAs by histone deacetylase inhibitors (HDACi): Rationalizing epigenetics-based therapies for breast cancer
Gao et al. miR‑202 acts as a potential tumor suppressor in breast cancer
Chen et al. Long non-coding RNA (lncRNA) small nucleolar RNA host gene 15 (SNHG15) alleviates osteoarthritis progression by regulation of extracellular matrix homeostasis
Zhang et al. miR-206 inhibits metastasis-relevant traits by degrading MRTF-A in anaplastic thyroid cancer
US20170015999A1 (en) Compositions and methods for treatment of ovarian cancer
Alarcón‑Millán et al. Regulation of GKN1 expression in gastric carcinogenesis: A problem to resolve
US11013754B2 (en) Compositions and methods for treating cancer
Ding et al. The emerging role of small non-coding RNA in renal cell carcinoma
Stevanovic et al. Interplay of SOX transcription factors and microRNAs in the brain under physiological and pathological conditions

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10787107

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10787107

Country of ref document: EP

Kind code of ref document: A1