WO2022022541A1 - Utilisation d'un gène rbm10 - Google Patents

Utilisation d'un gène rbm10 Download PDF

Info

Publication number
WO2022022541A1
WO2022022541A1 PCT/CN2021/108791 CN2021108791W WO2022022541A1 WO 2022022541 A1 WO2022022541 A1 WO 2022022541A1 CN 2021108791 W CN2021108791 W CN 2021108791W WO 2022022541 A1 WO2022022541 A1 WO 2022022541A1
Authority
WO
WIPO (PCT)
Prior art keywords
rbm10
gene
protein
mrna
ovarian cancer
Prior art date
Application number
PCT/CN2021/108791
Other languages
English (en)
Chinese (zh)
Inventor
山松
潘德思
乔伟
姚何
周游
宁志强
Original Assignee
深圳微芯生物科技股份有限公司
成都微芯药业有限公司
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 深圳微芯生物科技股份有限公司, 成都微芯药业有限公司 filed Critical 深圳微芯生物科技股份有限公司
Publication of WO2022022541A1 publication Critical patent/WO2022022541A1/fr

Links

Images

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57449Specifically defined cancers of ovaries
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • 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
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • 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
    • C12Q2600/156Polymorphic or mutational markers

Definitions

  • the present invention relates to the technical field of medicine, in particular to the use of RBM10 gene.
  • Ovarian cancer ranks third among gynecological malignant tumors, and its mortality ranks first, and its incidence is increasing year by year.
  • Ovarian cancer patients can achieve complete remission (CR) after ideal cytoreduction (residual tumor lesions ⁇ 1 cm) combined with platinum and paclitaxel systemic chemotherapy, but 60%-70% of patients will be Relapse occurs.
  • Relapsed patients are less effective for re-chemotherapy: platinum-sensitive relapsed populations have an effective rate of about 30-80% for platinum-containing chemotherapy.
  • platinum-sensitive patients will become platinum-resistant after 2 to 3 platinum-containing treatments; platinum-refractory
  • the effective rate of platinum-free single-agent chemotherapy is only 5-17%, the progression-free survival time is about 3 months, and the overall survival time is less than 1 year.
  • the relevant research mainly includes:
  • GOG0170D study a phase II study of bevacizumab monotherapy in patients with platinum-refractory/resistant ovarian cancer, enrolled 62 subjects, ORR was 21%, of which 2 (3.2%) CR, 11 Case (17.7%) PR. Median PFS and OS were 4.7 and 17 months, respectively.
  • the GOG0170F study a phase II study of sorafenib monotherapy in ovarian cancer patients with recurrence and disease progression less than 12 months from the last platinum-containing chemotherapy, enrolled 71 patients, including platinum-sensitive 21 (29.6%) and platinum-sensitive Drug resistance was found in 50 cases (70.4%), including 59 evaluable cases. Two patients (3.4%) achieved PR, ORR was 3.4%, and median PFS and OS were 2.1 and 16.3 months, respectively.
  • the VEG104450 study pazopanib monotherapy for patients with high recurrence risk ovarian cancer with elevated CA125 after initial treatment, enrolled 36 subjects, of whom 14 (39.0%) were platinum-resistant and 22 (61.0%) were platinum-resistant. %) platinum sensitive.
  • the response rate was 11 (31.0%), and the response rate of the evaluable cases according to the RECIST standard was 0.
  • PARP polyadenosine diphosphate ribose polymerase
  • olapa Niraparib and Niraparib have clear curative effect on ovarian cancer patients with BRCA mutation, but they have not obtained significant benefit for most platinum-refractory/resistant recurrent ovarian cancer patients without BRCA mutation.
  • Ovarian cancer requires the search for more effective treatment options.
  • Cioroni is a small molecule anti-tumor drug targeting multiple protein kinases. It has high selective inhibitory activity on Aurora B, VEGFR/PDGFR/c-Kit, and CSF-1R targets, and has the ability to inhibit tumor cells. Multi-channel synergistic mechanisms such as mitosis, anti-tumor angiogenesis, and regulation of tumor immune microenvironment play a comprehensive anti-tumor effect. Due to the unique anti-tumor mechanism of cioronib, the research on the marker genes related to the efficacy of cioroni will help to find the potential benefit patient population and better therapeutic effect for the clinical application of cioroni .
  • the purpose of the present invention is to provide the use of RBM10 gene or its mRNA or its encoded protein or protein fragment as a biomarker in evaluating the efficacy of cioronitrile or instructing the administration of cioronib or predicting the therapeutic effect of ovarian cancer .
  • Cioroni capsules are used to conduct Phase Ib clinical trials for relapsed and refractory advanced ovarian cancer.
  • ctDNA plasma cell-free tumor DNA
  • 548 tumor-related genes are used to conduct accompanying research on therapeutic efficacy-related biomarkers.
  • Blood samples were drawn from all patients before enrollment, and gene sequences were detected for tumor-related genes, including gene mutations and copy number abnormalities.
  • the detection results selected all genes with a mutation rate of more than 0.4%, and used the patient's progression-free survival (PFS) as the efficacy index to analyze the correlation between tumor-related gene abnormalities and the efficacy of cioronib.
  • PFS progression-free survival
  • the present invention accordingly relates to the preparation of a reagent for detecting the RBM10 gene or its mRNA or its encoded protein or protein fragment for evaluating the curative effect of cioronib or guiding the administration of cioronib or predicting the treatment of ovarian cancer Use in kits or microarrays for effect.
  • a reagent for detecting the RBM10 gene or its mRNA or its encoded protein or protein fragment for evaluating the curative effect of cioronib or guiding the administration of cioronib or predicting the treatment of ovarian cancer Use in kits or microarrays for effect.
  • a reagent for detecting the RBM10 gene or its mRNA or its encoded protein or protein fragment for evaluating the curative effect of cioronib or guiding the administration of cioronib or predicting the treatment of ovarian cancer Use in kits or microarrays for effect.
  • cioronib if there is variation in the RBM10 gene or its
  • the reagent for detecting the RBM10 gene or its mRNA or its encoded protein or protein fragment is a binding agent that binds to the RBM10 gene encoded protein or protein fragment, or hybridizes with the RBM10 gene or its mRNA Or a substance that amplifies the RBM10 gene or its mRNA.
  • the binding agent that binds to the protein or protein fragment encoded by the RBM10 gene is an anti-RBM10 antibody.
  • the substance that hybridizes to or amplifies the RBM10 gene or its mRNA is an oligonucleotide primer or probe.
  • the present invention relates to a kit or microarray for evaluating the curative effect of cioronib or instructing the administration of cioronib or predicting the therapeutic effect of ovarian cancer, comprising a kit or microarray for detecting the RBM10 gene or its mRNA or its Reagents for encoded proteins or protein fragments.
  • a kit or microarray for detecting the RBM10 gene or its mRNA or its Reagents for encoded proteins or protein fragments.
  • if there is variation in the RBM10 gene or its mRNA or the protein encoded by the subject compared with the wild type it indicates that for the subject, cioronib has better curative effect and can The use of cioroni therapy or ovarian cancer treatment is better.
  • the reagent for detecting the RBM10 gene or its mRNA or its encoded protein or protein fragment is a binding agent that binds to the RBM10 gene encoded protein or protein fragment, or hybridizes with the RBM10 gene or its mRNA Or a substance that amplifies the RBM10 gene or its mRNA.
  • the binding agent that binds to the protein or protein fragment encoded by the RBM10 gene is an anti-RBM10 antibody
  • the substance that hybridizes with the RBM10 gene or its mRNA or amplifies the RBM10 gene or its mRNA is an oligonucleotide Nucleotide primers or probes.
  • the present invention relates to the use of cioronib in the preparation of a medicament for the treatment of ovarian cancer, especially ovarian cancer with RBM10 gene mutation.
  • the present invention relates to a biomarker for evaluating the efficacy of cioronib or guiding the administration of cioronib or predicting the treatment effect of ovarian cancer
  • the biomarker is the RBM10 gene or its mRNA or its encoding protein or protein fragment.
  • the present invention relates to the use of the RBM10 gene or its mRNA or its encoded protein or protein fragment as a biomarker in evaluating the efficacy of cioronib or guiding the administration of cioronib or predicting the therapeutic effect of ovarian cancer.
  • the present invention relates to the use of the RBM10 gene or its mRNA or its encoded protein or protein fragment in the preparation of biomarkers for evaluating the efficacy of cioronib or guiding the administration of cioronib or predicting the therapeutic effect of ovarian cancer.
  • the present invention relates to a method for evaluating the curative effect of cioronitrile or instructing the administration of cioronib or predicting the therapeutic effect of ovarian cancer, comprising the following steps:
  • the curative effect of cioronib is better, and the cioronib can be used for treatment or ovarian cancer. Cancer treatment is better.
  • the biological sample is ctDNA, tumor tissue, circulating tumor cells, or tissue from other sources of the human body.
  • the detection method is gene sequencing, PCR, FISH, immunohistochemistry, ELISA, Western or flow cytometry.
  • the ovarian cancer tumor tissue ctDNA is used as the detection sample, and the next-generation sequencing technology is used for detection.
  • the present invention relates to the use of cioronib in the treatment of ovarian cancer, especially ovarian cancer with mutations in the RBM10 gene.
  • Figure 1 shows the progression-free survival of RBM10 mutant and wild-type.
  • the present invention relates to the relationship between the mutation and/or expression of a newly discovered biomarker (ie, RBM10) and the efficacy of cioronib, the administration of cioronib, and/or the therapeutic effect of ovarian cancer.
  • the biomarkers described herein provide methods for evaluating the efficacy of cioronib, guiding the administration of cioronib, and/or predicting the effect of treatment in ovarian cancer.
  • one embodiment of the present invention represents an improvement in biomarkers suitable for evaluating the efficacy of cioronib, guiding the administration of cioronib and/or predicting the effect of ovarian cancer treatment.
  • the newly discovered biomarkers of the present invention may be combined with one or more other cancer markers known in the art (eg, CEA, NSE, CA 19-9, CA 125, CA 72-4, PSA, proGRP, SCC, NNMT, VEGFR2, HER2, MISIIR, VEGFA, CD24) in combination, for example, for evaluating the efficacy of cioronib, guiding the administration of cioronib and/or predicting the treatment effect of ovarian cancer or For the preparation of kits for this purpose.
  • cancer markers known in the art eg, CEA, NSE, CA 19-9, CA 125, CA 72-4, PSA, proGRP, SCC, NNMT, VEGFR2, HER2, MISIIR, VEGFA, CD24
  • the invention verifies the correlation between the RBM10 gene variation and the curative effect of cioroni by taking the patient's progression-free survival (PFS) as the curative effect index, and the detection of the RBM10 gene mutation information can guide the clinical medication and treatment of cioroni.
  • PFS progression-free survival
  • the curative effect of cioronib is better, the treatment effect of cioronib can be better, or the effect of ovarian cancer treatment is better
  • the kit of the present invention is used to evaluate the efficacy of cioronib in the treatment of ovarian cancer and/or to guide the administration of cioronib for the treatment of ovarian cancer.
  • sample means a material known or suspected to express or contain a biomarker (i.e. RBM10) or a binding agent, such as an antibody specific for the biomarker (i.e. RBM10).
  • Samples may be derived from biological sources ("biological samples"), such as tissues (eg, biopsy samples), extracts or cell cultures including cells (eg, tumor cells), cell lysates, and biological or physiological fluids, such as Whole blood, plasma, serum, saliva, cerebrospinal fluid, sweat, urine, milk, peritoneal fluid, etc. Samples obtained from sources or after pretreatment to improve sample characteristics (eg, preparation of plasma from blood, dilution of mucus, etc.) can be used directly.
  • biological samples such as tissues (eg, biopsy samples), extracts or cell cultures including cells (eg, tumor cells), cell lysates, and biological or physiological fluids, such as Whole blood, plasma, serum, saliva, cerebrospinal fluid, sweat, urine, milk, peritoneal fluid, etc.
  • the sample is a human physiological fluid, such as human serum.
  • the sample is a biopsy sample such as tumor tissue or cells obtained by tissue examination.
  • the sample is a malignant or normal tissue sample such as a paracancerous normal tissue sample.
  • Samples that can be analyzed in accordance with the present invention include polynucleotides of clinical origin.
  • target polynucleotides can include RNA, including but not limited to total cellular RNA, poly(A)+ messenger RNA (mRNA) or a portion thereof, cytoplasmic mRNA, or RNA transcribed from cDNA (i.e. cRNA).
  • the target polynucleotide can be detectably labeled on one or more nucleotides using methods known in the art.
  • Detectable labels can be, without limitation, luminescent, fluorescent, bioluminescent, chemiluminescent, radioactive, and colorimetric labels.
  • marker refers to a molecule to be used as a target for analyzing a patient's experimental sample.
  • molecular targets are genes, proteins or polypeptides.
  • the genes, proteins or polypeptides used as markers in the present invention are intended to include naturally occurring variants of said genes or proteins as well as fragments of said genes or proteins or said variants, particularly immunologically detectable fragments .
  • Immunologically detectable fragments preferably comprise at least 6, 7, 8, 10, 12, 15 or 20 contiguous amino acids of the marker polypeptide.
  • proteins released by cells or present in the extracellular matrix may be damaged (eg, during inflammation) and may be degraded or cleaved into such fragments.
  • markers are synthesized in an inactive form, which can then be activated by proteolysis.
  • proteins or fragments thereof can also be present as part of a complex.
  • Such complexes can also be used as markers in the sense of the present invention.
  • the amino acid sequence of the variant is 95%, 96%, 97%, 98%, 99% or more identical to the corresponding marker sequence.
  • marker polypeptides or variants thereof may carry post-translational modifications.
  • Non-limiting examples of post-translational modifications are glycosylation, acylation and/or phosphorylation.
  • Expression of the marker can also be identified by detecting translation of the marker (ie, detection of the marker protein in the sample).
  • Methods suitable for detecting marker proteins include any suitable method for detecting and/or measuring proteins from cells or cell extracts. Such methods include, but are not limited to, immunoblotting (eg, Western blotting), enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), immunoprecipitation, immunohistochemistry, and immunofluorescence.
  • Particularly preferred methods for detecting proteins include any cell-based assay, including immunohistochemistry and immunofluorescence assays. Such methods are well known in the art.
  • subject refers to a warm-blooded animal, such as a mammal.
  • the term includes, but is not limited to, livestock, rodents (eg, rats and mice), primates, and humans. Preferably the term refers to humans.
  • RBM10 (NG_012548.1) belongs to a family of RNA motif-containing binding proteins, located on chromosome Xp11.23, encoding a 930 amino acid nuclear protein, and contains two RNA recognition domains (RRM), two zinc finger domains and A G-patch domain. This gene is also known as DXS8237E, GPATC9, GPATCH9, S1-1, TARPS and ZRANB5.
  • wild-type is to be understood according to the general understanding of those skilled in the art and refers to a nucleic acid or amino acid sequence in its naturally occurring form without any artificial mutation, nucleotide change or amino acid modification.
  • mutant should be understood according to the general understanding of those skilled in the art.
  • a nucleic acid sequence is referred to as “mutated” if it contains at least one nucleotide addition, deletion or substitution in its nucleic acid sequence compared to its native or native nucleic acid sequence, ie if it contains a nucleic acid mutation.
  • An amino acid sequence is referred to as “mutated” if it contains at least one added, deleted or substituted amino acid in its amino acid sequence compared to its native or native amino acid sequence, ie if it contains an amino acid mutation.
  • the variation of the RBM10 gene or its mRNA refers to the mutation in the wild-type RBM10 gene or its mRNA, which includes addition, deletion and substitution of one or more nucleotides and changes in copy number (including amplification and missing).
  • the nucleotides may be nucleotides of a coding region or a non-coding region.
  • the coding region may be a nucleotide sequence encoding an RNA recognition domain (RRM), a zinc finger domain and/or a G-patch domain.
  • RRM RNA recognition domain
  • the variation of the RBM10 gene or its mRNA can lead to the increase or decrease of the expression level of the RBM10 gene and/or the amplification or deletion of the copy number.
  • the level or copy number is at least about 1.1, 1.25, 1.5, 2, 3, 4, 5, 6, 7, 8, 9 or 10 times or more or at most about 1 times the control or standard, respectively /1.1, 1/1.25, 1/1.5, 1/2, 1/3, 1/4, 1/5, 1/6, 1/7, 1/8, 1/9 or 1/10 or less.
  • Copy number amplifications or deletions can be detected by techniques well known in the art, such as whole gene sequencing.
  • the variation of the protein or protein fragment encoded by the RBM10 gene includes: 1) mutations in the protein or protein fragment encoded by the wild-type RBM10 gene, which include addition, deletion or substitution of one or more amino acids; and 2 ) Changes in the expression level of the protein encoded by the wild-type RBM10 gene.
  • the amino acid may be an amino acid of an RNA recognition domain (RRM), a zinc finger domain and/or a G-patch domain.
  • the protein fragments refer to polypeptides having amino-terminal deletions, carboxy-terminal deletions and/or intermediate deletions compared to the full-length native protein.
  • the fragments may also contain modified amino acids compared to the native protein. In certain embodiments, fragments are about 5-500 amino acids in length.
  • fragments can be at least 5, 6, 8, 10, 14, 20, 50, 70, 100, 110, 150, 200, 250, 300, 350, 400, or 450 amino acids in length.
  • the fragment is an immunologically detectable fragment preferably comprising at least 6, 7, 8, 10, 12, 15 or 20 contiguous amino acids of the marker polypeptide.
  • the change in the protein expression level refers to at least about 1.1, 1.25, 1.5, 2, 3, 4, 5, 6, 7, 8, 5, 5, 6, 7, 8, 9 or 10 times or more or at most about 1/1.1, 1/1.25, 1/1.5, 1/2, 1/3, 1/4, 1/5, 1/6 of the expression level of a control or standard , 1/7, 1/8, 1/9 or 1/10 or less.
  • polypeptide and “protein” are used interchangeably herein to refer to at least one molecular chain of amino acids linked by covalent and/or non-covalent bonds.
  • the term includes post-translational modifications of peptides, oligopeptides, and proteins and polypeptides, such as glycosylation, acetylation, phosphorylation, and the like. Protein fragments, analogs, muteins or variant proteins, fusion proteins, etc. are also included within the meaning of the term.
  • determining "protein expression level", “gene expression” or “gene expression level” as used herein includes, but is not limited to, determining corresponding RNA, protein or peptide levels (or combinations thereof).
  • the present invention is not limited to specific methods and reagents for measuring protein, peptide or RNA levels, all of which are well known in the art.
  • Methods for determining the amount or concentration of protein in a sample are known to the skilled artisan. Such methods include radioimmunoassays, competitive binding assays, Western blot analysis and ELISA assays.
  • methods using antibodies both monoclonal and polyclonal antibodies are suitable.
  • the antibody may be immunologically specific for a protein, protein epitope or protein fragment.
  • oligonucleotide refers to a polymeric form of nucleotides of any length, either ribonucleotides or deoxyribonucleotides.
  • the term includes double- and single-stranded DNA and RNA, modified and unmodified forms such as methylation or capping of polynucleotides.
  • polynucleotide and oligonucleotide are used interchangeably herein.
  • the oligonucleotide may, but need not, include other coding or non-coding sequences, or it may, but need not, be linked to other molecules and/or vectors or support materials.
  • oligonucleotides used in the methods or kits of the invention can be of any length suitable for the particular method.
  • the term refers to an antisense nucleic acid molecule (eg, an mRNA or DNA strand in the opposite orientation to a sense polynucleotide encoding a cancer marker of the invention (eg, RBM10)).
  • Oligonucleotides for use in the present invention include complementary nucleic acid sequences and nucleic acids substantially identical to those sequences, and also include sequences that differ from nucleic acid sequences due to the degeneracy of the genetic code. Oligonucleotides useful in the present invention also include nucleic acids that hybridize to oligonucleotide cancer marker nucleic acid sequences under stringent conditions, preferably high stringency conditions.
  • Nucleotide hybridization assays are well known in the art. Hybridization assay procedures and conditions will vary depending on the application and are chosen according to known general binding methods, see eg, J. Sambrook et al., Molecular Cloning: A Laboratory Guide (Third Edition. Science Press, 2002); and Young and Davis , PNAS, 80:1194 (1983). Methods and apparatus for performing repetitive and controlled hybridization reactions have been described in US Pat. Nos. 5,871,928, 5,874,219, 6,045,996, 6,386,749, and 6,391,623, each of which is incorporated herein by reference.
  • Genomic samples can be amplified by various mechanisms, some of which employ PCR. Samples can be amplified on the array. See, eg, US Patent No. 6,300,070 and US Patent Application Serial No. 09/513,300.
  • LCR ligase chain reaction
  • LCR ligase chain reaction
  • Genomics 4 eg Wu and Wallace, Genomics 4, 560 (1989), Landegren et al. Science 241, 1077 (1988) and Barringer et al. Gene 89:117 (1990)
  • transcription Amplification Kwoh et al., Proc. Natl. Acad. Sci. USA 86, 1173 (1989) and WO 88/10315
  • self-sustaining sequence replication (Guatelli et al., Proc. Nat. Acad. Sci. USA, 87, 1874 (1990) ) and WO90/06995)
  • selective amplification of target polynucleotide sequences US Pat. No.
  • CP-PCR consensus-primed polymerase chain reaction
  • AP-PCR arbitrarily primed polymerase chain reaction reaction
  • NABSA Nucleic Acid-Based Sequence Amplification
  • Reagents that can be used to detect RBM10 expression levels and/or copy number are well known in the art. Such reagents suitable for use in the present invention are commercially available or routinely prepared by methods well known to those skilled in the art.
  • binding agent refers to substances such as polypeptides, antibodies, ribosomes or aptamers that specifically bind to the biomarkers of the invention (RBM10).
  • a substance “specifically binds" to a biomarker of the invention if it reacts at a detectable level with the biomarker of the invention but not detectably reacts with a peptide containing an unrelated sequence or a sequence of a different polypeptide. Binding properties can be assessed using an ELISA that can be readily performed by those skilled in the art.
  • the binding agent may be a ribosome, RNA or DNA molecule or polypeptide with or without a peptide component.
  • the binding agent may be a polypeptide comprising a polypeptide biomarker sequence, a peptide variant thereof, or a non-peptide mimetic of such a sequence.
  • Aptamers include DNA or RNA molecules that bind nucleic acids and proteins. Aptamers that bind the markers of the present invention can be generated using conventional techniques without undue experimentation. [See, for example, the following publications describing in vitro selection of aptamers: Klug et al., Mol. Biol. Reports 20:97-107 (1994); Wallis et al., Chem. Biol. 2:543-552 (1995); Ellington, Curr. Biol. 4:427-429 (1994); Lato et al., Chem. Biol. 2:291-303 (1995); Conrad et al., Mol. Div. 1:69-78 (1995); and Uphoff et al., Curr. Opin . Struct. Biol. 6:281-287 (1996)].
  • Antibodies useful in the present invention include, but are not limited to, synthetic antibodies, monoclonal antibodies, polyclonal antibodies, recombinant antibodies, antibody fragments (eg, Fab, Fab', F(ab')2), dAbs (domain antibodies; see Ward et al.
  • antibody heavy chain intrabody, humanized antibody, human antibody, antibody light chain, single-chain Fv (scFv) (eg, including monospecific, bispecific, etc.) , anti-idiotypic (ant-Id) antibodies, proteins comprising antibody moieties, chimeric antibodies (eg, antibodies containing the binding specificity of a murine antibody but where the remainder is of human origin), derivatives such as enzyme conjugates or labeled Derivatives, diabodies, linear antibodies, disulfide-linked Fvs (sdFv), multispecific antibodies (eg, bispecific antibodies), epitope-binding fragments of any of the foregoing, and antigen recognition comprising the desired specificity Any other modified configuration of the immunoglobulin molecule at the site.
  • scFv single-chain Fv
  • anti-Id anti-idiotypic antibodies
  • proteins comprising antibody moieties
  • chimeric antibodies eg, antibodies containing the binding specificity of a murine antibody but where the remainder is of human origin
  • derivatives
  • Antibodies include antibodies of any class (e.g., IgA, IgD, IgE, IgG, IgM, and IgY), any class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2), or any subclass (e.g., IgG2a and IgG2b), and antibodies do not have to be is any particular type, class or subclass.
  • the antibody is an IgG antibody or a class or subclass thereof.
  • Antibodies can be from any animal source, including birds and mammals (eg, human, murine, donkey, sheep, rabbit, goat, guinea pig, camel, horse, or chicken).
  • antibodies for use in the present invention are commercially available from, for example, Invitrogen (Cat. Nos. PA5-83253, PA5-40331, PA5-40330, etc.), Abcam (eg, antibody under Cat. No. ab224149), and the like.
  • the antibodies can be prepared by recombinant methods well known in the art.
  • the antibody is a monoclonal antibody.
  • monoclonal antibodies see, eg, Kohler et al. (1975) Nature 256:495-497; Kozbor et al. (1985) J. Immunol Methods 81:31-42; Cote et al. (1983) Proc Natl Acad Sci 80:2026-2030 and Cole et al. (1984) Mol Cell Biol 62:109-120.
  • kits can be prepared by conventional methods in the art.
  • the kits may contain materials or reagents (including reagents for detecting the RBM10 gene or its mRNA or its encoded protein or protein fragment) for use in carrying out the methods of the present invention.
  • Kits may include storage reaction reagents (eg, primers, dNTPs, enzymes, etc. in suitable containers) and/or support materials (eg, buffers, instructions for performing assays, etc.).
  • a kit may include one or more containers (eg, cassettes) containing the respective reaction reagents and/or support materials. Such contents may be delivered to the intended recipient together or separately.
  • the kit may contain reagents, buffers, and instructions for use in detecting the RBM10 gene or its mRNA or its encoded protein or protein fragment.
  • the kit may also contain polymerase and dTNP, among others.
  • the kit may also contain internal standards, positive and negative controls, etc. for quality control.
  • the kit may also contain reagents for preparing nucleic acid, eg, DNA, from the sample.
  • nucleic acid eg, DNA
  • a microarray refers to a solid support with a flat surface that has an array of nucleic acids, each member of the array comprising identical copies of oligonucleotides or polynucleotides immobilized on spatially defined regions or sites, so The regions or sites do not overlap with regions or sites of other members of the array; that is, the regions or sites are spatially discrete.
  • a spatially defined hybridization site may be "addressable" in that its location and the identity of its immobilized oligonucleotide are known or predetermined (eg, known or predetermined prior to its use). definite).
  • the oligonucleotide or polynucleotide is single stranded and is usually covalently attached to the solid support from the 5'- or 3'-end.
  • the density of nucleic acids containing non-overlapping regions in the microarray is typically greater than 100/cm 2 , more preferably greater than 1000/cm 2 .
  • Microarray technology is disclosed, for example, in the following references: Microarrays: A Practical Approach, edited by Schena (IRL Press, Oxford, 2000); Southern, Current Opin. Chem. Biol., 2:404-410, 1998, the entire contents of which are via Reference is incorporated herein.
  • the invention discloses the use of the RBM10 gene, and those skilled in the art can learn from the content of this article and appropriately improve the process parameters to achieve. It should be particularly pointed out that all similar substitutions and modifications are obvious to those skilled in the art, and they are deemed to be included in the present invention.
  • the uses described in the present invention have been described through the preferred embodiments, and it is obvious that relevant persons can make changes or appropriate changes and combinations of the uses described herein without departing from the content, spirit and scope of the present invention, so as to realize and apply the technology of the present invention .
  • Example 1 Phase Ib clinical trial of cioronib monotherapy in the treatment of relapsed and refractory ovarian cancer
  • Test drug Cioroni capsules, specifications: 5mg, 25mg. Produced by Shenzhen Microchip Biotechnology Co., Ltd.
  • Cioroni capsules were administered at 50 mg/day, QD (no adjustment for body weight or body surface area). Take each morning on an empty stomach, with water, and swallow the capsule whole. Continuous administration for 28 days is one treatment cycle, and there is no interval between each treatment cycle.
  • epithelial ovarian cancer Histologically diagnosed epithelial ovarian cancer, fallopian tube cancer or primary peritoneal cancer;
  • Platinum-resistant patients should have received at least 2 different chemotherapy regimens for disease progression or recurrence;
  • platinum-sensitive patients should have received at least 2 chemotherapy regimens for disease progression or recurrence, or the subject refuses to receive further chemotherapy;
  • the interval between the previous chemotherapy, radiotherapy, targeted therapy, immunotherapy or study drug treatment should be more than 4 weeks. If the chemotherapy regimen includes mitomycin, the interval should be more than 6 weeks;
  • Coagulation function prothrombin time-international normalized ratio (PT-INR) ⁇ 1.5 times ULN.
  • test subjects took cioroni capsules 50 mg orally once a day, every 28 days as a treatment cycle, and there was no discontinuation interval during the treatment cycle. Throughout the trial, all subjects continued treatment until any of the following occurred (whichever occurred first): disease progression, intolerable toxicity, death, withdrawal of informed consent, or loss to follow-up.
  • Efficacy evaluation According to RECIST1.1 criteria, the evaluation was performed at the baseline and the 4th week after treatment, and repeated every 8 weeks until disease progression. Tumor imaging examinations include CT or MRI of the neck, chest, whole abdomen, and pelvis. Examinations of other parts are performed according to clinical indications and when necessary. The same techniques and methods should be used for baseline and follow-up assessment of lesions.
  • Safety assessment including physical examination, vital signs, ECOG performance score, blood routine, urine routine, 12-lead ECG, blood biochemistry, electrolytes, coagulation function, cardiac enzymes, troponin, TSH, FT3, FT4, amylase, Echocardiography, 24-hour urine protein quantification (if necessary), adverse events.
  • Test results include genetic mutations and copy number abnormalities.
  • a concomitant study of efficacy-related biomarkers was performed on 548 tumor-related genes for the detection and analysis of cell-free tumor DNA (ctDNA) in the plasma of the evaluated patients.
  • the detection results selected all genes with a mutation rate of more than 0.4%, and used the patient's progression-free survival (PFS) as the efficacy index to analyze the correlation between tumor-related gene abnormalities and the efficacy of cioronib.
  • PFS progression-free survival
  • the median PFS was 232 days in 7 of the 23 evaluable subjects with RBM10 mutations (30.4%, these mutations included nucleotide additions, deletions and substitutions, and copy number changes) and 16 with RBM10 wild-type (69.6%).
  • the median PFS of ) was 106 days, and there was a significant difference between the two groups, suggesting that the RBM10 gene is associated with the efficacy of cioronib in the treatment of ovarian cancer and can be a potential biomarker for evaluating the efficacy. See Table 1 and Figure 1 for the results.
  • Table 1 RBM10 gene mutation and median PFS correlation analysis

Abstract

L'invention concerne un biomarqueur pour évaluer l'efficacité de Chiauranib ou guider l'administration correspondante ou prédire l'effet thérapeutique sur le cancer de l'ovaire. Le biomarqueur est le gène RBM10, un ARNm correspondant ou une protéine ou un fragment de protéine codé par celui-ci. La corrélation entre une variation du gène RBM10 et l'efficacité de Chiauranib est vérifiée au moyen de la survie sans progression (SSP) d'un patient en tant qu'indice d'efficacité. La détection d'informations de mutation du gène RBM10 peut guider l'administration clinique de Chiauranib et évaluer son efficacité dans le traitement de tumeurs, en particulier pour le cancer de l'ovaire récurrent et avancé.
PCT/CN2021/108791 2020-07-29 2021-07-28 Utilisation d'un gène rbm10 WO2022022541A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010744602 2020-07-29
CN202010744602.3 2020-07-29

Publications (1)

Publication Number Publication Date
WO2022022541A1 true WO2022022541A1 (fr) 2022-02-03

Family

ID=80037600

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/108791 WO2022022541A1 (fr) 2020-07-29 2021-07-28 Utilisation d'un gène rbm10

Country Status (3)

Country Link
CN (1) CN114058700A (fr)
TW (1) TW202204639A (fr)
WO (1) WO2022022541A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114672562A (zh) * 2022-03-01 2022-06-28 武汉凯德维斯医学检验实验室有限公司 一种针对parp抑制剂耐药性监测的方法、装置、设备及介质

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109893654A (zh) * 2017-12-11 2019-06-18 江苏恒瑞医药股份有限公司 Vegfr抑制剂治疗肿瘤的方法
CN110833544A (zh) * 2018-08-17 2020-02-25 深圳微芯生物科技股份有限公司 组蛋白去乙酰化酶抑制剂与蛋白激酶抑制剂之组合及其制药用途
CN111093706A (zh) * 2017-12-06 2020-05-01 江苏恒瑞医药股份有限公司 Parp抑制剂用于治疗化疗耐药的卵巢癌或乳腺癌的用途

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111093706A (zh) * 2017-12-06 2020-05-01 江苏恒瑞医药股份有限公司 Parp抑制剂用于治疗化疗耐药的卵巢癌或乳腺癌的用途
CN109893654A (zh) * 2017-12-11 2019-06-18 江苏恒瑞医药股份有限公司 Vegfr抑制剂治疗肿瘤的方法
CN110833544A (zh) * 2018-08-17 2020-02-25 深圳微芯生物科技股份有限公司 组蛋白去乙酰化酶抑制剂与蛋白激酶抑制剂之组合及其制药用途

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
ANONYMOUS: "Phase Ib Study of Chiauranib in Patients With Ovarian Cancer", CHIPSCREEN BIOSCIENCES LTD., 18 January 2020 (2020-01-18), XP055853624, Retrieved from the Internet <URL:https://clinicaltrials.gov/ct2/show/NCT03166891> [retrieved on 20211021] *
JIN XIN: "Molecular Mechanism of RBM10 Inhibiting Proliferation of Non-small Cell Lung Cancer by Regulating RAP1/AKT/CREB Signal Transduction Pathway", CHINESE SELECTED DOCTORAL DISSERTATIONS AND MASTER'S THESES FULL-TEXT DATABASES, MEDICINE & PUBLIC HEALTH, 15 November 2019 (2019-11-15), XP055890521 *
SUN YONGKUN, YANG LIN, HAO XUEZHI, LIU YUTAO, ZHANG JINWEN, NING ZHIQIANG, SHI YUANKAI: "Phase I dose-escalation study of chiauranib, a novel angiogenic, mitotic, and chronic inflammation inhibitor, in patients with advanced solid tumors", JOURNAL OF HEMATOLOGY & ONCOLOGY, vol. 12, no. 1, 1 December 2019 (2019-12-01), XP055853626, DOI: 10.1186/s13045-018-0695-0 *
SUTHERLAND LESLIE C., THIBAULT PHILIPPE, DURAND MATHIEU, LAPOINTE ELVY, KNEE JOSE M., BEAUVAIS ARIANE, KALATSKAYA IRINA, HUNT SARA: "Splicing arrays reveal novel RBM10 targets, including SMN2 pre-mRNA", BMC MOLECULAR BIOLOGY, vol. 18, no. 1, 1 December 2017 (2017-12-01), pages 19, XP055890353, DOI: 10.1186/s12867-017-0096-x *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114672562A (zh) * 2022-03-01 2022-06-28 武汉凯德维斯医学检验实验室有限公司 一种针对parp抑制剂耐药性监测的方法、装置、设备及介质

Also Published As

Publication number Publication date
TW202204639A (zh) 2022-02-01
CN114058700A (zh) 2022-02-18

Similar Documents

Publication Publication Date Title
JP7401710B2 (ja) 正規化バイオマーカースコアからがん治療を特定するためのシステム及び方法
Wan et al. METTL3/IGF2BP3 axis inhibits tumor immune surveillance by upregulating N6-methyladenosine modification of PD-L1 mRNA in breast cancer
ES2370054T3 (es) Biomarcadores y procedimientos para determinar la sensibilidad a moduladores del receptor del factor de crecimiento epidérmico.
US20090233286A1 (en) Methods of diagnosis and prognosis of pancreatic cancer
JP2005503779A (ja) 致死性の高い癌の分子シグネチャー
KR20110036610A (ko) 신규 온코진 nrf2
US20140030257A1 (en) Agtr1 as a marker for bevacizumab combination therapies
US10190170B2 (en) Maker for diagnosing HER2 inhibitor resistant cancer, diagnostic kit comprising same, and method for diagnosing HER2 inhibitor resistant cancer
WO2022022541A1 (fr) Utilisation d&#39;un gène rbm10
CN112626207B (zh) 一种用于区分非侵袭性和侵袭性无功能垂体腺瘤的基因组合
US20210108276A1 (en) Tumor-associated markers for detection of minimal residual disease using digital droplet pcr
KR20200092382A (ko) 유전자 발현 검정을 이용한 펩티드 수용체 방사선요법의 예측
US20110151469A1 (en) Interferon epsilon (ifne1) as a marker for targeted cancer therapy
JP5145549B2 (ja) 腫瘍マーカー
US9109258B2 (en) Molecular markers for the diagnosis and treatment of tumors
KR102259708B1 (ko) 대장암에 대한 항암제 감수성 예측을 위한 신규 바이오마커
US20170044627A1 (en) Six-Gene Biomarker of Survival and Response to Platinum Based Chemotherapy in Serious Ovarian Cancer Patients
KR102259695B1 (ko) 대장암에 대한 항암제 감수성 예측을 위한 신규 바이오마커
KR102325972B1 (ko) 간암 치료 반응 예측용 spp1 바이오마커 및 이의 용도
KR102326119B1 (ko) 암의 면역 치료 후 예후 예측용 바이오 마커
US9075067B2 (en) Marker for the diagnosis of cancer
CN114763573A (zh) Fgf19扩增作为预测fgfr抑制剂在食管鳞癌中敏感性的生物标志物及其潜在应用
CN114763574A (zh) Cdkn2a/2b缺失作为预测cdk4/6抑制剂在食管鳞癌中敏感性的生物标志物
US20160340736A1 (en) Method for Detecting Lung Cancer
JP2017032588A (ja) 結核検査用バイオマーカー

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: 21851270

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 26.06.2023)

122 Ep: pct application non-entry in european phase

Ref document number: 21851270

Country of ref document: EP

Kind code of ref document: A1