专利摘要:
Determination of methylation and levels of a mirna in response to an antitumor compound based on platinum. The present invention relates to a method for determining the response to a platinum-based antitumor compound in an ovarian cancer patient comprising (I) determining the level of methylation on the cpg island of sequence seq id no: 1 and (II) ) compare the level of methylation on said cpg island in the gene encoding mir-7 or the expression level of mir-7 with a corresponding reference value, where an increase in the level of methylation obtained in (I) or a decrease in the level of expression obtained in (I) with respect to the corresponding reference value, is indicative that the ovarian cancer of said patient is resistant to said platinum compound. The invention also relates to the use of mir-7 or a precursor thereof for the manufacture of a medicament for the treatment of a subject suffering from a cancer resistant to an anti-tumor compound based on platinum. (Machine-translation by Google Translate, not legally binding)
公开号:ES2596711A1
申请号:ES201530997
申请日:2015-07-09
公开日:2017-01-11
发明作者:Inmaculada IBÁÑEZ DE CÁCERES;Olga PERNÍA ARIAS;Javier De Castro Carpeño;Rosario PERONA ABELLÓN;Federico ROJO TODO;Olga VERA PUENTE;Julia JIMÉNEZ HERNÁNDEZ
申请人:Consejo Superior de Investigaciones Cientificas CSIC;Universidad Autonoma de Madrid;Fundacion para la Investigacion Biomedica del Hospital Universitario La Paz;Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz;
IPC主号:
专利说明:

Determination of the methylation and levels of a miRNA in response to a platinum-based antitumor compound FIELD OF THE INVENTION
The present invention relates to a method for determining the response to a platinum-based antitumor compound in an ovarian cancer patient and to the cancer treatment. BACKGROUND OF THE INVENTION
Ovarian cancer is the gynecological disease that causes the most deaths in countries
10 developed. With an incidence of 204,000 cases a year, it causes 125,000 deaths worldwide. The 5-year mortality is around 70-80%, being in most cases due to the progression and metastasis of the tumor.
The most common type of ovarian cancer is epithelial (90%). Among epithelial tumors there are benign tumors, tumors with low malignant potential or borderline tumors and
15 malignant tumors, whose prognosis depends primarily on grade (I-III) (Ovarian Cancer Detailed Guideline, ACS 2013). The appropriate surgical and pharmacological treatment for each type of tumor depends on its extension and its risk of progression, as in the rest of tumors; but in ovarian cancer the surgical decision should be as conservative as possible, in order to avoid the loss of fertility in patients.
20 Although most patients respond to drug treatment, many patients develop resistance to platinum-based antitumor compounds, resulting in rapid disease progression. The exact mechanism by which ovarian cancer cells become resistant to cisplatin treatment is currently unknown.
25 MicroRNAs are small non-coding RNAs (21-22 base pairs) occur naturally and primarily recognize the 3 'non-coding region of mRNA and inhibit protein synthesis.
Several recent studies indicate that deregulation of miRNAs and their target genes promote cancer onset, progression and drug resistance. For these
30 reasons have been attempted to identify the relationship of miRNAs with resistance to ovarian cancer.


It is known that levels of various miRNAs are increased in cisplatin-resistant ovarian cancer, for example mir-214 (Yang H. et al., Cancer Res. 2008 Jan 15; 68 (2): 425-33), mir -21 (Echevarría-Vargas IE. Et al., PLOS May 2014 Volume 9 Issue 5), and others whose levels are reduced in cisplatin-resistant ovarian cancer cells, for example miR-106a (Rao YM. Et al. , J Huazhong Univ Sci Technolog Med Sci. 2013 Aug; 33 (4): 567-72).
However, platinum resistance in ovarian cancer is a complex and multifactorial process, which involves several mechanisms and processes, and it is a challenge to predict it from a miRNA profile.
Therefore, there is a need in the art for methods to determine the response to a platinum-based antitumor compound in patients with ovarian cancer and the identification of new compounds for the treatment of platinum-based antitumor-resistant cancers. SUMMARY OF THE INVENTION
In a first aspect, the invention relates to a method for determining the response to a platinum-based antitumor compound in an ovarian cancer patient comprising
(i) determining the level of methylation on the CpG island of sequence SEQ ID NO: 1 in the gene encoding miR-7 or the expression level of miR-7 in a sample of said patient, and
(ii) compare the level of methylation on said CpG island in the gene encoding miR-7 or
the expression level of miR-7 with a corresponding reference value, where an increase in the level of methylation obtained in (i) or a decrease in the level of expression obtained in (i) with respect to the corresponding reference value, It is indicative that the ovarian cancer of said patient is resistant to said platinum compound.
In a second aspect, the invention relates to the use of miR-7 or a precursor thereof for the manufacture of a medicament for the treatment of a subject suffering from a cancer resistant to a platinum-based antitumor compound.
BRIEF DESCRIPTION OF THE FIGURES


Figure 1. Cisplatin response (CDDP) of the sensitive (S) and resistant (R) lines used in the H460S / R, H23S / R, A2780S / R and OVCAR3S / R study.
Figure 2: Quantifications of changes in expression of miRNAs in the lines used. All change their expression in at least one of the R (resistant) lines compared to the sensitive (S) lines, used as a gauge in the graph and are recovered after epigenetic reactivation (RT).
Figure 3: Cell viability after overexpression of the miRNA-7 precursor. This figure shows how the overexpression of miR-7 in resistant lines H23R and A2780R induces the 50% mortality of the cells in culture, so that their function could be closely linked to tumor progression. A: Controls the effectiveness of transfection of the miRNA-7 precursor in H23R and A2780R cells. B: Cell viability after transfection with the miRNA precursor in the H23R and A2780R lines.
Figure 4: Kaplan Meier curves showing the time to the first progression (TP) in months (upper figure) and overall survival time (OS) in months (lower figure). DETAILED DESCRIPTION
Method to determine the response
In a first aspect, the invention relates to a method for determining the response to a platinum-based antitumor compound in an ovarian cancer patient comprising
(i) determining the level of methylation on the CpG island of sequence SEQ ID NO: 1 in the gene encoding miR-7 or the expression level of miR-7 in a sample of said patient, and
(ii) compare the level of methylation on said CpG island in the gene encoding miR-7 or the
miR-7 expression level with a corresponding reference value, where an increase in the level of methylation obtained in (i) or a decrease in the level of expression obtained in (i) with respect to the corresponding reference value, is indicative that the ovarian cancer of said patient is resistant to said platinum compound.
The expression "determine the response of a patient" refers to the assessment of the response of a platinum-based antitumor therapy in a patient suffering from cancer.


"Platinum-based antitumor compound," as used in the present invention, refers to platinum-based antineoplastic drugs, commonly called platinum and that result in inhibition of DNA repair and / or DNA synthesis, as a consequence of the cross-linking of said agents with the DNA. Illustrative, non-limiting examples of platinum compounds are cisplatin, carboplatin, oxaliplatin, satraplatin, picoplatin, nedaplatin, triplatin, lipoplatin.
In a preferred embodiment the platinum-based antitumor compound is cisplatin, also known as cis-diaminodichloroplatin (II) or CDDP.
By "ovarian cancer", as used in the present invention it refers to a malignant tumor originating in any part of the ovary. The most frequent location is the epithelium that covers the ovary. It also develops from germ cells or connective tissue around the ovary.
In a particular embodiment ovarian cancer is ovarian carcinoma of epithelial origin.
By "ovarian carcinoma of epithelial origin", as used in the present invention, it refers to a type of cancer that derives from the epithelial surface that covers the surface of the ovary or endometrial tissue.
The person skilled in the art knows various methods to determine if a patient has ovarian cancer, among others by determining tumor markers such as CA-125, imaging studies, computed tomography, magnetic resonance imaging or ultrasound among others.
The term "patient", "subject", "individual" applied to cancer refers to a member of a species of a mammalian animal, and includes, but is not limited to, domestic animals, primates and humans; The subject is preferably a human being, male or female, of any age or race. In the case of ovarian cancer the patient is female.
In a first step, the method of the invention comprises determining the level of methylation in the CpG island of sequence SEQ ID NO: 1 in the gene encoding miR-7 in a patient sample.
As used herein, the term "CpG island" refers to a short DNA sequence rich in the CpG dinucleotide and that can be found in the 5 ′ region of approximately half of the genes. CpG islands are typically, but not always, between about 0.2 to about 1 kb in length. "CpG" is the abbreviation of "Cytosine-phosphate-Guanine", that is, cytosine and guanine separated by only


a phosphate; Phosphate binds any two nucleosides together in the DNA. The term "CpG" is used to distinguish this linear sequence from the pairing of cytosine and guanine CG bases. Cytosines in CpG dinucleotides may be methylated to form 5-methylcytosine.
5 The gene encoding mir-7 has several CpG islands. Specifically, the CpG island analyzed in the invention is found in the precursor mir-7-3 and is located on chromosome 19p13.3 positions 4769132-4769799, that is, the region between nucleotides 1550 and 883 before the start of the coding region from miR-7.
The term "methylation" as used herein, refers to the covalent attachment of a methyl group at the C5 position of the cytosine nucleotide base in the CpG dinucleotides of gene regulatory regions. The term "methylation status" or "methylation level" refers to the presence or absence of 5-methyl-cytosine ("5-mCyt") in
One or a plurality of CpG dinucleotides in a DNA sequence. As used herein, the terms "methylation status" and "methylation level" are used interchangeably.
The term "miRNA", as used in the present invention, also known as
20 "micro RNA" refers to a single stranded RNA molecule, usually about 21-25 nucleotides in length, with the ability to regulate gene expression at the post-transcriptional level by degrading the mRNA or by inhibiting the process of protein translation.
"MiR-7", as used in the present invention, refers to mature miRNA derived from three precursors miR-7-1, miR-7-2 and miR-7-3 whose human sequences correspond to numbers Access MI0000263, MI0000264 and MI0000265, respectively, from the miRBase database as of April 8, 2015. According to the invention, the determination of the level of methylation on the CpG island of SEQ ID
30 NO: 1 of the gene encoding miR-7 is performed on a sample of the patient.
The term "sample" or "biological sample", as used herein, refers to biological material isolated from a subject. The biological sample contains any biological material suitable for detecting the desired methylation pattern at one or more CpG site (s) and may comprise cells and / or non-cellular material of the subject. In the present invention, sample 35 comprises genetic material, e.g. eg, DNA, genomic DNA (gDNA), complementary DNA


(CDNA), RNA, heterogeneous nuclear RNA (mRNA), mRNA, etc., of the subject under study. In a particular embodiment, the genetic material is DNA. In a preferred embodiment, the DNA is genomic DNA. In another preferred embodiment, the DNA is circulating DNA. The sample can be isolated from any suitable biological tissue or fluid such as, for example, blood, saliva, plasma, serum, urine, cerebrospinal fluid (CSF), feces, a buccal or oropharyngeal swab, a sample, a sample obtained from of a biopsy, and a tissue sample included in paraffin. Methods for isolating cells and tissue samples are well known to those skilled in the art. In a particular embodiment, the sample is selected from the group consisting of blood, urine, saliva, serum, plasma, an oral or buco-pharyngeal swab,
10 hair, a surgical sample of the tumor, and a sample obtained from a biopsy. In a preferred embodiment, the sample is selected from blood, serum, hair, plasma, urine and saliva.
In a preferred embodiment the sample is a sample containing tumor cells, preferably a sample of the primary tumor, metastatic tissue or a biofluid.
The term "tumor sample", as used herein, refers to a tissue sample from the primary cancer tumor. "Primary tumor" refers to a tumor that has its origin in the tissue or organ in which it is found and has not metastasized to that location from another location. "Metastatic tissue", as used herein
The invention refers to a cancerous tissue in an organ other than that in which it started.
The tumor tissue sample can be obtained by conventional methods, for example, by biopsy, using methods well known to those skilled in related medical techniques. Methods for obtaining a biopsy sample 25 include partitioning into large pieces of a tumor, or microdissection or other cell separation methods known in the art. Tumor cells can be obtained additionally by aspiration cytology with a fine needle. To simplify the preservation and handling of the samples, these can be fixed in formalin and embedded in paraffin or frozen first and then embedded in a cryosolidifiable medium, such as
OCT compound, by immersion in a highly cryogenic medium that allows rapid freezing.
The tumor sample can be treated to physically or mechanically disintegrate the structure of the tissue or cell, to release the intracellular components in an aqueous or organic solution to prepare the nucleic acids for further analysis. Nucleic acids


they are extracted from the sample by procedures known to the person skilled in the art and commercially available.
In those cases where the DNA sample is not enclosed in a membrane (for example, circulating DNA of a blood sample), standard methods can be employed in the art for the isolation and / or purification of DNA. Such methods include the use of a protein degenerating reagent, for example, chaotropic salt, for example guanidinium hydrochloride or urea; or a detergent, for example, sodium dodecyl sulfate (SDS), cyanogen bromide. Alternative methods include, but are not limited to ethanol precipitation or propanol precipitation, vacuum concentration among others by means of a centrifuge. The person skilled in the art can also make use of devices such as filter devices, for example, ultracentrifugation, silica surfaces or membranes, magnetic particles, polystyrene particles, polystyrene surfaces, positively charged surfaces and positively charged membranes, charged membranes, surfaces loaded, charged membranes changed, loaded surfaces changed.
Once nucleic acids have been extracted, genomic double stranded DNA is used in the analysis. Methylation analysis can be carried out by any means known in the art. A variety of methylation analysis procedures are known in the art and can be used to practice the invention. These assays allow the determination of the methylation status of one or a plurality of CpG sites in a tissue sample. In addition, these methods can be used for absolute or relative quantification of methylated nucleic acids. Such methylation tests involve, among other techniques, two main steps. The first step is a specific methylation reaction or separation, such as (i) bisulfite treatment, (ii) methylation specific binding, or (iii) methylation specific restriction enzymes. The second main step involves (i) amplification and detection, or (ii) direct detection, by a variety of methods such as (a) PCR (sequence specific amplification) such as Taqman®, (b) DNA sequencing of DNA without treat and treat with bisulfite, (c) sequencing by ligation of modified probes with dyes (including cyclic ligation and cutting), (d) pyrosequencing, (e) sequencing of single molecules, (f) mass spectrometry, or (g) analysis by Southern type transfer.
In addition, restriction enzyme digestion of amplified PCR products of bisulfite converted DNA can be used, for example, the method described by Sadri and Hornsby (1996, Nucl. Acids Res. 24: 5058-5059), or COBRA ( Combined bisulfite restriction analysis) (Xiong and Laird, 1997, Nucleic Acids Res. 25: 2532-2534). The analysis


COBRA is a quantitative methylation assay useful for determining DNA methylation levels in specific gene loci in small amounts of genomic DNA. Briefly, restriction enzyme digestion is used to reveal differences in methylation-dependent sequences in PCR products of DNA treated with sodium bisulfite. Methylation dependent sequence differences are first introduced into genomic DNA by standard bisulfite treatment according to the procedure described by Frommer et al. (Frommer et al, 1992, Proc. Nat. Acad. Sci. USA, 89, 1827-1831). PCR amplification of bisulfite-converted DNA is then performed using specific primers for the CpG sites of interest, followed by restriction endonuclease digestion, gel electrophoresis and detection using specific labeled hybridization probes. Methylation levels in the original DNA sample are represented by the relative amounts of digested and undigested PCR product in a linearly quantitative manner across a broad spectrum of DNA methylation levels. In addition, this technique can be applied reliably to DNA obtained from microdissected paraffin embedded tissue samples. Typical reagents (for example, as could be found in a typical COBRA-based kit) for COBRA analysis may include, but are not limited to: PCR primers for specific genes (or DNA sequence altered by methylation or CpG islands) ; restriction enzymes and appropriate buffer; oligo hybridization with genes; oligo hybridization control; kinase labeling kit for the oligo probe; and radioactive nucleotides. In addition, bisulfite conversion reagents may include: DNA denaturation buffer; sulfonation buffer; DNA recovery reagents or kits (eg, precipitation, ultrafiltration, affinity column); desulfonation buffer; and DNA recovery components.
In a preferred embodiment, the level of CpG island methylation of SEQ ID NO: 1 is determined using specific methylation PCR (MSP). MSP allows you to assess the methylation status of virtually any group of CpG sites on a CpG island, independent of the use of methylation-sensitive restriction enzymes. Briefly, the DNA is modified by sodium bisulfite which converts the cytosines without methylation, but not the methylated ones, to uracil and subsequently amplified with specific primers for methylated versus nonmethylated DNA. MSP requires only small amounts of DNA, is sensitive to 0.1 percent of methylated alleles of a given CpG island locus, and can be performed on DNA extracted from samples embedded in paraffin. Alternatively, quantitative multiplexed methylation specific PCR (QM-PCR) can be used.


In one embodiment, the methylation profile of selected CpG sites is determined using the MethyLight and Heavy Methyl methods. The MethyLight and Heavy Methyl assays are a high performance quantitative methylation assay that uses real-time fluorescence PCR technology (Taq Man®) that does not require additional manipulations after the PCR step. Briefly, the MethyLight process begins with a mixed sample of genomic DNA that is converted, in a reaction with sodium bisulfite, to a mixed set of methylation-dependent sequence differences according to standard procedures (the bisulfite process converts cytosine residues without methylation to uracil). PCR is then performed with fluorescence either in a "non-biased" PCR reaction (with primers that do not overlap with known CpG methylation sites) or in a "biased" reaction (with PCR primers that overlap with known CpG dinucleotides). Sequence discrimination can occur at the level of the amplification process or at the level of the fluorescence detection process, or both. The MethyLight assay can be used as a quantitative test for methylation patterns in the genomic DNA sample, where sequence discrimination occurs at the level of probe hybridization. In this quantitative version, the PCR reaction provides a biased amplification in the presence of a fluorescent probe that overlaps with a particular putative methylation site. Unbiased control is provided for the amount of input DNA by a reaction in which neither the primers nor the probe cover any
20 nucleotide CpG. Alternatively, a qualitative test for genomic methylation is achieved by testing the biased PCR set with control oligonucleotides that do not "cover" known methylation sites (a fluorescent version of the "MSP" technique) or with oligonucleotides that cover potential methylation sites .
Specific detection probes for methylated and unmethylated sites with two
25 different fluorophores provide a simultaneous quantitative measure of methylation. The "Heavy Methyl" technique begins with the bisulfite conversion of DNA. Then specific blockers prevent amplification of unmethylated DNA. Methylated genomic DNA does not bind to blockers and their sequences will be amplified. The amplified sequences are detected with a specific methylation probe.
30 The Ms-SNuPE technique is a quantitative method for assessing methylation differences at specific CpG sites based on DNA bisulfite treatment, followed by single nucleotide primer extension. Briefly, genomic DNA is reacted with sodium bisulfite to convert unmethylated cytosine to uracil while leaving 5-methylcytosine unchanged. The sequence amplification is then performed
Desired target using specific PCR primers for bisulfite converted DNA, and


The resulting product is isolated and used as a template for methylation analysis at the CpG site (s) of interest. Small amounts of DNA (for example, microdissected pathological sections) can be analyzed and avoids the use of restriction enzymes to determine the state of methylation at CpG sites.
In another embodiment, the methylation status of selected CpG sites is determined using methylation detection methods based on differential binding. For the identification of differentially methylated regions, one approach is to capture methylated DNA. This approach uses a protein, in which the MBD2 methyl binding domain is fused to the Fc fragment of an antibody (MBD-FC). This fusion protein has several advantages over conventional methylation specific antibodies. MCB-FC has a higher affinity for methylated DNA and binds to double stranded DNA. Most importantly, the two proteins differ in the way they bind DNA. Methylation-specific antibodies bind to DNA stochastically, which means that only a binary response can be obtained. The MBD-FC methyl binding domain, on the other hand, binds to DNA molecules regardless of their methylation status. The strength of this protein-DNA interaction is defined by the level of DNA methylation. After binding to genomic DNA, eluate solutions of increasing salt concentrations can be used to fractionate methylated and unmethylated DNA allowing for more controlled separation. Therefore, this method, called immunoprecipitation of methyl-CpG (MCIP), not only enriches, but also divides genomic DNA according to the level of methylation, which is particularly positive when the fraction of unmethylated DNA must also be investigated.
Alternatively, antibodies against 5-methylcytidine can be used to bind and precipitate methylated DNA. Antibodies are available from Abeam (Cambridge, MA), Diagenode (Sparta, NJ) or Eurogentec (c / o AnaSpec, Fremont, 30 CA). Once the methylated fragments are separated, they can be sequenced using microarray based techniques such as the methylated CpG island recovery test (MIRA) or methylated DNA immunoprecipitation (MeDIP) or other new generation sequencing techniques (NGS). Another technique is that of the methyl-CpG binding domain / segregation of partially molten molecules (MBD / SPM).
Alternatively, there are methyl-sensitive enzymes that preferentially or substantially cut or digest in their DNA recognition sequence if it is not methylated. Therefore, an unmethylated DNA sample will be cut into smaller fragments than a methylated DNA sample. Similarly, a sample of hypermethylated DNA will not be cut. In


Instead, there are methyl-sensitive enzymes that cut in their DNA recognition sequence only if it is methylated. Methyl-sensitive enzymes that digest unmethylated DNA suitable for use in the methods of the technology include, but are not limited to, HpaII, HhaI, MaeII, BstUI and AciI. An enzyme that can be used is HpaII that cuts only the CCGG sequence without methylation. Another enzyme that can be used is HhaI that cuts only the GCGC sequence without methylation. Both enzymes are available from New England BioLabs®, Inc. Combinations of two or more methyl sensitive enzymes that digest only unmethylated DNA can also be used. The cutting methods and procedures for the restriction enzymes selected to cut DNA at specific sites are well known to the person skilled in the art. For example, many restriction enzyme suppliers provide information on the conditions and types of DNA sequences cut by specific restriction enzymes, including New England BioLabs, Pro-Mega Biochems, Boehringer-Mannheim, and the like.
The MCA technique is a method that can be used to screen altered methylation patterns in genomic DNA, and to isolate specific sequences associated with these changes. Briefly, restriction enzymes with different sensitivities to cytosine methylation are used at their recognition sites to digest genomic DNA from primary tumors, cell lines and normal tissues before PCR amplification with arbitrary primers. The fragments showing differential methylation are cloned and sequenced after resolving the PCR products in high resolution polyacrylamide gels. The cloned fragments are then used as probes for Southern analysis to confirm the differential methylation of these regions. Typical reagents (for example, as could be found in a typical MCA-based kit) for MCA analysis may include, but are not limited to: PCR primers for arbitrary priming of genomic DNA; PCR buffers and nucleotides, restriction enzymes and appropriate buffers; oligos or gene hybridization probes; oligos or probes for control hybridization.
In another embodiment, the methylation status of selected CpG sites is determined using high resolution methylation-sensitive fusion (HRM). A variety of commercially available real-time PCR machines have HRM systems including Roche LightCycler480, Corbett Research RotorGene6000 and Applied Biosystems 7500. HRM can also be combined with other amplification techniques such as pyrosequencing.
In another embodiment, the methylation state of the selected CpG locus is determined using a primer extension assay, including a reaction of


Optimized PCR amplification that produces amplified targets for analysis using mass spectrometry. The test can also be done in multiplex. Mass spectrometry is a particularly effective method for the detection of polynucleotides associated with differentially methylated regulatory elements. The presence of the polynucleotide sequence is verified by comparing the mass of the detected signal with the expected mass of the polynucleotide of interest. The relative strength of the signal, for example, peak mass in a spectrum, for a particular polynucleotide sequence indicates the relative population of a specific allele, thus allowing the ratio of the allele to be calculated directly from the data. For the methylation analysis, the assay can be adopted to detect methylation-dependent C to T sequence changes introduced with bisulfite. These methods are particularly useful for performing multiplexed amplification reactions and multiplexed primer extension reactions (e.g., multiplexed homogenous primer mass extension assays (hME)) in a single well to further increase the yield and reduce the cost per reaction. for primer extension reactions.
Other methods for DNA methylation analysis include genomic scanning of restriction marks, analysis of representative differences sensitive to methylation (MS-RDA), comprehensive high performance matrix techniques for relative methylation (CHARM). Roche® NimbleGen® microarrays include chip chromatin immunoprecipitation (ChIP-chip) or chip-methylated DNA immunoprecipitation (MeDIP-chip).
After the reaction or separation of the nucleic acid in a specific manner of methylation, the nucleic acid can be subjected to sequence based analysis.
Alternatively, the method of the invention comprises in a first stage determining the level of miR-7 expression in a sample of said patient.
The term "expression level", as used in the present invention, refers to the expression level of miRNA, in particular miR-7. Expression levels of miR-7 are determined in a sample of the subject.
For the determination of the level of miR-7 expression, it is necessary to obtain RNA from the sample of the affected ovarian cancer subject to be analyzed. Techniques for the purification of RNA from a sample of a subject are widely known to those skilled in the art. Total RNA can be purified from a sample by homogenization in the presence of a nucleic acid extraction buffer, followed by centrifugation. Nucleic acids precipitate and DNA is removed by treatment


with DNAase and precipitation. Nucleic acids, specifically RNA and specifically miRNA, can be isolated by any technique known to those skilled in the art. There are two main methods to isolate RNA: (i) phenol-based extraction and (ii) silica matrix or glass fiber filter (GFF) binding. Phenol-based reagents contain a combination of denaturing and RNAase inhibitors for the breakdown of cells and tissues and the subsequent separation of RNA from contaminants. Phenol-based isolation procedures can recover RNA species in the range of 10-200 nucleotides for example, miRNA, ribosomal RNA (rRNA) and small nuclear RNA (snRNA). If a sample of total RNA was purified by the GFF procedure or conventional silica matrix column, small-sized RNAs may have been lost. However, extraction procedures such as those using Trizol or TriReagent will purify all RNAs, large and small, and are the recommended methods to isolate total RNA from biological samples that will contain miRNA. Any method required for the treatment of a sample before quantification of the expression level of miR-7 is within the scope of the present invention. Similarly, the use of commercial kits for RNA purification, in particular miRNA, including, without limitation, miRNeasy Mini kit from Qiagen, miRNA isolation kits from Life Technologies, mirPremier microRNA isolation kit from Sigma-Aldrich and High Pure miRNA isolation kit from Roche, in particular commercial kits for RNA purification, in particular miRNA, without limitation, PAXgene blood miRNA kit from Qiagen.
Once an RNA preparation is available from a sample to be analyzed, the method of the invention requires determining the expression levels of mir-7 in the RNA isolated from said sample. Methods for determining miRNA expression levels in cells or biological samples include generic methods for the detection and quantification of nucleic acids, especially RNA, optimized methods for the detection and quantification of small RNA species, since both mature miRNAs and precursors fall within this category, as well as methods specially designed for the detection and quantification of miRNA. Illustrative, non-limiting examples of methods that can be employed to determine the levels of one or more miRNAs include hybridization-based methods, such as Northern blot analysis and in situ hybridization, real-time multiplex and / or singleplex RT-PCR (reagents available from, for example, Applied Biosystems and System Biosciences (SBI)), including quantitative real-time reverse transcriptase PCR (qRT-PCR), individual molecule detection, pearl-based flow cytometry methods, and assays using nucleic acid matrices.


While all techniques of gene expression profile determination (RT-PCR, SAGE -Serial Analysis of Gene Expression, or serial analysis of gene expression-, expression microarrays or TaqMan) are suitable in the present invention, levels RNA expression are frequently determined by reverse transcription-polymerase chain reaction (RT-PCR). In a particular embodiment, expression levels are determined by quantitative PCR, preferably real-time PCR. Detection can be carried out in individual samples or in tissue microarrays.
In a particular embodiment, the expression level of mirR-7 is determined by real-time quantitative RT-PCR (qRT-PCR), a modification of the polymerase chain reaction (PCR) used to quickly measure the amount of a PCR product. This is preferably done in real time, therefore it is an indirect method to quantitatively measure starting quantities of DNA, complementary DNA or RNA. This is commonly used to determine if a genetic sequence is present or not, and if the number of copies in the sample is present. Like other forms of PCR, the procedure is based on the amplification of DNA samples, using thermal cycles and a thermostable DNA polymerase. The three commonly used quantitative PCR methods are: by agarose gel electrophoresis, by using SYBR Green (a double stranded DNA dye) and by a fluorescent indicator probe. The last two methods can be analyzed in real time, thus constituting real-time PCR methods.
The fluorescent indicator probe method is the most accurate and the most reliable of the methods. Use a sequence-specific nucleic acid-based probe, so that it only quantifies the sequence that hybridizes with the probe and not all double stranded DNA. Said probe, which has at its 3 ’end a fluorophore and at its 5’ end a molecule that blocks its fluorescence emission (quencher or quencher), specifically hybridizes to the central part of the PCR product to be obtained. Thus, when PCR is performed with the probe plus the pair of specific primers, the probe hybridizes in the amplicon but, due to the proximity of the fluorophore to the damper, fluorescence is not emitted; when the polymerase begins to synthesize the complementary chain for the single-stranded template DNA primed, as the polymerization progresses, it reaches the probe attached to its complementary sequence, so that the polymerase hydrolyzes the probe through its 5'-3 'exonuclease activity , thereby separating the fluorescent indicator and the switch. This results in an increase in the fluorescence that is detected. During the thermal cycles of the real-time PCR reaction, the increase in fluorescence is monitored at


as it is released from the double labeled hydrolyzed probe in each PCR cycle, which allows the precise determination of the final and initial DNA quantities.
Any PCR method that allows to determine the expression of miR-7 is within the scope of the present invention.
5 The determination of the level of expression by PCR requires the use of probes. A probe is a defined sequence oligonucleotide capable of specifically hybridizing with a complementary sequence of a nucleic acid, so it can be used to detect and identify complementary or substantially complementary sequences in nucleic acids. The length of the probe of the invention may vary.
10 within a wide range although, for practical reasons, probes of small length minus 10 nucleotides, preferably at least 15 nucleotides, preferably at least 20 nucleotides, preferably at least 25 nucleotides and, preferably not more than 100 nucleotides, more preferably are preferred between 15 bases and 30 bases, preferably between 16 bases and 22 bases.
Alternatively or additionally, the oligonucleotides used may contain modified bonds such as phosphodiester, phosphotriester, phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoranilothioate, phosphoramidate, methylphosphonate, boranophosphonate links, as well as combinations of the same or peptides (peptides). nucleic acids, PNA), in which the different nucleotides are linked by
20 amida links.
The use of probes of greater or lesser length would not affect the sensitivity or specificity of the technique, but could require the realization of a series of modifications of the conditions on which the same is performed by varying their melting temperature and its GC content, which would affect the temperature and time
25 hybridization fundamentally.
The expression levels of miR-7 can be quantified by comparison with an internal standard, for example, the level of messenger RNA of a maintenance or housekeeping gene present in the same sample, as a control gene or normalizing gene . Preferably, the normalizing gene is a gene whose level of expression does not change in a cell, such as a maintenance gene that encodes a protein that expresses itself is constitutive and performs essential cellular functions. Maintenance genes suitable for use as internal standards include, but are not limited to, myosin, β-2-microglobulin, ubiquitin, 18S ribosomal protein, cyclophilin,


glyceraldehyde-3-phosphate dehydrogenase (GADPH) and actin. Alternatively, an RNA may be taken as normalizer including, without limitation, snRNA U6 or snRNA RNU48.
The method of the invention requires, in a second step, to compare the level of methylation in the CpG island of sequence SEQ ID NO: 1 in the gene encoding miR-7 or the expression level of miR-7 obtained in a sample of a subject with the corresponding reference value.
The term "reference value", as used in the present invention, refers to a value obtained in the laboratory and used as a reference for the values or data obtained by laboratory tests of the patients or samples collected from patients. The reference value or reference level may be an absolute value, a relative value, a value that has an upper and / or lower limit, a range of values, an average value, a median value, or a value in comparison to a specific control or reference value. The reference value may be based on a value of the individual sample, such as a value obtained from a sample of the subject being tested, but at an earlier time. The reference value may be based on a large number of samples, such as population values of subjects of the same age group, or it may be based on a set of samples, including or excluding the sample to be tested.
The reference value corresponding to a miR-7 according to the present invention is an expression value of said miRNA. The expression reference value of a miRNA according to the invention can be determined by techniques well known in the state of the art, for example, by isolating RNA from each sample in the collection, determining the expression levels of said miRNA in each RNA. isolated and calculating the average expression levels of said miRNA in each sample. Alternatively, the reference value could be determined by measuring the miRNA expression levels in an RNA sample obtained by mixing equal amounts of RNA from each of the samples in the aforementioned collection. The collection of samples to be analyzed to calculate the reference value is preferably derived from a population of two or more subjects; for example, the population may comprise 3, 4, 5, 10, 15, 20, 30, 40, 50 or more subjects.
In a particular embodiment, the reference value corresponds to the value obtained in a sample located at a distance from the tumor tissue, by way of illustration at least 3 cm of the tumor tissue.


In another particular embodiment the reference value is obtained in a sample of a subject suffering from ovarian cancer sensitive to a platinum compound, preferably a sample of the primary tumor. In another particular embodiment the reference value is obtained in a sample of a healthy subject that does not have cancer.
Finally, the method of the invention comprises correlating a variation in the level of methylation in the CpG island of sequence SEQ ID NO: 1 in the gene encoding miR-7 or the expression level of miR-7 obtained with the response to a platinum based anti-tumor compound. In particular, an increase in the level of methylation obtained in step (i) or a decrease in the level of expression obtained in step (i) of the method of the invention, is indicative that the ovarian cancer of said patient is resistant to said platinum compound.
According to the present invention, an increase in the level of methylation refers to an increase in the number of molecules that exhibit methylation in a given CpG region, resulting in an increase in the average of methylated molecules in a given position.
In accordance with the present invention, the level of methylation of one or more CpG site (s) is increased when the level of methylation of said one or more CpG site (s) in a sample is higher than in the reference sample. The level of methylation of one or more CpG site (s) is considered to be higher than in the reference sample when it is at least 1.5%, at least 2%, at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 100%, at least 110%, at least 120%, at least 130%, at least 140%, at least 150% or higher than in the reference sample.
Similarly, in the context of the present invention, the level of methylation of one or more CpG site (s) is decreased when the level of methylation of said one or more CpG site (s) in a sample is less than a value. reference. The level of methylation of one or more CpG site (s) is considered to be lower than in the reference sample when it is at least 5%, at least 10%, at least 15%, at least 20%, at least 25% at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75% at least 80%, at least 85%, at least 90%, at least 95%, at least 100%, at least 110%, at least 120%, at least 130%, at least 140%, at least 150% or more lower than the reference sample.


In the context of the present invention, the expression "decreased expression level with respect to a reference value" refers to any statistically significant variation in the expression level of a miRNA below its corresponding reference level, in particular to any variation in the expression level of miR-7 below its corresponding reference level. A variation of the expression level of miR-7, below the reference value may be at least 0.95 times, 0.9 times, 0.75 times, 0.2 times, 0.1 times, 0.05 times, 0.025 times, 0.02 times, 0.01 times, 0.005 times or even less compared to the corresponding reference expression value.
The expression "increased expression level with respect to a reference value" refers to any statistically significant variation in the expression level of a miRNA above its corresponding reference level, in particular any variation in the expression level of miR -7 above its corresponding reference level, and / or any variation in the expression level of miR-7 above its corresponding reference level. A variation of the expression level of a miR-7, above its corresponding reference value may be at least 1.05 times, 1.1 times, 1.5 times, 5 times, 10 times, 20 times, 30 times, 40 times, 50 times, 60 times, 70 times, 80 times, 90 times, 100 times or even more compared to the corresponding reference expression value.
Medical uses
In another aspect, the invention relates to the use of miR-7 or a precursor thereof for the manufacture of a medicament for the treatment of a subject suffering from a cancer resistant to a platinum-based antitumor compound.
Alternatively, the invention relates to miR-7 or a precursor thereof for use in the treatment of a subject suffering from resistant cancer a platinum-based antitumor compound.
Alternatively, the invention relates to a method of treating a cancer resistant to a platinum-based antitumor compound in a subject by the administration of miR-7 or a precursor thereof.
The terms miR-7, subject, and platinum-based antitumor compound have been described above and are equally applicable to this aspect.


The term "treatment", as used herein, refers to any type of therapy, which aims to terminate, improve or reduce the susceptibility to suffer from a clinical condition, as described herein. Thus, "treatment," "treating," and its equivalent terms refer to obtaining a desired pharmacologically or physiologically desired effect, which covers any treatment of a pathological condition or disorder in a mammal, including humans. The effect can be prophylactic in terms of providing total or partial prevention of a disorder and / or adverse effect attributable to it. That is, "treatment" includes (1) inhibiting the disease, for example by stopping its development, (2) interrupting or ending the disorder or at least the symptoms associated with it, so that the patient would no longer suffer the disease or its symptoms, for example causing the regression of the disease or its symptoms by restoring or repairing a lost, absent or defective function, or stimulating an inefficient process,
or (3), reduce, alleviate or improve the disease, or the symptoms associated with it, where reducing is used in a broad sense to refer to at least a reduction in the magnitude of a parameter, such as inflammation, pain , or immune deficiency.
In a particular embodiment, the subject suffering from cancer resistant to a miR-7 treated platinum compound or a precursor thereof, is not treated with a platinum compound.
Within the context of the present invention, the term "cancer" includes any type of cancer or tumor. Illustrative, non-limiting examples of such cancers or tumors include hematological cancers (eg, leukemia or lymphomas), neurological tumors (eg, astrocytomas or glioblastomas), melanoma, breast cancer, lung cancer, head and neck cancer, gastrointestinal tumors (eg, stomach, pancreatic or colorectal cancer (CRC)), liver cancer (eg, hepatocellular carcinoma), renal cell cancer, genitourinary tumors (eg, ovarian cancer, vaginal cancer, cervical cancer, cancer of bladder, testicular cancer, prostate cancer), bone tumors, vascular tumors, etc. In a particular embodiment, said cancer is ovarian, lung, colon and pancreas cancer. More particularly, ovarian cancer is ovarian carcinoma of epithelial origin and lung cancer is non-small cell lung cancer.
In a preferred embodiment the platinum-based antitumor compound is cisplatin.
The expert knows various methods to know if the cancer to be treated according to the present invention is resistant to a platinum-based antitumor compound.


The microRNA (miRNA) is produced from a precursor microRNA (pre-miRNA), which in turn is formed from a primary microRNA transcript (pri-miRNA).
Thus, by "miR-7 precursor" as used in the present invention, both pre-miR-7 and pri-miR-7 are included. In a particular embodiment the precursor of miR-7 is selected from miR-7-1, miR-7-2 and miR-7-3.
The invention contemplates the administration of one or more miR-7 precursors alone or in combination with miR-7.
The invention relates both to the administration of miR-7 as such or its precursors and to the administration of a polynucleotide comprising the sequence encoding miR7 or a precursor thereof.
Thus, in another embodiment miR-7 or a precursor thereof is administered by a polynucleotide comprising the sequence encoding miR-7 or a precursor thereof.
The expression "sequence encoding miR-7 or a precursor thereof" is a polynucleotide that comprises the sequence encoding the pri-miRNA sequence, premiRNA for miR-7 or the mature miR-7 sequence. The polynucleotide comprising the mature, pre-miRNA, or pri-miRNA sequence can be single stranded or double stranded. The polynucleotides may contain one or more chemical modifications, such as blocked nucleic acids, peptide nucleic acids, modifications with sugar, such as 2'-O-alkyl (e.g., 2'-O-methyl, 2'-O-methoxyethyl modifications ), 2'-fluoro, and 4'-thio, and modifications of the main structure, such as one or more phosphorothioate, morpholino or phosphonocarboxylate junctions. In some embodiments, the polynucleotide is conjugated to a steroid, such as cholesterol, a vitamin, a fatty acid, a carbohydrate or glycoside, a peptide, or other small molecule ligand.
Alternatively, the DNA molecule encoding the miR-7 or precursor thereof is in an expression cassette.
The expression cassette comprises one or more regulatory sequences, selected based on the cells to be used for expression, operably linked to a polynucleotide encoding the miR-7 or precursor thereof. "Operationally linked" is intended to mean that the nucleotide sequence of interest (ie, a DNA encoding the miR-7 or precursor thereof) binds to the regulatory sequence (s) in a way that allows nucleotide sequence expression (for example, in a system of


transcription / translation in vitro or in a cell when the cassette or expression vector is introduced into a cell). "Regulatory sequences" include promoters, enhancers and other expression control elements (eg, polyadenylation signals). Regulatory sequences include those that direct the constitutive expression of a nucleotide sequence in many types of host cells and those that direct the expression of the nucleotide sequence only in certain host cells (eg, tissue-specific regulatory sequences). Those skilled in the art will appreciate that the design of the expression cassette may depend on factors such as the choice of host cell to be transformed, the level of expression of the miR-7 or precursor thereof, and the like. Such expression cassettes typically include one or more sites properly located for restriction enzymes, to facilitate the introduction of nucleic acid into a vector.
Appropriate promoter and / or regulatory elements can be readily selected to allow expression of the miR-7 or precursor thereof in the cell of interest. In certain embodiments, the promoter used to direct the intracellular expression of a miR-7 or precursor thereof is a promoter for RNA polymerase III (Pol III). According to other embodiments, a promoter for RNA polymerase I can be used, for example, a tRNA promoter.
In a preferred embodiment, the polynucleotide encoding miR-7 or a precursor thereof is in a vector.
Various vectors are known in the art and can be easily modified to direct the transcription of miR-7 or a precursor thereof. When double-stranded miRNA precursors are synthesized in vitro, the chains can be allowed to hybridize before introducing them into a cell or prior to administration to a subject. miR-7 or precursors thereof can be supplied or introduced into a cell as a single RNA molecule that includes autocomplementary parts (for example, an hRNA that can be processed intracellularly to produce a miRNA), or as two chains hybridized to each other. In other embodiments, miR-7 or the precursor thereof is transcribed in vivo. Regardless of whether the miRNA or precursor thereof is transcribed in vivo or in vitro, in any scenario, a primary transcript can be produced that can then be processed (for example, by one or more cellular enzymes) to generate the miRNA that achieves gene inhibition. .


In another approach, miRNAs can be expressed by attenuated viral vectors or hosts or bacterial vectors. Recombinant vaccinia virus, adeno-associated virus (VAA), herpesvirus, retrovirus, or other viral vectors vectors can be used.
In one example, a viral vector is used. These vectors include, but are not limited to, adenovirus, herpesvirus, vaccinia, or an RNA virus such as a retrovirus. In one example, the retroviral vector is a derivative of a murine or avian retrovirus. Examples of retroviral vectors into which a single foreign gene can be inserted include, but are not limited to: Moloney murine leukemia virus (VLMuMo), Harvey murine sarcoma virus (VSMuHa), murine mammary tumor virus (VTMMu ) and Rous sarcoma virus (RSV). When the subject is a human being, a vector such as the gibbon monkey leukemia virus (VLMG) can be used. Several additional retroviral vectors can incorporate multiple genes. All these vectors can transfer or incorporate a gene for a selection marker so that transduced cells can be identified and generated. By inserting a nucleic acid sequence that codes for miR-7 or precursor thereof into the viral vector, together with another gene that codes for the ligand for a receptor in a specific target cell, for example, the vector is now target specific . Target specific retroviral vectors can be prepared by linking, for example, a sugar, a glycolipid or a protein. Preferred target selection is achieved using an antibody to select the retroviral vector as the target. Those skilled in the art will know how, or can easily determine without undue experimentation, specific polynucleotide sequences that can be inserted into the retroviral genome or attached to a viral envelope to allow specific target delivery of the retroviral vector containing the polynucleotide encoding miR -7 or a precursor thereof.
miR-7 or a precursor thereof for the manufacture of the medicament for the treatment according to the invention may further comprise a pharmaceutically acceptable carrier.
A "pharmaceutically acceptable carrier", "pharmaceutically acceptable diluent", or "pharmaceutically acceptable carrier", or "pharmaceutically acceptable carrier", used interchangeably herein, refers to a non-toxic filler, encapsulation material or formulation, solids, semi-solids or auxiliary liquids of any conventional type. A pharmaceutically acceptable carrier is essentially non-toxic to the receptors at the dosages and concentrations employed, and is compatible with other components of the formulation. For example, the carrier for a formulation containing polypeptides would not normally include oxidizing agents and other compounds known to be harmful to the polypeptides. The bearers


Suitable include, but are not limited to water, dextrose, glycerol, saline, ethanol and combinations thereof. The carrier may contain additional agents such as wetting or emulsifying agents, pH buffering agents or adjuvants that enhance the effectiveness of the formulation.
5 miR-7 or a precursor thereof for the manufacture of a medicament for use according to the present invention can be administered by any suitable route of administration, for example an oral, sublingual, topical, rectal or parenteral route including subcutaneous route. , intraperitoneal, intradermal, intramuscular, intravenous. intravascular, intratumoral, intracranial, intrathecal, intrasplenic, intramuscular, subretinal, and mucosa. In
A preferred embodiment is administration intratumorally or intravenously.
The present invention includes all forms of nucleic acid delivery, including synthetic oligonucleotides, naked DNA, plasmid and viral, whether or not integrated into the genome.
Both miRNAs and polynucleotides encoding the miRNAs of interest can
Introduced into the desired host cells by methods known in the art, including but not limited to transfection, electroporation (eg, transcutaneous electroporation), microinjection, transduction, cell fusion, DEAE-dextran, calcium phosphate precipitation, use of a gene gun, or by lipofection.
Both miRNAs and polynucleotides encoding miRNAs can be administered in a single dose, or multiple doses separated by a time interval.
The therapeutic amounts for therapeutic use may depend on the severity of the disease and age, weight, general condition of the patient and other clinical factors. Therefore, the final determination of the appropriate treatment regimen will be made by the attending physician.
The invention is now described in detail by means of the following examples that should be considered as merely illustrative and not limiting the scope of the invention.
EXAMPLES
Materials and methods
1. Cellular models


To carry out this study, two human tumor cell lines have been used: non-small cell lung cancer (NSCLC) and ovarian cancer.
• H23. Cell line from a patient not treated with non-small cell lung adenocarcinoma, obtained from the ATCC (American type culture collection; Manassas, VA, USA), CRL-5800. H460 Undifferentiated NSCLC cell line derived from the pleural fluid of a patient, obtained from the ATCC, HTB-177. A2780 Cell line from an ovarian carcinoma of epithelial origin, obtained from the ECACC (European Collection of Cell Cultures; Sigma-Aldrich, Madrid, Spain), 93112519. OVCAR3. Ovarian adenocarcinoma cell line of epithelial origin, obtained from the ATCC, HTB-161.
Obtaining the resistant subtypes of each of the cell lines described was carried out in previous works, subjecting them to increasing doses of CDDP (Ibanez de Caceres, I. et al Oncogene 29, 1681–1690 (2010). obtained four pairs of sensitive (S) and resistant (R) cell lines: H23S / H23R, H460S / H460R, A2780S / A2780R and OVCAR3S / OVCAR3R. These lines were cultured in RPMI medium 10% FBS (fetal bovine serum) supplemented with 4 ml of Glutamine, 4 ml of Fungizona and 350 µl of Gentamicin.On the other hand, the four resistant lines (H23R, H460R, A2780R and OVCAR3R) were treated with the demethylating agent 5aza-dC and the histone deacetylase inhibitor TSA2 , thus obtaining a third group of treated resistant lines (RT): H23RT, H460RT, A2780RT and OVCAR3RT.
2. Selection of candidate miRNAs
The selection of the candidate miRNAs was carried out following the criteria described below. Initially, those miRNAs were selected that had a decreased expression pattern in R (with respect to S) and recovered in RT in at least 2 of the 4 lines that were analyzed. From there, those who possess a CpG island in their promoter region or, in the case of intragenic miRNAs, in the promoter region of the gene in which they are encoded were selected. Next, the selection was made based on the cellular pathways in which the target genes of each of the selected miRNAs participate, in search of those involved in tumor progression and response to chemotherapy. The bioinformatic resources that were used were the following:
• GeneCards (www.genecards.org) and miRBase (www.miRBase.org). Genecards integrates information from more than 100 databases about all known human genes. miRBase contains information on all miRNA gene sequences


known, their location and the mature sequences thereof. These two databases were used for a first search for information on the miRNAs in the list.
• Ensembl (www.ensembl.org). Ensembl constitutes a genome information center for various species, focusing on the human species, through the collection, creation and organization of data resources. With this database we obtained information about the region of the genome in which the miRNAs (inter or intragenic) are found, as well as the elements adjacent to them. It also allows to know the cDNA sequences of the promoter regions to check the presence of CpG islands.
• CpG Island Searcher (www.cpgislands.com) and Webgene (www.itb.cnr.it/webgene). The criteria and algorithms that use these bioinformatics tools for the prediction of CpG islands were proposed by Takai and Jones: GC ≥ 55%, CpGObs / CpGEsp ≥ 65 and a length> 200 bp (Takai, D. & Jones, PA Proc. Natl Acad. Sci. USA 99, 3740–3745 (2002) In all cases the presence of CpG islands was verified by both resources.
• WebGestalt (bioinfo.vanderbilt.edu/webgestalt). This analysis tool is designed for large-scale functional studies, based on a large list of genes and their respective miRNAs described. Specifically, Gene Ontology Tree Machine (GOTM) Zhang, B., et al., BMC Bioinformatics 5, 16 (2004), a web tool for the analysis and visualization of gene sets, was used for the functional classification of the genes obtained. of interest based on gene ontology hierarchies.
3. Expression analysis by qRT-PCR
The validation of the expression change seen in the arrays of the different miRNAs in the three experimental groups (S, R and RT) of each of the cell lines was performed by qRT-PCR from RNA samples that were extracted following the guanidine thiocyanate method using TRIZOL (Invitrogen), and which were purified by the RNeasy Kit (Qiagen).
To obtain the cDNA of the miRNAs, 11 ng of purified total RNA was used and the TaqMan® MicroRNA Reverse Transcription kit (ID: 4366596, Applied Biosystems) was used, together with the specific oligos for retrotranscription of each of the assessed miRNAs (TaqMan® test numbers) as well as miR-RNU48 (ID: 001006, Applied Biosystems). The latter was used as an endogenous control because its expression is constitutive, that is, its levels do not vary as a result of


treatments received by the cells. The specific program used followed the following parameters: 30 min at 16 ° C; 30 min at 42 ° C; 5 min at 85 ° C and a final temperature of 4 ° C. The final product was stored at -20 ° C until use.
Once the cDNA was obtained, the expression levels between S, R and RT of each of the lines were compared by quantitative PCR, using TaqMan® quantitative expression assays (Applied Biosystems). For this, TaqMan® Universal PCR Master Mix (ID: 4304437, Applied Biosystems) and TaqMan® Small RNA Array (20X) probes were used for quantitative PCR specific to each miRNA. In each reaction 1.5 µl cDNA was used, in a final volume of 20 µl. Each sample was analyzed in triplicate at least twice, using the 7900HT Fast Real-Time PCR System (Applied Biosystems), with a program that followed the following parameters: 10 min at 95 ° C and forty amplification cycles of 15 s at 95 ° C and 1 min at 60 ° C. With this technique the fluorescence emitted after each amplification cycle is measured, with which the quantification is performed in real time, thus allowing to estimate the relative amount of the sequence in the different samples during the logarithmic phase of the reaction Higuchi, R. , et al.,. Bio / Technology 11, 1026-30 (1993).
The relative quantification of gene expression was performed with the RQ Manager (Life Technologies) program, which is based on the 2-ΔΔCt comparative method (Livak KJ et a., Methods San Diego Calif 25, 402–408 (2001) by that the relative expression changes of each sample or treatment are calculated with respect to a reference (in this case untreated controls) In addition the expression of all samples is previously normalized with respect to the expression of the endogenous control miR-RNU48. they are presented as the “change of expression in number of times” (RQ) and the error bars are expressed as the maximum calculation (RQmax) and the minimum calculation (RQmin) of the expression levels, representing the standard deviation of the mean of the expression level RQ.
4. DNA modification by bisulfite, subsequent sequencing of CpG islands and specific methylation PCR in patient samples (MSP)
The extraction of DNA from the different experimental groups of each line, in addition to DNA from normal tissue samples used as controls (lung, ovary, saliva and lymphocytes) and from the tumor lines PANC-1 and IMIMPC3 (pancreas), LOVO and HT29 (colon), OV4 and SKOV3 (ovary), PC-3 and LNCAP (prostate), BT474 (breast), SW780 (bladder) H727 and A549 (lung), was performed using conventional K proteinase digestion techniques (Invitrogen) and extraction with phenol-chloroform.


For the modification of the samples, 1 µg of genomic DNA that was denatured with NaOH (0.2 M) was started for 10 min at 37 ° C, then the DNA was modified by treating it with hydroquinone and sodium bisulfite at 50 ° C for 17 h under mineral oil layer. The modified DNA was purified using Wizard DNA Clean-Up system (Promega).
5 To complete the modification, the DNA was treated with NaOH (0.3 M) for 5 minutes at room temperature and then precipitated with glycogen, ammonium acetate (10 M) and ethanol. DNA modification results in the conversion of unmethylated cytosines into uracils, while those that are methylated are resistant to modification and therefore continue to be cytosines.
In the bisulfite sequencing, fragments of the modified DNA of the lines and of the relevant controls, which were between 355 and 560 bp and that contained all or part of the CpG islands of the promoters of each miRNA, were amplified by PCR. The PCR reactions were performed under the following conditions: a) 1 cycle at 94 ° C for 5 min, b) 40 cycles of 1 min at 94 ° C, 1 min between 60-61 ° C and 1 min at 72 ° C. Finally c) one
15 extension of 10 min at 72 ° C. To run the PCR products, 1.5% agarose gels were used and the bands were purified with MinElute Gel Extraction Kit (Qiagen). In all cases, direct sequencing was performed using the ABI 3100A genetic analyzer. The data obtained were analyzed with Sequencher 5.1 software.
OLIGOS miR-7-3miR-132miR-335miR-148
Sec. By Bisulfite Direct5-TTAGGAAGAAGT TAGGAGGGGAA A-3 (SEQ ID NO: 2)5-GTTTTAGGTTTTTA YGGGAGTTTA-3 (SEQ ID NO: 10)5-TGGGAAAGAGGA GGTGAGAAA-3 (SEQ ID NO: 14)5-GTYGTTT TTTTTTTA GTTAGGA GATA-3 (SEQ ID NO: 16)
Back 5-CAAACACCTCAA ACCACCCTCT-3 (SEQ ID NO: 3)5-TACRACCRCRACT CCTACACACT-3 (SEQ ID NO: 11)5-CGCTTCCTAAAAC CAAAAATTCT-3 (SEQ ID NO: 15)5-CCRCTCC CTTCCAT CTTAACT3 (SEQ ID NO: 17)
Amplicon size 372bp441bp528bp560bp
Sec. By Bisulfite 2 Direct5-TAGTTGGGGAA GTTTTTTTTAGG A-3 (SEQ ID NO: 4)5-AGGAGTYGYGGTY GTATGAATGA-3 (SEQ ID NO: 12)
Back 5-TTTCCCCTCCTA5-GTCTCCTAAAACR


ACTTCTTCCT-3 (SEQ ID NO: 5) CCAACACCTT-3 (SEQ ID NO: 13)
Amplicon size 428bp441bp
MSP Not Methylated Direct5-GGGGTGGGGTT TTTTAAGAATT-3 (SEQ ID NO: 6)
Back 5-ATAACATTCTCC TCCTTCAATCA-3 (SEQ ID NO: 7)
Amplicon size 131bp
MSP Methylated Direct5-GGGTGGGGTTT TTTAAGAATC -3 (SEQ ID NO: 8)
Back 5-ACATTCTCCTCC TTCGATCG-3 (SEQ ID NO: 9)
Amplicon size 127bp
Table 1. Oligos used to determine the methylation of miR-7
For amplification assays with specific methylation PCR (MSP), bisulfite modified DNA from the analyzed patients was amplified with miRNA 5 specific oligos for methylated DNA versus unmethylated DNA for miRNA-7. As a negative control we use DNA from healthy lung tissue, of which we know that it does not have the methylated miRNA, and as a positive control we use lymphocyte DNA from patients, without any previous important pathology, artificially methylated in our laboratory with the enzyme S-adenosylmethionine (SAM ). The reaction was carried out with a program with the
10 following parameters: 5 minutes at 95 ° C and 2 minutes at 80 ° C, at which time we incorporate the DNA polymerase followed by 35 cycles with 1 minute at 95 ° C, 1 minute at 58 ° C and 50 seconds at 72 ° C followed by a final extension of 8 minutes at 72 ° C. The PCR products were separated on a 5% polyacrylamide gel under non-denaturing conditions.
15 5. Cellular transfection with the miR-7 precursor and associated cell viability
To study the existence of a true connection between miR-7 expression and cell viability, functional tests were performed using precursors of miR-7, hsa


miR-7-5p (PremiR hsa-miR-7, ID: PM10047, Life technologies) to induce its expression, which are chemically modified double stranded RNAs that mimic endogenous miRNAs and allow functional analysis of the regulation of miRNA activity. Overexpression was performed in H23R and A2780R, since it is the cell lines in which the expression of miR-7 was validated by qRT-PCR after treatment with cisplatin. Negative controls were also transfected on the same cell lines, H23R and A2780R, which serves as an internal control of miR-7 expression. Transfection with the miR-7 precursor was carried out using lipofectamine 2000 (Invitrogen), as recommended by the commercial house. The amount of negative control and transfected miR-7 went to
10 a concentration of 50 nM per p60.
The response in cell viability after transfection of the miR-7 precursor was assessed in the H23R (RM) and A2780R (RM) cells transfected with said precursor compared to the corresponding resistant transfected with the negative control A2780 (RC and RC) to identify if miR-7 is acting as a possible tumor suppressor. This test is carried out in quadruplicate, sowing 80,000 cells per well of the four experimental groups in 24-well plates and then treating them with the negative control.
or with the precursor of miR-7 for 72 hours. Subsequently, the cells are fixed and stained with violet crystal, assessing the intensity of the staining by an ELISA test, calculating the proportion of staining in the cells transfected with the miR-7 precursor with respect to
20 controls, following the method previously described by Chattopadhyay (Chattopadhyay, S .. Oncogene 25 (23), 3335-3345 (2006))
EXAMPLE 1 Identification of candidate miRNAs
The platinum resistance of the cell lines used in the study can be seen in Figure 1.
IC50 (µg / ml) ± SD CDDP-IRP value
H23S 0.28 ± 0.01--
H23R 0.94 ± 0.093.35<0.001
A2780S 0.15 ± 0.08--
A2780R 0.45 ± 0.043.00<0.001
H460S 0.46 ± 0.37--
H460R 1.60 ± 0.262.50<0.001
OVCAR3S 0.60 ± 0.05--
OVCAR3R 1.50 ± 0.142.96<0.001


Table 2. IC50 against the agent Cisplatin and Resistance Index (IR).
Among all the miRNAs identified in the expression arrays, 129 were found with a decreased pattern of expression in the resistant lines (R), as a result of the resistance acquired after cisplatin exposures, expression that was partially recovered, after epigenetic reactivation (RT) treatment. On the other hand, of all the genes analyzed in the complete genome expression arrays, 2755 genes were found with an expression pattern contrary to that sought in the tests with miRNAs, that is, expression increased in R and decreased in RT, which also they presented a p-value
10 adjusted FDR less than 0.05.
Of those 129 miRNAs, those that only presented the pattern of expression described in 1 of the 4 lines analyzed were discarded, so the list of candidates was reduced to 56 miRNAs. Next, the promoter region of each miRNA was analyzed to find the presence of some CpG island with the characteristics described by Takai and Jones, supra and
15 found that 14 of the 56 miRNAs had a CpG island in the 2000 bp before and after the start of their sequence, and in another 5 it was in the 2000 bp before and after the transcription start site of the gene in which they were They found encoded. That is, in a total of 19 miRNAs the presence of at least one potentially regulatory CpG island was found.
20 To continue with the identification of the best candidate miRNAs, a third filtering was carried out based on the ontological grouping of the possible target genes of the 19 identified miRNAs and thus have a global vision of the potential involvement of the miRNA in tumor progression and response to chemotherapy. . 19 analyzes were performed based on the information obtained with Gene Ontology Tree Machine (GOTM) using two listings for
25 each miRNA studied, one of them with the 2755 genes selected from the array and the other with the genes regulated by each of the 19 miRNAs, obtained by sequence complementarity, as detailed in section 3 of the materials and methods. This tool compares and analyzes both listings by marking those genes that, with a level of significance p <0.005, have an appearance greater than expected if they had been chosen
30 at random. This allowed identifying routes with involvement in cell growth and proliferation, apoptosis, membrane transport or chromatin remodeling, among others, that could be regulated by the same miRNA. This resulted in the selection of 10 candidate miRNAs for subsequent epigenetic validation.


EXAMPLE 2. Validation of the expression changes of the selected miRNAs
The results obtained by qRT-PCR show the expression changes in the 3 experimental groups (S, R and RT) of the 10 selected miRNAs. Each miRNA was validated in the 4 lines (H23, H460, A2780 and OVCAR3), although in Figure 2 only the lines in which significant changes of expression had been previously identified in the arrays are represented, to simplify their understanding.
In 8 of the 10 candidate miRNAs there was a decrease in R expression with respect to S and a recovery in RT after epigenetic treatment, in the H23 line in the expression arrays, data that were fully validated by qRT-PCR . In the H460 line, in the arrays, 8 of the 10 miRNAs with the defined expression pattern were also found, 7 of them shared with the other H23 NSCLC line, of which the miR-148a and the miR- were fully validated 335. In the rest of the miRNAs analyzed, except in miR-340, there is partial validation, since in miR-7, miR-9, miR-10a and miR-132 the expression in RT increases, although there were no significant changes in the experimental group R, and in the miR-124 a decrease of the expression in R was registered, but no restatement was observed after epigenetic treatment. The miR-340 is the only miRNA in which no changes were validated. In the case of the A2780 ovarian cancer line, in the arrays the pattern of expression defined in 6 of the 10 candidate miRNAs was observed, and in 3 of them it was validated by qRT-PCR: miR-7, miR-10a and miR-132 In miRNAs 124 and 203, no significant expression changes were observed between sensitive and resistant cells, however in both cases there is an increase in expression in epigenetically treated cells. MicroRNA 149 has no relevant expression changes in this line. Finally, OVCAR3 is the line in which, according to the arrays, the lowest number of miRNAs with specific expression changes was observed, and in fact only partial validations are observed, since miR-124 and 132 are reactivated in RT but there is no decrease in expression in R, and in miR-149 there is a decrease in R but there is no recovery in RT.
In total, 4 miRNAs are validated in at least 2 lines: miR-148a and miR-335 in the CPNM lines H23 and H460, and miR-132 and 7 in H23 and in the ovarian cancer line A2780. And miR-7 in 2 of the 3 that predicted expression arrays (H23 and A2780). These miRNAs were chosen for the next phase of the study, the assessment of the methylation status of its regulatory CpG island.
EXAMPLE 3. Methylation status of the CpG islands analyzed


The study of the methylation status of the CpG islands was performed in the 4 miRNAs selected after validation of the expression changes by qRT-PCR, miR-148a, miR335, miR-132 and miR-7. The miR-148a is located on the short arm of chromosome 7 and has a 1663 bp CpG island, located 150 bp from the first nucleotide of its sequence. A 560 bp region of the CpG island was analyzed in the experimental groups S and R of the H23 and H460 lines, in addition to normal lung tissue. The results of bisulfite sequencing showed no methylation in any of the tumor lines or in the normal tissue controls examined. The miR-335 is found on the long arm of chromosome 7, in the second intron of transcript 002 of the MEST gene. In the promoter region of the gene there is an 1123 bp CpG island, of which a 528 bp fragment was analyzed. The analyzes were done on the HNCP and H23S, R, H460S and R lines of CPNM, in addition to normal lung tissue. The results showed methylation in the H460 line, both in S and R. However, the same did not happen with H23S or R or with the normal lung. Additionally, the DNA methylation status of 3 tumor lines (LOVO, OV4 and PC-3) and a sample of normal saliva was analyzed. The results of this analysis did not show methylation in any of the cases. MicroRNA 132 is located on the short arm of chromosome 17. In its promoter region, also encompassing the miRNA sequence and part of the posterior region, there is a 2080 bp CpG island. The analyzed region covers 866bp and was performed using 2 pairs of oligos (SEQ ID NO: 10 and 11; SEQ ID NO: 12 and 13). In this case, the lines analyzed were H23 (S and R) and A2780 (S and R), together with normal lung and ovarian tissue. In all cases the results showed absence of methylation. The miR-7 has 2 CpG islands in its promoter region. One of them is located at a distance of 861 bp from the first nucleotide of the miRNA and has a length of 667 bp. The second island has an extension of 269 bp and encompasses the miRNA. For the analysis of the first island, 2 25 pairs of oligos were designed (SEQ ID NO: 2 and 3; SEQ ID NO: 4 and 5) covering a total of 776 bp, including the entire island and part of the areas adjacent. The study was conducted on lines H23 and A2780, in sensitive and resistant cells in both cases, in addition to normal lung and ovarian tissue controls. The sequencing results showed the presence of methylation in the resistant cells of the H23 and A2780 lines. The 30 lung and ovarian controls had no methylation. These results were complemented by the analysis of 11 tumor lines, among which are: BT474, SKOV3, LOVO, IMIMPC2 and SW780. The results in these lines show the presence of the same methylation pattern in 2 lines, LOVO and IMIMPC2, and absence of methylation in the others. On the other hand, from the second island a region of 355 bp in the lines was analyzed
35 H23S and R, A2780S and R, together with the normal lung and ovarian tissue controls. In all cases the sequences were methylated. 33


In summary, of the four validated miRNAs, especially 2 of them (miR-335, miR-7) have specific methylation on a CpG island near the region where they are encoded.
EXAMPLE 4. The methylation of miR-7 is a frequent event in samples of patients with early NSCLC.
The assessment of miR-7 methylation in 36 samples of lung cancer patients indicates: 1) that this miRNA methylation may be a frequent event in patients with lung cancer (53% of affected individuals) and that it could be a prognosis of the future development of lung cancer since it also appears in samples of patients with non-tumor diseases (emphysema).
EXAMPLE 5. Overexpression of miR-7 induces a mortality of approximately 60% in platinum resistant lines.
The correct transfection of miR-7 was validated by qRT-PCRs only in cases where an inhibited expression in Resistant cells had previously been validated in the arrays data. Thus, it was found that after the test a true overexpression of miR-7 occurs, in order to ensure that a correct transfection has taken place and that the miR-7 precursor acts correctly.
After transfection of the miR-7 precursor, a cell viability test was carried out to determine the response produced as a result of the overexpression of said microRNA in the H23R and A2780R lines, since they are those in which an expression was validated inhibited resistance of mir-7. At zero time we obtained a high mortality of the treated cells as the precursor of the miR-7 against the controls, reaching levels of 63% of cell viability in H23 and 52% in A2780 (Figure 3).
EXAMPLE 6. The methylation of miRNA-7 is a potential biomarker predictive of recurrence and overall survival in patients with ovarian cancer.
The assessment of miR-7 methylation in 83 patients with ovarian cancer indicates that these patients present in a frequency of 29% of affected women, but nevertheless it is closely related to a worse response to treatment with platinum derivatives and less time to tumor progression. The data indicate that 50% of patients with methylated miR-7 who receive platinum therapy resort before 18 months. On the other hand, 75.5% of women without relapse of the disease have unmethylated miR-7. In addition, overall survival in women with no methylation


of this marker, it is 40 months higher than the group of patients with methylated miR-7 (> 3 years) (Figure 4 and Table 3).
Complete Series (n = 83) Not methylated (n = 59)Methylated (n = 24)
features No. of patients%No. of patients%No. of patients%P
Age (median, range) 55 (17-84)59 (17-80)55 (18-84)0.88
Menopausal status 0.565
Premenopause 3. 441.02. 339.0eleven45.8
Postmenopause 4959.03661.01354.2
Parity 0.974
Do not 2428.91728.8729.2
Yes 5971.14271.21770.8
Family history 0.684
Do not 5869.94271.21666.7
Yes 2530.11728.8833.3
ECOG scale 0.025
0 twenty-one25.3twenty33.9one4.2
one 3643.42237.31458.3
2 twenty24.11220.3833.3
3 67.258.5one4.2
Ascites 0.025
Do not 4756.63864.4937.5
Yes 3643.4twenty-one35.6fifteen62.5
Tumor Grade 0,346
I 3. 4412745.8729.2
II 2428.9fifteen25.4937.5
III 2530.11728.8833.3
Histology 0.851
Serous 4959.03. 457.6fifteen62.5
Mucinous 910.8711.928.3
Clear Cell 89.658.5312.5
Others 1720.51322.0416.7
Chemotherapy 0.956
Adjuvant 5971.14372.91666.7
Neoadjuvant 67.235.1312.5
Metastatic 1821.71322.0520.8
Relapse 0.286
Do not 4959.03762.71250.0


Yes 3. 441.02237.31250.0
Exitus 0.119
Do not 49593864.4eleven45.8
Yes 3. 441twenty-one35.61354.2
Table 3. Demographic table showing the tumor characteristics of 83 patients with ovarian cancer and the methylation status of miRNA-7.
The term "Sequence listing" in the sequence list refers to "Sequence list", "Artificial Sequence" to "Artificial sequence" and the term "DNA" refers to "DNA".

权利要求:
Claims (1)
[1]
1-A method for determining the response to a platinum-based antitumor compound in an ovarian cancer patient comprising
5 (i) determine the level of methylation on the CpG island of sequence SEQ ID NO: 1 in the gene encoding miR-7 or the expression level of miR-7 in a sample of said patient, and
(ii) compare the level of methylation on said CpG island in the gene encoding miR-7 or the expression level of miR-7 with a reference value
10, where an increase in the level of methylation obtained in (i) or a decrease in the level of expression obtained in (i) with respect to the corresponding reference value, is indicative that the ovarian cancer of said patient is resistant to said platinum compound.
The method according to claim 1, wherein the platinum-based antitumor compound is cisplatin.
3-Method according to any of claims 1 or 2, wherein the ovarian cancer is an ovarian carcinoma of epithelial origin.
4-Method according to any one of claims 1 to 3 wherein the level of methylation on the CpG island of the gene encoding mir-7 is determined by specific methylation PCR.
5-Method according to any of claims 1 to 3 wherein the level of expression of mir-7 is determined by qRT-PCR.

TO
B
Fig. 1

D
Fig. 1 (cont.)

Fig 2

Expression levels delmiRNA Log10
Cell viability (%)
2. 3. 4 S miRcontrol R miRcontrol R miR7
one
0
-one H23A2780
S-C
R-C
R-M 25 0
Fig. 3

<110> FOUNDATION FOR BIOMEDICAL RESEARCH OF THE HOSPITALUNIVERSITY OF LA PAZ
5 SUPERIOR SCIENTIFIC RESEARCH COUNCILAUTONOMOUS UNIVERSITY OF MADRIDJIMÉNEZ DÍAZ FOUNDATION HEALTH RESEARCH INSTITUTE
<120> Determination of the methylation and levels of a miRNA in response to a platinum-based antitumor compound
<130> P11783ES00
<160> 17
<170> PatentIn version 3.5
<210> 1
<211> 668 20 <212> DNA
<213> Artificial Sequence
<220>
<223> CpG island in the gene encoding miR-7
<400> 1cgcctcggca gccacgggac acctgcatct gccaacaaga ctggaagcag gtgaggcaca 60
cagaggggga ggcccgcagc tgcgtgggag gaggggtggt ctgagggacg tgggatgccg 120
ggaatgaggc tggtttgcag gttggcgcat ggacattttc ccagaaaggg acagagacgg 180
cgaagtttga cggtctggaa agcagagacc agcagggctg actgcttggg aggtaagttc 240
35 tggggacatg gtacagggtg aggagcaggt atcagtgcta gttgcgaccc ctctgtgtct 300
cccccccgcc accccattgc cattctgaag ctccccagga agaagctagg aggggaaata 360
one

aattgagtgg gggtggggtt tcccaagaat cggaggaacc gagaacgaag aggggtgggg 420
gaacggggaa agagagagga aaatcaagtt ttcttcagca cgagggacag ctctccaccg 480
accgaaggag gagaatgcta tttatttcag caccaaatat ccggacagcg cctctcggga 540
ggtccgagaa gagaaccgcg atctgtttca gcaccggggc tcaggacagt tcccagcggg 600
10 ctccgtttcg tctccagaac cctggacagc tcctccaggt aacgggagag ccctttgacc 660
ctgatttt 668
15 <210> 2
<211> 24
<212> DNA
<213> Artificial Sequence
20 <220>
<223> Oligo direct miR-7-3 bisulfite sequencing
<400> 2ttaggaagaa gttaggaggg gaaa 24
<210> 3
<211> 22
<212> DNA 30 <213> Artificial Sequence
<220>
<223> Oligo reverse mir-7-3 bisulfite sequencing
35 <400> 3 caaacacctc aaaccaccct ct 22
2

<210> 4
<211> 24
<212> DNA5 <213> Artificial Sequence
<220>
<223> oligo direct mir-7-3 bisulfite 2 sequencing
10 <400> 4 tagttgggga agtttttttt agga 24
<210> 5 15 <211> 22
<212> DNA
<213> Artificial Sequence
<220> 20 <223> Oligo reverse mir-7-3 bisulfite sequencing 2
<400> 5tttcccctcc taacttcttc ct 22
<210> 6
<211> 22
<212> DNA
<213> Artificial Sequence
<220>
<223> Oligo direct miR-7-3 MSP not methylated
<400> 6 35 ggggtggggt tttttaagaa tt 22
3

5 <210> <211> <212> <213> <220> <223>7 23 Artificial DNA Sequence Oligo reverse miR-7-3 MSP not methylated
10 <400> 7 ataacattct cctccttcaa tca2. 3
fifteen <210> <211> <212> <213>8 21 Artificial DNA Sequence
twenty <220> <223> Oligo direct miR-7-3 MSP methylated <400> 8 gggtggggtt ttttaagaat ctwenty-one
25 <210> <211> <212> <213>9 20 Artificial DNA Sequence
30 <220> <223>Oligo reverse miR-7-3Methylated MSP
35 <400> 9 acattctcct ccttcgatcgtwenty
<210> 10
4

<211> <212> <213> 24 Artificial DNA Sequence
5 <220> <223>Oligo direct miR-132 bisulfite sequencing
10 <400> 10 gttttaggtt tttaygggag ttta24
fifteen <210> <211> <212> <213>11 23 Artificial DNA Sequence
<220> <223> Oligo reverse miR-132 bisulfite sequencing
twenty <400> 11 tacraccrcr actcctacac act2. 3
25 <210> <211> <212> <213>12 23 Artificial DNA Sequence
30 <220> <223>Oligo direct miR-132 sequencing by bisulfite 2
<400> 12 aggagtygyg gtygtatgaa tga 2. 3
35 <210> <211>13 23
5

<212> <213> Artificial DNA Sequence
5 <220> <223>Oligo reverse miR-132 bisulfite 2 sequencing
<400> 13 gtctcctaaa acrccaacac ctt 2. 3
10 15 <210> <211> <212> <213> <220> <223>14 21 Artificial DNA Sequence Oligo direct miR-335 bisulfite sequencing
twenty <400> 14 tgggaaagag gaggtgagaa atwenty-one
25 <210> <211> <212> <213>15 23 Artificial DNA Sequence
30 <220> <223> Oligo reverse miR-335 bisulfite sequencing <400> 15 cgcttcctaa aaccaaaaat tct2. 3
35 <210> <211> <212>16 26 DNA
6

<213> Artificial Sequence
<220>
<223> Oligo direct miR-148 bisulfite sequencing
<400> 16gtygtttttt ttttagttag gagata 26
10 <210> 17
<211> 21
<212> DNA
<213> Artificial Sequence
15 <220>
<223> Oligo reverse miR-148 bisulfite sequencing
<400> 17ccrctccctt ccatcttaac t 21
7
类似技术:
公开号 | 公开日 | 专利标题
US20190376143A1|2019-12-12|Microrna assay for detection and management of pancreatic cancer precursors
ES2443230T3|2014-02-18|New markers for cancer
US20150126374A1|2015-05-07|Hypermethylated gene markers for head and neck cancer
JP5867757B2|2016-02-24|Cancer detection method and detection kit, and cancer therapeutic agent
Zheng et al.2011|Genetic variants at the miR-124 binding site on the cytoskeleton-organizing IQGAP1 gene confer differential predisposition to breast cancer
Zhou et al.2014|Methylation of OPCML promoter in ovarian cancer tissues predicts poor patient survival
US20140271455A1|2014-09-18|Dna methylation biomarkers for small cell lung cancer
US20140154303A1|2014-06-05|Treating cancer by inhibiting expression of olfm4, sp5, tob1, arid1a, fbn1 or hat1
WO2013148151A1|2013-10-03|Plasma microribonucleic acids as biomarkers for endometriosis and endometriosis-associated ovarian cancer
KR20110015013A|2011-02-14|Methods for assessing colorectal cancer and compositions for use therein
Zhang et al.2019|MicroRNA-596 acts as a tumor suppressor in gastric cancer and is upregulated by promotor demethylation
CN106906290B|2019-12-20|CDSN as diagnosis and treatment target of tongue squamous cell carcinoma
Jiang et al.2014|Transcription factor WT1 and promoter CpG hypomethylation coactivate HOXA10 expression in ovarian cancer
TWI539002B|2016-06-21|Biomarker dact1 for gastric cancer
ES2596711B1|2017-10-27|Determination of the methylation and levels of a miRNA in response to a platinum-based antitumor compound
WO2019035100A2|2019-02-21|Prognostic markers for cancer recurrence
WO2011015040A1|2011-02-10|Methods and compositions for the diagnosis and prognosis of cervical intraepithelial neoplasia and cervical cancer
CN110387423B|2021-03-05|Biomarker for diagnosing vestibular nerve sheath tumor
CN109371136B|2021-07-02|Lung adenocarcinoma-related lncRNA and application thereof
CN108220446B|2020-06-30|Application of LINC01356 as molecular marker in gastric cancer
CN111518913A|2020-08-11|Non-coding RNA-LINC01819 for diagnosis and treatment of gastric cancer
CN108707672B|2020-02-18|Application of DUXAP8 in diagnosis and treatment of hepatocellular carcinoma
Wu et al.2021|Hypermethylation of the PZP gene is associated with hepatocellular carcinoma cell proliferation, invasion and migration
CN106702009B|2021-07-02|Application of gene related to lung adenocarcinoma
Lee et al.2011|MicroRNA-23a: a novel serum based diagnostic biomarker for lung adenocarcinoma
同族专利:
公开号 | 公开日
WO2017005957A1|2017-01-12|
ES2596711B1|2017-10-27|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题
US20130096185A1|2008-06-24|2013-04-18|Heiko Hermeking|INACTIVATION OF miR-34a BY ABERRANT CpG METHYLATION IN CANCER|
US20130059015A1|2010-03-11|2013-03-07|H. Lee Moffitt Cancer Center & Research Institute|Human Cancer micro-RNA Expression Profiles Predictive of Chemo-Response|
WO2013056217A1|2011-10-14|2013-04-18|The Ohio State University|Methods and materials related to ovarian cancer|
US9598734B2|2010-04-29|2017-03-21|Medical Prognosis Institute A/S|Methods and devices for predicting treatment efficacy|
EP2908830A4|2012-10-18|2016-11-30|Univ Western Australia|CANCER THERAPY USING miRNAs|EP3681515A1|2017-09-15|2020-07-22|Fundación para la Investigación Biomédica del Hospital Universitario de la Paz |Mafg as a potential therapeutic target to restore chemosensitivity in platinum-resistant cancer cells|
法律状态:
2017-10-27| FG2A| Definitive protection|Ref document number: 2596711 Country of ref document: ES Kind code of ref document: B1 Effective date: 20171027 |
优先权:
申请号 | 申请日 | 专利标题
ES201530997A|ES2596711B1|2015-07-09|2015-07-09|Determination of the methylation and levels of a miRNA in response to a platinum-based antitumor compound|ES201530997A| ES2596711B1|2015-07-09|2015-07-09|Determination of the methylation and levels of a miRNA in response to a platinum-based antitumor compound|
PCT/ES2016/070516| WO2017005957A1|2015-07-09|2016-07-08|Determination of methylation and mirna-7 levels for predicting the response to a platinum-based antitumor compound|
[返回顶部]