WO2009036332A1 - Micrornas differentially expressed in cervical cancer and uses thereof - Google Patents

Micrornas differentially expressed in cervical cancer and uses thereof Download PDF

Info

Publication number
WO2009036332A1
WO2009036332A1 PCT/US2008/076246 US2008076246W WO2009036332A1 WO 2009036332 A1 WO2009036332 A1 WO 2009036332A1 US 2008076246 W US2008076246 W US 2008076246W WO 2009036332 A1 WO2009036332 A1 WO 2009036332A1
Authority
WO
WIPO (PCT)
Prior art keywords
hsa
mir
asg
stl
mirna
Prior art date
Application number
PCT/US2008/076246
Other languages
French (fr)
Inventor
Sylvie Beaudenon
Emmanuel Labourier
Laura Elizondo
Original Assignee
Asuragen, Inc.
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 Asuragen, Inc. filed Critical Asuragen, Inc.
Priority to EP08831073A priority Critical patent/EP2198050A1/en
Publication of WO2009036332A1 publication Critical patent/WO2009036332A1/en

Links

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
    • 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
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/178Oligonucleotides characterized by their use miRNA, siRNA or ncRNA

Definitions

  • the present invention relates generally to the field of molecular biology. More particularly, it concerns methods and compositions involving microRNA (miRNAs) molecules. Certain aspects of the invention include applications for miRNAs in diagnostics, therapeutics, and prognostics of cervical cancer.
  • miRNAs microRNA
  • miRNAs small RNAs
  • C. elegans, Drosophila, and humans Lagos-Quintana et al, 2001; Lau et al, 2001; Lee and Ambros, 2001.
  • miRNAs Several hundred miRNAs have been identified in plants and animals — including humans — which do not appear to have endogenous siRNAs. Thus, while similar to siRNAs, miRNAs are distinct.
  • miRNAs thus far observed have been approximately 21-22 nucleotides in length, and they arise from longer precursors, which are transcribed from non-protein-encoding genes. See review of Ca ⁇ ngton et al (2003). The precursors form structures that fold back on themselves in self-complementary regions; they are then processed by the nuclease Dicer (in animals) or DCLl (in plants) to generate the short double-stranded miRNA.
  • One of the miRNA strands is incorporated into a complex of proteins and miRNA called the RNA- induced silencing complex (RISC).
  • RISC RNA- induced silencing complex
  • the miRNA guides the RISC complex to a target mRNA, which is then cleaved or translationally silenced, depending on the degree of sequence complementarity of the miRNA to its target mRNA.
  • a target mRNA which is then cleaved or translationally silenced, depending on the degree of sequence complementarity of the miRNA to its target mRNA.
  • perfect or nearly perfect complementarity leads to mRNA degradation, as is most commonly observed in plants.
  • imperfect base pairing as is primarily found in animals, leads to translational silencing.
  • recent data suggest additional complexity (Bagga et al, 2005; Lim et al, 2005), and mechanisms of gene silencing by miRNAs remain under intense study.
  • miRNAs have also been implicated in regulating cell growth and cell and tissue differentiation - cellular processes that are associated with the development of cancer.
  • Cervical cancer is the second most common cause of cancer in women worldwide (Pisani et al, 2002; Parkin et al, 2005). About 470,000 new cases are diagnosed and approximately 230,000 women die of cervical cancer every year (Pisani et al, 1999). While the majority (-80%) of these new cases and deaths occur in developing countries, it is estimated that approximately 3,700 women will die from invasive cervical cancer in the United States in 2007 (Jemal et al, 2007).
  • HPVs human papillomaviruses
  • HPVs human papillomaviruses
  • HPV types are the etiological agents of the vast majority (99.7%) of cervical cancers and their intraepithelial precursors (Pisani, et al, 2002; Parkin et al, 2005; Ober Hausen, 2002).
  • Approximately, fifty HPV types infect the anogenital tract including the uterine cervix (Pisani, et al, 2002; Parkin et al, 2005).
  • "High-risk" HPV types are associated with intraepithelial lesions that can progress into invasive carcinomas.
  • HPV 16 and HPV 18 are associated with 50% and 20%, respectively, of cervical squamous cell carcinomas (Bosch and de Sanjose, 2002; Ober Hausen, 2002; Clifford et al, 2003).
  • Other high-risk HPV types are found in 20-30% of cervical cancers (Bosch and de Sanjose, 2002; Ober Hausen, 2002; Clifford et al, 2003).
  • High-risk HPV types are also associated with 25% of head and neck tumors, in particular tumors of the mouth, tonsils, esophagus and larynx (Gillison et al, 2000; Rose et al, 2006).
  • Pap smears Cytological examination of cervical smears with Papanicolaou staining (Pap smears) is the screening method universally accepted for early detection of cervical cancer and its precursors. Pap smear screening has been very effective in reducing cervical cancer incidence and mortality. Abnormal pap smear results include mild dysplasias referred to as low-grade squamous intraepithelial lesions (LSIL) and moderate to severe dysplasias referred to as high- grade squamous intraepithelial lesions (HSIL). A Pap smear with LSIL or HSIL indicates a need for further examination and possible treatment.
  • LSIL low-grade squamous intraepithelial lesions
  • HSIL high- grade squamous intraepithelial lesions
  • Embodiments of the invention include compositions and methods of identifying miRNAs that are differentially expressed or mis-regulated in various states of normal, precancerous, cancerous, and/or abnormal tissues, including but not limited to normal cervical tissue, pre-cancerous diseased cervical tissue ⁇ e.g., low-grade squamous intraepithelial lesions (LSIL) and high-grade squamous intraepithelial lesions (HSIL)), squamous cell carcinoma, cervical cancer ⁇ e.g., cervical squamous cell carcinoma), squamous cell carcinoma of the vulva and vagina, penile intraepithelial lesions, anal cancer, cancers of the digestive tract, oral cancers, a subset of head and neck cancers, and tonsil cancer.
  • LSIL low-grade squamous intraepithelial lesions
  • HSIL high-grade squamous intraepithelial lesions
  • squamous cell carcinoma cervical cancer ⁇ e.g., cervical
  • the cancers are related to or a result of HPV infection.
  • a subject known or at risk of infection with HPV can be monitored.
  • a subject can be monitored periodically, e.g. rm ' RNA assays can be conducted in conjunction with pap smears or noninvasive procedures or procedures with limited invasiveness ⁇ e.g., swabbing or flushing and the like). The method of sampling is not intended to be a limiting factor and is at the discretion of the care giver.
  • the invention describes a method for diagnosing normal, pre-cancerous, and cancerous tissues or cells, including but not limited to cervical squamous intraepithelial lesions and cervical cancer based on determining levels (increased or decreased) of selected miRNAs in patient-derived samples.
  • Samples may be obtained and/or analyzed from patients, including but not limited to patients having or suspected of having pre-cancerous cervical lesion or cervical cancer, or a patient suspected of having one or the other condition.
  • miRNA is used according to its ordinary and plain meaning and refers to a microRNA molecule found in eukaryotes that is involved in RNA-based gene regulation. See, e.g., Carrington et al, 2003, which is hereby incorporated by reference. Individual miRNAs in a variety of organisms have been identified, sequenced, and given names. Names of miRNAs and their sequences related to the present invention are provided herein.
  • a "synthetic nucleic acid” of the invention means that the nucleic acid does not have a chemical structure or sequence of a naturally occurring nucleic acid or is made by non-natural processes. Consequently, it will be understood that the term “synthetic miRNA” refers to a “synthetic nucleic acid” that functions as or inhibits the functions of an miRNA, at least in part, in a cell or under physiological conditions.
  • nucleic acid molecule(s) need not be "synthetic.”
  • a non-synthetic miRNA employed in methods and compositions of the invention may have all or part of the sequence and structure of a naturally occurring miRNA precursor or the mature miRNA.
  • non-synthetic miRNAs used in methods and compositions of the invention may not have one or more modified nucleotides or nucleotide analogs.
  • the non-synthetic miRNA may or may not be recombinantly produced.
  • the nucleic acid in methods and/or compositions of the invention is specifically a synthetic miRNA; though in other embodiments, the invention specifically involves a non-synthetic miRNA and not a synthetic miRNA. Any embodiments discussed with respect to the use of synthetic miRNAs can be applied with respect to non-synthetic miRNAs, and vice versa.
  • a synthetic miRNA molecule does not have the sequence of a naturally occurring miRNA molecule
  • a synthetic miRNA molecule may have the sequence of a naturally occurring miRNA molecule, but the chemical structure of the molecule, particularly in the part unrelated specifically to the precise sequence (non-sequence chemical structure) differs from chemical structure of the naturally occurring miRNA molecule with that sequence.
  • the synthetic miRNA has both a sequence and non-sequence chemical structure that are not found in a naturally-occurring miRNA.
  • the sequence of the synthetic molecules will identify which miRNA is effectively being provided or inhibited; the endogenous miRNA will be referred to as the "corresponding miRNA.”
  • Corresponding miRNA sequences that can be used in the context of the invention include, but are not limited to, all or a portion of those sequences in SEQ ID NOs: 1-562, as well as any other of miRNA sequence, miRNA precursor sequence, or any complementary sequence.
  • the sequence is or is derived from or contains all or part of a sequence identified in Table 1 below to target a particular miRNA (or set of miRNAs) or mRNA (or set of mRNA).
  • methods include assaying a cell or a sample containing a cell for the presence of one or more miRNA.
  • a sample may comprise RNA or nucleic acid isolated from a tissue or cells of a patient or reference. Consequently, in some embodiments, methods include a step of generating an miRNA profile for a sample.
  • miRNA profile refers to a set of data regarding the expression pattern for a plurality of miRNAs (e.g., one or more miRNA from Table 1) in the sample; it is contemplated that the miRNA profile can be obtained using a set of miRNAs, using for example nucleic acid amplification or hybridization techniques well known to one of ordinary skill in the art. It is contemplate the any one or subset of the miRNA listed in this application can be included or excluded from the claimed invention.
  • an miRNA profile is generated by steps that include: (a) labeling miRNA in the sample; (b) hybridizing miRNA to a number of probes, or amplifying a number of miRNA, and (c) determining miRNA hybridization to the probes or detection of miRNA amplification products, wherein an miRNA profile is generated.
  • Methods of the invention involve diagnosing a patient based on an miRNA expression or expression profile.
  • the presence, absence, elevation, or reduction in the level of expression of a particular miRNA or set of miRNA in a cell is correlated with a disease state compared to the expression level of that miRNA or set of miRNAs in a normal cell. This correlation allows for diagnostic methods to be carried out when the expression level of an miRNA is measured in a biological sample being assessed and then compared to the expression level of a normal cell.
  • miRNA profiles for patients can be generated by evaluating any miRNA or sets of the miRNAs discussed in this application.
  • the miRNA profile that is generated from the patient will be one that provides information regarding the particular disease or condition.
  • the miRNA profile is generated using miRNA hybridization or amplification, (e.g., array hybridization or RT-PCR).
  • a miRNA profile can be used in conjunction with other diagnostic tests, such as serum protein profiles.
  • Embodiments of the invention include methods for diagnosing, prognosing, and/or assessing a condition in a patient comprising measuring an expression profile of one or more miRNAs in a sample from the patient.
  • the difference in the expression profile in the sample from the patient and a reference expression profile is indicative of a pathologic, disease, or cancerous condition.
  • An miRNA or probe set comprising or identifying a segment of a corresponding miRNA can include all or part of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 ,13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 125, 150, 175, 200, 250, 300, 350, 400, 500, 550 to 562 or any integer or range derivable there between, of a miRNA or a probe or its complement listed in Table 1. It is contemplate the any one or subset of the miRNA listed in this application can be included or excluded from the claimed invention.
  • the methods for diagnosing a condition in a patient comprise measuring an expression profile of one or more miRNAs, from Table 1, in a sample from the patient, wherein the difference between the expression profile in the sample from the patient and an expression profile of a normal sample or a reference is indicative of a pathological condition.
  • the methods for diagnosing a condition in a patient comprises measuring an expression profile of one or more miRNAs in a sample from the patient, wherein the difference between the expression profile of the sample from the patient and an expression profile of a reference is indicative of a cervical tissue disease, or condition; wherein the miRNA is one or more of hsa-let-7a, hsa-let-7b, hsa-let-7c, hsa-let-7i, hsa-miR- 100, hsa-miR-101, hsa-miR-106a, hsa-miR-125a, hsa-miR-125b, hsa-miR-126-AS, hsa-miR- 127, hsa-miR-130a, hsa-miR-134, hsa-miR-142-3p, hsa-miR-143, hsa-miR-143,
  • a decrease in expression or down-regulation of hsa-miR-7 and/or hsa-miR-215 is also indicative of a cervical tissue.
  • the methods for diagnosing a condition in a patient comprise measuring an expression profile of one or more miRNAs in a sample or a cervix sample from the patient believed to be pre-cancerous or cancerous or contain precancerous or cancerous cells, wherein the difference between the expression profile in the sample from the patient and an expression profile of normal adjacent tissue (NAT) or a reference tissue is indicative of a disease or condition; wherein the miRNA is one or more miRNA listed in Table 1. It is contemplate the any one or subset of the miRNA listed in this application can be included or excluded from the claimed invention.
  • NAT normal adjacent tissue
  • the methods for diagnosing a condition in a patient comprise measuring an expression profile of one or more miRNAs in a cervix sample from a patient believed to be precancerous or cancerous, or contain precancerous or cancerous cells, or from a patient suspected of having or at risk of developing a pre-cancerous or cancerous condition, wherein the difference between the expression profile in the sample from the patient and an expression profile of normal cervix (NCX) or normal adjacent tissue (NAT) or a reference tissue is indicative of a disease or condition; wherein the miRNA is one or more of hsa-let-7a, hsa-let-7b, hsa-let-7c, hsa-let-7d, hsa-let-7e, hsa-let-7f, hsa-let-7g, hsa-let-7i, hsa-miR-1, hsa-miR-100, hsa-miR
  • the reduced expression or down regulation of hsa-miR-1, hsa-miR-133a, hsa-miR-204, hsa-miR-218, hsa-miR-143, hsa-miR-368, hsa-miR-99a, hsa- miR-100, hsa-miR-195, hsa-miR-376a, hsa-miR-424, hsa-miR-497, hsa-miR-299_5p, hsa- miR-154, hsa-miR-134, ambi-miR-7101 and/or ambi-miR-7029 or complement thereof relative to normal cervix tissue is indicative of a cancerous condition or tissue.
  • the increased or upregulation of hsa-miR-205, hsa-miR-183, hsa-miR-31, hsa-miR- 224, hsa-miR-182, hsa-miR-21 and/or hsa-miR-203 or complement thereof relative to normal cervix tissue is indicative of a cancerous condition or cancerous tissue.
  • the increased or upregulation of hsa-asg-11688_stl, hsa-miR-18b, hsa-miR-18a, hsa-asg- 3711_st2, hsa-miR-183, hsa-asg-2919_stl, hsa-asg-924_stl, and/or hsa-cand317_stl, or complement thereof relative to normal cervix tissue is indicative of a cancerous condition or cancerous tissue.
  • hsa-miR-93*, hsa-miR-940, hsa-miR-99a*, and/or hsa-miR-99b* or complement thereof relative to normal cervix tissue is indicative of a cancerous condition or tissue.
  • the increased or upregulation of hsa-miR-31*, hsa-miR-96, hsa-miR-21*, and/or hsa- miR-944 or complement thereof relative to normal cervix tissue is indicative of a cancerous condition or cancerous cells or cancerous tissue.
  • hsa-miR-16 can be used as reference.
  • Embodiments of the invention include analysis of miR expression by amplification assays.
  • the PCR assay is quantitative PCR and in particular real time quantitative reverse transcription PCR (qRT- PCR).
  • the methods for diagnosing a condition in a patient comprise measuring an expression profile of one or more miRNAs in a cervix sample from the patient suspected of having a cancerous condition, (e.g. , cervical squamous cell carcinoma) wherein the difference between the expression profile in the sample from the patient and an expression profile of normal tissue or a reference tissue is indicative of a cancerous disease or condition; wherein the miRNA is one or more of hsa-miR-1, hsa-miR- 100, hsa-miR-133a, hsa-miR-134, hsa-miR-143, hsa-miR-154, hsa-miR-182, hsa-miR-183, hsa-miR-195, hsa-miR-204, hsa-miR-205, hsa-miR-21, hsa-
  • the methods for diagnosing a condition in a patient comprise measuring an expression profile of one or more miRNAs in a cervix sample from the patient suspected of having a precancerous condition (e.g., cervical squamous intraepithelial lesion (SIL), also known as cervical intraepithelial neoplasias), wherein the difference between the expression profile in the sample from the patient and an expression profile of normal tissue or a reference is indicative of a disease or condition; wherein the miRNA is one or more of hsa-miR-1, hsa-miR-133a, hsa-miR-124a, hsa-miR-187, hsa-miR- 204, hsa-miR-145, hsa-miR-143, hsa-miR-325, hsa-miR-500, hsa-miR-196a, h
  • SIL cervical squamous
  • decreased expression, in a patient sample, of hsa- miR-1, hsa-miR-133a, hsa-miR-124a, hsa-miR-187, hsa-miR-204, hsa-miR-145, hsa-miR- 143, hsa-miR-325, hsa-miR-500, hsa-miR-196a, hsa-miR-125a, hsa-miR-376a, hsa-miR-505, hsa-miR-100, and/or hsa-miR-99a or complements thereof are indicative of SIL.
  • Increased expression of hsa-miR-141, hsa-miR-200a, ambi-miR-7029, hsa-miR-233, hsa-miR-205, hsa- miR-146a, hsa-miR-429, hsa-miR-200b, hsa-miR-182, hsa-miR-142-5p, hsa-miR-203, hsa- miR-21, hsa-miR-513, and/or hsa-miR-31 can be indicative of SIL.
  • Ct cycle threshold
  • Ct values may be determined for one or more of the miRNA listed in Table 1 or their complements.
  • the Ct values for miR-1, miR-21, or both miR-1 and miR-21 can be used to distinguish between pre-cancerous or cancerous cervical samples or tissues and normal or non-cancerous tissues.
  • a sample may be taken from a patient having or suspected of having a disease or pathological condition.
  • a sample may also comprise nucleic acids or RNA isolated from a tissue or cell sample from a patient.
  • the sample can be, but is not limited to tissue (e.g., biopsy, particularly fine needle biopsy), blood, serum, plasma, or cervical samples (e.g., pap smear or punch biopsy).
  • tissue e.g., biopsy, particularly fine needle biopsy
  • blood serum
  • plasma or cervical samples (e.g., pap smear or punch biopsy).
  • cervical samples e.g., pap smear or punch biopsy.
  • the sample can be fresh, frozen, fixed (e.g., formalin fixed), or embedded (e.g., paraffin embedded) tissues or cells.
  • the sample is a cervical sample or nucleic acid or RNA isolated therefrom.
  • Methods of the invention can be used to diagnose or assess a pathological condition.
  • the condition is a non-cancerous condition, such as pre-cancerous cervical lesion.
  • the condition is a cancerous condition, such as cervical cancer.
  • the methods can further comprise one or more steps including: (a) obtaining a sample from the patient, (b) isolating nucleic acids from the sample, (c) labeling the nucleic acids isolated from the sample, and (d) hybridizing the labeled nucleic acids to one or more probes or primers.
  • Nucleic acids of the invention include one or more nucleic acid comprising at least one segment having a sequence or complementary sequence of one or more of the miRNA sequences in Table 1. In certain aspects, the nucleic acids identify one or more miRNAs listed in Table 1.
  • Nucleic acids of the invention are typically coupled to a support. Such supports are well known to those of ordinary skill in the art and include, but are not limited to glass, plastic, metal, or latex. In particular aspects of the invention, the support can be planar or in the form of a bead or other geometric shapes or configurations.
  • an amplification assay can be a quantitative amplification assay, such as quantitative RT-PCR or the like.
  • a hybridization assay can include in situ hybridization, array hybridization assays or solution hybridization assays.
  • aspects of the invention can be used to diagnose or assess a patient's condition.
  • the methods can be used to screen for a pathological condition, assess prognosis of a pathological condition, stage a pathological condition, or assess response of a pathological condition to therapy.
  • Embodiments of the invention concern nucleic acids that perform the activities of or inhibit endogenous miRNAs when introduced into cells.
  • nucleic acids are synthetic or non-synthetic miRNA.
  • Sequence-specific miRNA inhibitors can be used to inhibit sequentially or in combination the activities of one or more endogenous miRNAs in cells, as well those genes and associated pathways modulated by the endogenous miRNA.
  • the present invention concerns, in some embodiments, short nucleic acid molecules that function as miRNAs or as inhibitors of miRNA in a cell.
  • short refers to a length of a single polynucleotide that is 5, 10, 15, 20, 25, 50, 100, or 150 nucleotides or fewer, including all integers or range derivable there between.
  • kits containing compositions of the invention or compositions to implement methods of the invention.
  • kits can be used to evaluate one or more miRNA molecules.
  • a kit contains, contains at least or contains at most 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 100, 200, 300, 400, 500 or more miRNA probes, miRNA molecules or miRNA inhibitors, or any range and combination derivable therein.
  • there are kits for evaluating or modulating miRNA activity in a cell are kits for evaluating or modulating miRNA activity in a cell.
  • Kits may comprise components, which may be individually packaged or placed in a container, such as a tube, bottle, vial, syringe, or other suitable container means.
  • Individual components may also be provided in a kit in concentrated amounts; in some embodiments, a component is provided individually in the same concentration as it would be in a solution with other components. Concentrations of components may be provided as Ix, 2x, 5x, 10x, or 2Ox or more.
  • Kits for using miRNA probes or primers, synthetic miRNAs, nonsynthetic miRNAs, and/or miRNA inhibitors of the invention for therapeutic, prognostic, or diagnostic applications are also included as part of the invention. Specifically contemplated are any such molecules corresponding to any miRNA reported to influence biological activity, such as those discussed herein.
  • negative and/or positive control synthetic miRNAs and/or miRNA inhibitors are included in some kit embodiments. The control molecules can be used to verify transfection efficiency and/or control for transfection-induced changes in cells.
  • any method or composition described herein can be implemented with respect to any other method or composition described herein and that different embodiments may be combined. It is specifically contemplated that any methods and compositions discussed herein with respect to miRNA molecules or miRNA may be implemented with respect to synthetic miRNAs to the extent the synthetic miRNA is exposed to the proper conditions to allow it to become a mature miRNA under physiological circumstances.
  • the claims originally filed are contemplated to cover claims that are multiply dependent on any filed claim or combination of filed claims.
  • any one or more of the miRNA listed, particularly in Table 1 may be specifically excluded from any particular set or subset of miRNA or nucleic acid.
  • Any embodiment of the invention involving specific miRNAs by name is contemplated also to cover embodiments involving miRNAs whose sequences are at least 70, 75, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98% identical to the mature sequence of the specified miRNA. This also includes the various fragments of these miRNA or nucleic acid sequences.
  • Embodiments of the invention include kits for analysis of a pathological sample by assessing miRNA profile for a sample comprising, in suitable container means, two or more miRNA probes, wherein the miRNA probes detect one or more of the miRNAs described in Table 1.
  • the kit can further comprise reagents for labeling miRNA in the sample.
  • the kit may also include the labeling reagents include at least one amine-modified nucleotide, poly(A) polymerase, and poly(A) polymerase buffer. Labeling reagents can include an amine-reactive dye.
  • miR-16-1 and miR-16-2 are members of the miR-16 gene family and "mir-7" refers to miR-7-1, miR-7-2 and miR-7-3.
  • a shorthand notation refers to related miRNAs (distinguished by a letter).
  • let-7 refers to let-7a-l, let7-a-2, let-7b, let- 7c, let-7d, let-7e, let-7f-l, and let-7f-2.” Exceptions to this shorthand notations will be otherwise identified.
  • FIG. 1 Principal Component Analysis of miRNAs expressed in normal cervix sample (NCX), cancerous cervix samples (Ca), and paired normal adjacent cervix samples (NAT).
  • FIGs. 2A-2B Comparison between array and qRT-PCR data.
  • Real time RT-PCR were performed using 15 ng of total RNA input from the 34 samples previously profiled (16 NCX and 9 paired Ca and NAT samples).
  • miRNA expression data obtained with primer sets specific for the indicated miRNAs were normalized to miR-16 expression level for each sample (miRNA Ct - miR-16 Ct).
  • the graphs show the individual normalized miRNA expression levels determined by array or qRT-PCR and associated p-values (p (ANOVA) and *p (p (pairwise t-test)).
  • FIG. 3 Principal Component Analysis of miRNAs expressed in normal cervix sample (NCX), cancerous cervix samples (Ca) and paired normal adjacent cervix samples (NAT), and cervical cancer cell lines (CL).
  • FIG. 4 Expression of miR-21 and miR-1 classifies normal and cancerous cervical tissues. Real time RT-PCR were performed with primer sets specific for miR-1 and miR-21 using 15 ng total RNA from 16 normal cervix (NCX), 9 cancerous cervix (Ca), or 9 normal adjacent cervix (NAT) samples. Raw Ct values were directly used to calculate the ratio of miR-21 to miR-1 expression, i.e., miR-21 Ct - miRl Ct in the logarithmic space.
  • the present invention is directed to compositions and methods relating to preparation and characterization of miRNAs, as well as use of miRNAs for therapeutic, prognostic, and diagnostic applications, particularly those methods and compositions related to assessing and/or identifying cervical diseases and conditions.
  • cervical cancer is the second most common cause of cancer in women worldwide (Pisani et al, 2002; Parkin et al, 2005). About 470,000 new cases are diagnosed and approximately 230,000 women die of cervical cancer every year (Pisani et al, 1999). While the majority (-80%) of these new cases and deaths occur in developing countries, it is estimated that approximately 3,700 women will die from invasive cervical cancer in the United States in 2007 (Jemal et al, 2007).
  • Pap smear Cytological examination of cervical smears with Papanicolaou staining (Pap smear) is the screening method universally accepted for early detection of cervical cancer and its precursors. Pap smear screening programs have been very effective in reducing cervical cancer incidence and have reduced mortality rates by 60% among women aged 30 and over. However, even in those countries where these programs are routinely used, cervical cancer is still a significant public health problem.
  • the Bethesda system categorizes Pap smear results as negative, ASC-US (atypical squamous cells of undetermined significance), ASCU-H (atypical squamous cells, cannot exclude high grade lesion), LSIL (low grade squamous intraepithelial lesion), HSIL (high grade intraepithelial lesion) and carcinoma. While guidelines and protocols for the management of women diagnosed with ASC-H, LSIL, HSIL and cancer are well established, the ASC-US entity is a significant problem for clinicians. The morphological criteria for ASC-US are suggestive, resulting in great variations in reported rates for ASC-US between laboratories.
  • HPVs human papillomaviruses
  • HPV types including types 16, 18, 31 and 45
  • HPV types are called "high-risk" types because they can lead to cervical cancer, as well as anal cancer, vulvar cancer, and penile cancer.
  • HPV-induced cancers often have viral sequences integrated into the cellular DNA.
  • HPV "early" genes such as E6 and E7, are known to act as oncogenes that promote tumor growth and malignant transformation.
  • miRNAs are generally 21 to 22 nucleotides in length, though lengths of 19 and up to 23 nucleotides have been reported.
  • the miRNAs are each processed from a longer precursor RNA molecule ("precursor miRNA").
  • Precursor miRNAs are transcribed from non-protein-encoding genes.
  • the precursor miRNAs have two regions of complementarity that enables them to form a stem-loop- or fold-back-like structure, which is cleaved in animals by a ribonuclease Ill-like nuclease enzyme called Dicer.
  • the processed miRNA is typically a portion of the stem.
  • the processed miRNA (also referred to as "mature miRNA”) become part of a large complex to down-regulate a particular target gene.
  • animal miRNAs include those that imperfectly basepair with the target, which halts translation (Olsen et al, 1999; Seggerson et al, 2002).
  • siRNA molecules also are processed by Dicer, but from a long, double-stranded RNA molecule. siRNAs are not naturally found in animal cells, but they can direct the sequence-specific cleavage of an mRNA target through a RNA-induced silencing complex (RISC) (Denli et al, 2003).
  • RISC RNA-induced silencing complex
  • the nucleic acid molecules are typically synthetic.
  • nucleic acid molecule is isolated and not identical in sequence and/or chemical structure to a naturally-occurring nucleic acid molecule, such as an endogenous precursor miRNA or miRNA molecule. While in some embodiments, nucleic acids of the invention do not have an entire sequence that is identical to a sequence of a naturally-occurring nucleic acid, such molecules may encompass all or part of a naturally-occurring sequence. It is contemplated, however, that a synthetic nucleic acid administered to a cell may subsequently be modified or altered in the cell such that its structure or sequence is the same or similar as non-synthetic or naturally occurring nucleic acid, such as a mature miRNA sequence.
  • a synthetic nucleic acid may have a sequence that differs from the sequence of a precursor miRNA, but that sequence may be altered once in a cell to be the same as an endogenous, processed miRNA.
  • isolated means that the nucleic acid molecules of the invention are initially separated from different (in terms of sequence or structure) and unwanted nucleic acid molecules such that a population of isolated nucleic acids is at least about 90% homogenous, and may be at least about 95, 96, 97, 98, 99, or 100% homogenous with respect to other polynucleotide molecules.
  • a nucleic acid is isolated by virtue of it having been synthesized in vitro separate from endogenous nucleic acids in a cell. It will be understood, however, that isolated nucleic acids may be subsequently mixed or pooled together in a variety of combinations.
  • synthetic miRNA of the invention are RNA or RNA analogs.
  • miRNA inhibitors may be DNA or RNA, or analogs thereof.
  • miRNA and miRNA inhibitors of the invention are typically "synthetic nucleic acids.”
  • RNA molecules that are, are at least, or are at most 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 61, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106
  • synthetic miRNA have (a) an "miRNA region” whose sequence from 5' to 3' is identical to all or a segment of a mature miRNA sequence, and (b) a "complementary region” whose sequence from 5' to 3' is between 60% and 100% complementary to the miRNA sequence.
  • miRNA region refers to a region on the synthetic miRNA that is at least 75, 80, 85, 90, 95, or 100% identical, including all integers there between, to all or part of the sequence of a mature, naturally occurring miRNA sequence.
  • the miRNA region is or is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9 or 100% identical to the sequence of a naturally-occurring miRNA.
  • complementary region refers to a region of a synthetic miRNA that is or is at least 60% complementary to a corresponding naturally occurring miRNA sequence.
  • the complementary region is or is at least 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9 or 100% complementary to its corresponding naturally occurring miRNA, or any range derivable therein.
  • the complementary region is on a different nucleic acid molecule than the miRNA region, in which case the complementary region is on the complementary strand and the miRNA region is on the active or functional strand.
  • an miRNA inhibitor is between about 17 to 25 nucleotides in length and comprises a 5' to 3' sequence that is at least 90% complementary to the 5' to 3' sequence of a mature miRNA.
  • an miRNA inhibitor molecule is 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleotides in length, or any range derivable therein.
  • an miRNA inhibitor has a sequence (from 5' to 3') that is or is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9 or 100% complementary, or any range derivable therein, to the 5' to 3' sequence of a mature miRNA, particularly a mature, naturally occurring miRNA.
  • One of skill in the art could use a portion of a sequence that is complementary to the sequence of a mature miRNA as the sequence for an miRNA inhibitor. Moreover, that portion of a sequence can be altered so that it is still 90% complementary to the sequence of a mature miRNA.
  • a synthetic miRNA contains one or more design elements. These design elements include, but are not limited to: (i) a replacement group for the phosphate or hydroxyl of the nucleotide at the 5' terminus of the complementary region; (ii) one or more sugar modifications in the first or last 1 to 6 residues of the complementary region; or (iii) noncomplementarity between one or more nucleotides in the last 1 to 5 residues at the 3' end of the complementary region and the corresponding nucleotides of the miRNA region.
  • a synthetic miRNA has a nucleotide at its 5' end of the complementary region in which the phosphate and/or hydroxyl group has been replaced with another chemical group (referred to as the "replacement design").
  • the replacement design referred to as the "replacement design”.
  • the phosphate group is replaced, while in others, the hydroxyl group has been replaced.
  • the replacement group is biotin, an amine group, a lower alkylamine group, an acetyl group, 2'0-Me (2 Oxygen-methyl), DMTO (4,4'-dimethoxytrityl with oxygen), fluorescein, a thiol, or acridine, though other replacement groups are well known to those of skill in the art and can be used as well.
  • This design element can also be used with an miRNA inhibitor.
  • Additional embodiments concern a synthetic miRNA having one or more sugar modifications in the first or last 1 to 6 residues of the complementary region (referred to as the "sugar replacement design").
  • sugar modifications in the first 1, 2, 3, 4, 5, 6 or more residues of the complementary region, or any range derivable therein.
  • first and last are with respect to the order of residues from the 5' end to the 3' end of the region.
  • the sugar modification is a 2'O- Me modification.
  • an miRNA inhibitor can have this design element and/or a replacement group on the nucleotide at the 5' terminus, as discussed above.
  • noncomplementarity design there is a synthetic miRNA in which one or more nucleotides in the last 1 to 5 residues at the 3' end of the complementary region are not complementary to the corresponding nucleotides of the miRNA region.
  • the noncomplementarity may be in the last 1, 2, 3, 4, and/or 5 residues of the complementary miRNA.
  • synthetic miRNA of the invention have one or more of the replacement, sugar modification, or noncomplementarity designs.
  • synthetic RNA molecules have two of them, while in others these molecules have all three designs in place.
  • the miRNA region and the complementary region may be on the same or separate polynucleotides. In cases in which they are contained on or in the same polynucleotide, the miRNA molecule will be considered a single polynucleotide. In embodiments in which the different regions are on separate polynucleotides, the synthetic miRNA will be considered to be comprised of two polynucleotides.
  • the RNA molecule is a single polynucleotide
  • the single polynucleotide is capable of forming a hairpin loop structure as a result of bonding between the miRNA region and the complementary region.
  • the linker constitutes the hairpin loop. It is contemplated that in some embodiments, the linker region is, is at least, or is at most 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 residues in length, or any range derivable therein. In certain embodiments, the linker is between 3 and 30 residues (inclusive) in length.
  • flanking sequences as well at either the 5' or 3' end of the region.
  • the present invention concerns miRNAs that can be labeled or amplified, used in array analysis, or employed in diagnostic, therapeutic, or prognostic applications, particularly those related to diseases and conditions of the cervix.
  • the RNA may have been endogenously produced by a cell, or been synthesized or produced chemically or recombinantly. They may be isolated and/or purified.
  • miRNA refers to the processed RNA, after it has been cleaved from its precursor. Table 1 indicates which SEQ ID NO correspond to a mature miRNA sequence.
  • miRNAs referred to in the application are human sequences identified as mir-X or let-X, where X is a number and/or letter.
  • a miRNA is designated with a "5P" or "3P" suffix.
  • “5P” indicates that the mature miRNA derives from the 5' end of the precursor and a corresponding "3P” indicates that it derives from the 3' end of the precursor, as described on the world wide web at sanger.ac.uk.
  • a miRNA is used that does not correspond to a known human miRNA. It is contemplated that these non-human miRNA probes may be used in embodiments of the invention or that there may exist a human miRNA that is homologous to the non-human miRNA. While the invention is not limited to human miRNA, in certain embodiments, miRNA from human cells or a human biological sample is evaluated. In other embodiments, any mammalian cell, biological sample, or preparation thereof may be employed.
  • nucleic acids may be, be at least, or be at most 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103,
  • Such lengths cover the lengths of processed miRNA, miRNA probes, precursor miRNA, miRNA containing vectors, control nucleic acids, and other probes and primers.
  • miRNA sequences are 19-24 nucleotides in length
  • miRNA probes are 19-35 nucleotides in length, depending on the length of the processed miRNA and any flanking regions added.
  • miRNA precursors are generally between 62 and 110 nucleotides in humans.
  • Nucleic acids, and mimetics thereof, of the invention may have regions of identity or complementarity to another nucleic acid.
  • the region of complementarity or identity can be at least 5 contiguous residues, though it is specifically contemplated that the region is, is at least, or is at most 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 110, 120, 130, 140, 150, 160, 170
  • complementarity within a precursor miRNA or between a miRNA probe and a miRNA or a miRNA gene are such lengths.
  • the complementarity may be expressed as a percentage, meaning that the complementarity between a nucleic acid and its target is 90% or greater over the length of the nucleic acid.
  • complementarity is or is at least 90%, 95% or 100%.
  • such lengths may be applied to any nucleic acid comprising a nucleic acid sequence identified in any of SEQ ID NO:1 through SEQ ID NO:562 or any other sequence disclosed herein. Each of these SEQ ID NOs is disclosed herein.
  • miRNA probe refers to a nucleic acid probe that can identify a particular miRNA or structurally related miRNAs.
  • a miRNA is derived from genomic sequences or a gene.
  • the term "gene” is used for simplicity to refer to the genomic sequence encoding the precursor miRNA for a given miRNA.
  • embodiments of the invention may involve genomic sequences of a miRNA that are involved in its expression, such as a promoter or other regulatory sequences.
  • nucleic acid is well known in the art.
  • a “nucleic acid” as used herein will generally refer to a molecule (one or more strands) of DNA, RNA or a derivative or analog thereof, comprising a nucleobase.
  • a nucleobase includes, for example, a naturally occurring purine or pyrimidine base found in DNA (e.g., an adenine "A,” a guanine “G,” a thymine “T” or a cytosine “C”) or RNA (e.g., an A, a G, an uracil “U” or a C).
  • DNA e.g., an adenine "A,” a guanine "G,” a thymine “T” or a cytosine "C”
  • RNA e.g., an A, a G, an uracil "U” or a C.
  • nucleic acid encompasses the terms “oligonucleotide” and “polynucleotide,” each as a subgenus of the term “nucleic acid.”
  • miRNA generally refers to a single-stranded molecule, but in specific embodiments, molecules implemented in the invention will also encompass a region or an additional strand that is partially (between 10 and 50% complementary across length of strand), substantially (greater than 50% but less than 100% complementary across length of strand) or fully complementary to another region of the same single-stranded molecule or to another nucleic acid.
  • nucleic acids may encompass a molecule that comprises one or more complementary or self-complementary strand(s) or "complement(s)" of a particular sequence comprising a molecule.
  • precursor miRNA may have a self- complementary region, which is up to 100% complementary.
  • miRNA probes or nucleic acids of the invention can include, can be or can be at least 60, 65, 70, 75, 80, 85, 90, 95, 96, 97, 98, 99 or 100% complementary to their target.
  • hybridization As used herein, “hybridization,” “hybridizes,” or “capable of hybridizing” is understood to mean the forming of a double or triple stranded molecule or a molecule with partial double or triple stranded nature.
  • anneal as used herein is synonymous with “hybridize.”
  • hybridization encompasses hybridization under "stringent condition(s)” or “high stringency” and “low stringency” or “low stringency condition(s).”
  • stringent condition(s) or “high stringency” are those conditions that allow hybridization between or within one or more nucleic acid strand(s) containing complementary sequence(s), but preclude hybridization of random sequences. Stringent conditions tolerate little, if any, mismatch between a nucleic acid and a target strand. Such conditions are well known to those of ordinary skill in the art, and are preferred for applications requiring high selectivity. Non-limiting applications include isolating a nucleic acid, such as a gene or a nucleic acid segment thereof, or detecting at least one specific mRNA transcript or a nucleic acid segment thereof, and the like.
  • Stringent conditions may comprise low salt and/or high temperature conditions, such as provided by about 0.02 M to about 0.5 M NaCl at temperatures of about 42°C to about 70°C. It is understood that the temperature and ionic strength of a desired stringency are determined in part by the length of the particular nucleic acid(s), the length and nucleobase content of the target sequence(s), the charge composition of the nucleic acid(s), and to the presence or concentration of formamide, tetramethylammonium chloride or other solvent(s) in a hybridization mixture.
  • low stringency or “low stringency conditions”
  • non-limiting examples of low stringency include hybridization performed at about 0.15 M to about 0.9 M NaCl at a temperature range of about 20°C to about 50°C.
  • hybridization performed at about 0.15 M to about 0.9 M NaCl at a temperature range of about 20°C to about 50°C.
  • nucleobase refers to a heterocyclic base, such as for example a naturally occurring nucleobase (i.e., an A, T, G, C or U) found in at least one naturally occurring nucleic acid (i.e., DNA and RNA), and naturally or non-naturally occurring derivative(s) and analogs of such a nucleobase.
  • a nucleobase generally can form one or more hydrogen bonds (“anneal” or “hybridize”) with at least one naturally occurring nucleobase in manner that may substitute for naturally occurring nucleobase pairing (e.g., the hydrogen bonding between A and T, G and C, and A and U).
  • Preferredine and/or "pyrimidine” nucleobase(s) encompass naturally occurring purine and/or pyrimidine nucleobases and also derivative(s) and analog(s) thereof, including but not limited to, those a purine or pyrimidine substituted by one or more of an alkyl, carboxyalkyl, amino, hydroxyl, halogen (i.e., fluoro, chloro, bromo, or iodo), thiol or alkylthiol moiety.
  • Preferred alkyl (e.g., alkyl, carboxyalkyl, etc.) moieties comprise of from about 1, about 2, about 3, about 4, about 5, to about 6 carbon atoms.
  • a purine or pyrimidine include a deazapurine, a 2,6-diaminopurine, a 5-fluorouracil, a xanthine, a hypoxanthine, a 8-bromoguanine, a 8-chloroguanine, a bromothymine, a 8-aminoguanine, a 8- hydroxyguanine, a 8-methylguanine, a 8-thioguanine, an azaguanine, a 2-aminopurine, a 5- ethylcytosine, a 5-methylcyosine, a 5-bromouracil, a 5-ethyluracil, a 5-iodouracil, a 5- chlorouracil, a 5-propyluracil, a thiouracil, a 2-methyladenine, a methylthioadenine, a N 5 N- diemethyladenine, an azaguanine
  • a nucleobase may be comprised in a nucleoside or nucleotide, using any chemical or natural synthesis method described herein or known to one of ordinary skill in the art. Such nucleobase may be labeled or it may be part of a molecule that is labeled and contains the nucleobase.
  • nucleoside refers to an individual chemical unit comprising a nucleobase covalently attached to a nucleobase linker moiety.
  • a non-limiting example of a “nucleobase linker moiety” is a sugar comprising 5-carbon atoms (i.e., a "5-carbon sugar"), including but not limited to a deoxyribose, a ribose, an arabinose, or a derivative or an analog of a 5-carbon sugar.
  • Non-limiting examples of a derivative or an analog of a 5-carbon sugar include a 2'-fluoro-2'-deoxyribose or a carbocyclic sugar where a carbon is substituted for an oxygen atom in the sugar ring.
  • nucleoside comprising a purine (i.e., A or G) or a 7-deazapurine nucleobase typically covalently attaches the 9 position of a purine or a 7-deazapurine to the 1 '-position of a 5-carbon sugar.
  • a nucleoside comprising a pyrimidine nucleobase typically covalently attaches a 1 position of a pyrimidine to a 1 '-position of a 5-carbon sugar (Kornberg and Baker, 1992).
  • nucleotide refers to a nucleoside further comprising a "backbone moiety.”
  • a backbone moiety generally covalently attaches a nucleotide to another molecule comprising a nucleotide, or to another nucleotide to form a nucleic acid.
  • the "backbone moiety” in naturally occurring nucleotides typically comprises a phosphorus moiety, which is covalently attached to a 5-carbon sugar. The attachment of the backbone moiety typically occurs at either the 3'- or 5'-position of the 5-carbon sugar.
  • other types of attachments are known in the art, particularly when a nucleotide comprises derivatives or analogs of a naturally occurring 5-carbon sugar or phosphorus moiety.
  • a nucleic acid may comprise, or be composed entirely of, a derivative or analog of a nucleobase, a nucleobase linker moiety and/or backbone moiety that may be present in a naturally occurring nucleic acid.
  • RNA with nucleic acid analogs may also be labeled according to methods of the invention.
  • a "derivative” refers to a chemically modified or altered form of a naturally occurring molecule, while the terms “mimic” or “analog” refer to a molecule that may or may not structurally resemble a naturally occurring molecule or moiety, but possesses similar functions.
  • a "moiety” generally refers to a smaller chemical or molecular component of a larger chemical or molecular structure.
  • Nucleobase, nucleoside and nucleotide analogs or derivatives are well known in the art, and have been described (see for example, Scheit, 1980, incorporated herein by reference).
  • nucleosides examples include those in: U.S. Patent 5,681,947, which describes oligonucleotides comprising purine derivatives that form triple helixes with and/or prevent expression of dsDNA; U.S. Patents 5,652,099 and 5,763,167, which describe nucleic acids incorporating fluorescent analogs of nucleosides found in DNA or RNA, particularly for use as fluorescent nucleic acids probes; U.S.
  • Patent 5,614,617 which describes oligonucleotide analogs with substitutions on pyrimidine rings that possess enhanced nuclease stability
  • U.S. Patents 5,670,663, 5,872,232 and 5,859,221 which describe oligonucleotide analogs with modified 5-carbon sugars (i.e., modified T- deoxyfuranosyl moieties) used in nucleic acid detection
  • U.S. Patent 5,446,137 which describes oligonucleotides comprising at least one 5-carbon sugar moiety substituted at the 4' position with a substituent other than hydrogen that can be used in hybridization assays
  • Patent 5,886,165 which describes oligonucleotides with both deoxyribonucleotides with 3'-5' internucleotide linkages and ribonucleotides with 2'-5' internucleotide linkages
  • U.S. Patent 5,714,606 which describes a modified internucleotide linkage wherein a 3'-position oxygen of the internucleotide linkage is replaced by a carbon to enhance the nuclease resistance of nucleic acids
  • U.S. Patent 5,672,697 which describes oligonucleotides containing one or more 5' methylene phosphonate internucleotide linkages that enhance nuclease resistance
  • Patents 5,466,786 and 5,792,847 which describe the linkage of a substituent moiety which may comprise a drug or label to the 2' carbon of an oligonucleotide to provide enhanced nuclease stability and ability to deliver drugs or detection moieties;
  • U.S. Patent 5,223,618, which describes oligonucleotide analogs with a 2 or 3 carbon backbone linkage attaching the 4' position and 3' position of adjacent 5-carbon sugar moiety to enhanced cellular uptake, resistance to nucleases and hybridization to target RNA;
  • Patent 5,470,967 which describes oligonucleotides comprising at least one sulfamate or sulfamide internucleotide linkage that are useful as nucleic acid hybridization probe;
  • U.S. Patents 5,378,825, 5,777,092, 5,623,070, 5,610,289 and 5,602,240 which describe oligonucleotides with three or four atom linker moiety replacing phosphodiester backbone moiety used for improved nuclease resistance, cellular uptake, and regulating RNA expression;
  • U.S. Patent 5,858,988, which describes hydrophobic carrier agent attached to the 2'-O position of oligonucleotides to enhanced their membrane permeability and stability;
  • Patent 5,214,136 which describes oligonucleotides conjugated to anthraquinone at the 5' terminus that possess enhanced hybridization to DNA or RNA; enhanced stability to nucleases; U.S. Patent 5,700,922, which describes PNA-DNA-PNA chimeras wherein the DNA comprises T- deoxy-erythro-pentofuranosyl nucleotides for enhanced nuclease resistance, binding affinity, and ability to activate RNase H; and U.S. Patent 5,708,154, which describes RNA linked to a DNA to form a DNA-RNA hybrid; U.S. Patent 5,728,525, which describes the labeling of nucleoside analogs with a universal fluorescent label.
  • nucleoside analogs and nucleic acid analogs are U.S. Patent 5,728,525, which describes nucleoside analogs that are end-labeled; U.S. Patent 5,637,683, 6,251,666 (L-nucleotide substitutions), and 5,480,980 (7-deaza-2'deoxyguanosine nucleotides and nucleic acid analogs thereof). 5. Modified Nucleotides
  • Labeling methods and kits of the invention specifically contemplate the use of nucleotides that are both modified for attachment of a label and can be incorporated into a miRNA molecule.
  • Such nucleotides include those that can be labeled with a dye, including a fluorescent dye, or with a molecule such as biotin. Labeled nucleotides are readily available; they can be acquired commercially or they can be synthesized by reactions known to those of skill in the art.
  • Modified nucleotides for use in the invention are not naturally occurring nucleotides, but instead, refer to prepared nucleotides that have a reactive moiety on them.
  • Specific reactive functionalities of interest include: amino, sulfhydryl, sulfoxyl, aminosulfhydryl, azido, epoxide, isothiocyanate, isocyanate, anhydride, monochlorotriazine, dichlorotriazine, mono-or dihalogen substituted pyridine, mono- or disubstituted diazine, maleimide, epoxide, aziridine, sulfonyl halide, acid halide, alkyl halide, aryl halide, alkylsulfonate, N- hydroxysuccinimide ester, imido ester, hydrazine, azidonitrophenyl, azide, 3-(2-pyridyl dithio)-propionamide,
  • the reactive functionality may be bonded directly to a nucleotide, or it may be bonded to the nucleotide through a linking group.
  • the functional moiety and any linker cannot substantially impair the ability of the nucleotide to be added to the miRNA or to be labeled.
  • Representative linking groups include carbon containing linking groups, typically ranging from about 2 to 18, usually from about 2 to 8 carbon atoms, where the carbon containing linking groups may or may not include one or more heteroatoms, e.g. S, O, N etc., and may or may not include one or more sites of unsaturation.
  • alkyl linking groups typically lower alkyl linking groups of 1 to 16, usually 1 to 4 carbon atoms, where the linking groups may include one or more sites of unsaturation.
  • the functionalized nucleotides (or primers) used in the above methods of functionalized target generation may be fabricated using known protocols or purchased from commercial vendors, e.g., Sigma, Roche, Ambion, Biosearch Technologies and NEN.
  • Functional groups may be prepared according to ways known to those of skill in the art, including the representative information found in U.S. Patents 4,404,289; 4,405,711; 4,337,063 and 5,268,486, and U.K. Patent 1,529,202, which are all incorporated by reference.
  • Amine-modified nucleotides are used in several embodiments of the invention.
  • the amine-modified nucleotide is a nucleotide that has a reactive amine group for attachment of the label. It is contemplated that any ribonucleotide (G, A, U, or C) or deoxyribonucleotide (G, A, T, or C) can be modified for labeling.
  • Examples include, but are not limited to, the following modified ribo- and deoxyribo-nucleotides: 5-(3-aminoallyl)-UTP; 8-[(4- amino)butyl]-amino-ATP and 8-[(6-amino)butyl]-amino-ATP; N6-(4-amino)butyl-ATP, N6- (6-amino)butyl-ATP, N4-[2,2-oxy-bis-(ethylamine)]-CTP; N6-(6-Amino)hexyl-ATP; 8-[(6- Amino)hexyl] -amino- ATP; 5-propargylamino-CTP, 5-propargylamino-UTP; 5-(3- aminoallyl)-dUTP; 8-[(4-amino)butyl]-amino-dATP and 8-[(6-amino)butyl]-amino-d
  • nucleotides can be prepared according to methods known to those of skill in the art. Moreover, a person of ordinary skill in the art could prepare other nucleotide entities with the same amine-modification, such as a 5-(3-aminoallyl)-CTP, GTP, ATP, dCTP, dGTP, dTTP, or dUTP in place of a 5-(3-aminoallyl)-UTP.
  • a nucleic acid may be made or prepared by any technique known to one of ordinary skill in the art, such as for example, chemical synthesis, enzymatic production or biological production. It is specifically contemplated that nucleic acids of the invention are chemically synthesized.
  • miRNAs are recovered or isolated from a biological sample.
  • the miRNA may be recombinant or it may be natural or endogenous to the cell (produced from the cell's genome). It is contemplated that a biological sample may be treated in a way so as to enhance the recovery of small RNA molecules such as miRNA.
  • U.S. Patent Application Serial No. 10/667,126 describes such methods and it is specifically incorporated by reference herein. Generally, methods involve lysing cells with a solution having guanidinium and a detergent.
  • nucleic acid synthesis is performed according to standard methods. See, for example, Itakura and Riggs (1980). Additionally, U.S. Patents 4,704,362, 5,221,619, and 5,583,013 each describe various methods of preparing synthetic nucleic acids.
  • Non- limiting examples of a synthetic nucleic acid include a nucleic acid made by in vitro chemically synthesis using phosphotriester, phosphite, or phosphoramidite chemistry and solid phase techniques such as described in EP 266,032, incorporated herein by reference, or via deoxynucleoside H-phosphonate intermediates as described by Froehler et al, 1986 and U.S. Patent 5,705,629, each incorporated herein by reference.
  • one or more oligonucleotide may be used.
  • Various different mechanisms of oligonucleotide synthesis have been disclosed in for example, U.S. Patents 4,659,774, 4,816,571, 5,141,813, 5,264,566, 4,959,463, 5,428,148, 5,554,744, 5,574,146, 5,602,244, each of which is incorporated herein by reference.
  • a non-limiting example of an enzymatically produced nucleic acid include one produced by enzymes in amplification reactions such as PCRTM (see for example, U.S. Patents 4,683,202 and 4,682,195, each incorporated herein by reference), or the synthesis of an oligonucleotide described in U.S. Patent 5,645,897, incorporated herein by reference.
  • a non-limiting example of a biologically produced nucleic acid includes a recombinant nucleic acid produced (i.e., replicated) in a living cell, such as a recombinant DNA vector replicated in bacteria (see for example, Sambrook et al., 2001, incorporated herein by reference).
  • Oligonucleotide synthesis is well known to those of skill in the art. Various different mechanisms of oligonucleotide synthesis have been disclosed in for example, U.S. Patents 4,659,774, 4,816,571, 5,141,813, 5,264,566, 4,959,463, 5,428,148, 5,554,744, 5,574,146, 5,602,244, each of which is incorporated herein by reference.
  • Polynucleotide phosphorylase method is an enzymatic method of DNA synthesis that can be used to synthesize many useful oligonucleotides (Gillam et al., 1978; Gillam et al, 1979). Under controlled conditions, polynucleotide phosphorylase adds predominantly a single nucleotide to a short oligonucleotide. Chromatographic purification allows the desired single adduct to be obtained. At least a trimer is required to start the procedure, and this primer must be obtained by some other method. The polynucleotide phosphorylase method works and has the advantage that the procedures involved are familiar to most biochemists.
  • Phosphoramidite chemistry (Beaucage and Lyer, 1992) has become by far the most widely used coupling chemistry for the synthesis of oligonucleotides.
  • Phosphoramidite synthesis of oligonucleotides involves activation of nucleoside phosphoramidite monomer precursors by reaction with an activating agent to form activated intermediates, followed by sequential addition of the activated intermediates to the growing oligonucleotide chain (generally anchored at one end to a suitable solid support) to form the oligonucleotide product.
  • Recombinant methods for producing nucleic acids in a cell are well known to those of skill in the art. These include the use of vectors (viral and non- viral), plasmids, cosmids, and other vehicles for delivering a nucleic acid to a cell, which may be the target cell (e.g., a cancer cell) or simply a host cell (to produce large quantities of the desired RNA molecule). Alternatively, such vehicles can be used in the context of a cell free system so long as the reagents for generating the RNA molecule are present. Such methods include those described in Sambrook, 2003, Sambrook, 2001 and Sambrook, 1989, which are hereby incorporated by reference.
  • the present invention concerns nucleic acid molecules that are not synthetic.
  • the nucleic acid molecule has a chemical structure of a naturally occurring nucleic acid and a sequence of a naturally occurring nucleic acid, such as the exact and entire sequence of a single stranded primary miRNA (see Lee 2002), a single-stranded precursor miRNA, or a single-stranded mature miRNA.
  • non-synthetic nucleic acids may be generated chemically, such as by employing technology used for creating oligonucleotides.
  • Nucleic acids may be isolated using techniques well known to those of skill in the art, though in particular embodiments, methods for isolating small nucleic acid molecules, and/or isolating RNA molecules can be employed. Chromatography is a process often used to separate or isolate nucleic acids from protein or from other nucleic acids. Such methods can involve electrophoresis with a gel matrix, filter columns, alcohol precipitation, and/or other chromatography.
  • methods generally involve lysing the cells with a chaotropic (e.g., guanidinium isothiocyanate) and/or detergent (e.g., N- lauroyl sarcosine) prior to implementing processes for isolating particular populations of RNA.
  • a chaotropic e.g., guanidinium isothiocyanate
  • detergent e.g., N- lauroyl sarcosine
  • Methods may involve the use of organic solvents and/or alcohol to isolate nucleic acids, particularly miRNA used in methods and compositions of the invention.
  • Some embodiments are described in U.S. Patent Application Serial No. 10/667,126, which is hereby incorporated by reference.
  • this disclosure provides methods for efficiently isolating small RNA molecules from cells comprising: adding an alcohol solution to a cell lysate and applying the alcohol/lysate mixture to a solid support before eluting the RNA molecules from the solid support.
  • the amount of alcohol added to a cell lysate achieves an alcohol concentration of about 55% to 60%. While different alcohols can be employed, ethanol works well.
  • a solid support may be any structure, and it includes beads, filters, and columns, which may include a mineral or polymer support with electronegative groups. A glass fiber filter or column has worked particularly well for such isolation procedures.
  • miRNA isolation processes include: a) lysing cells in the sample with a lysing solution comprising guanidinium, wherein a lysate with a concentration of at least about 1 M guanidinium is produced; b) extracting miRNA molecules from the lysate with an extraction solution comprising phenol; c) adding to the lysate an alcohol solution for form a lysate/alcohol mixture, wherein the concentration of alcohol in the mixture is between about 35% to about 70%; d) applying the lysate/alcohol mixture to a solid support; e) eluting the miRNA molecules from the solid support with an ionic solution; and, f) capturing the miRNA molecules.
  • the sample is dried down and resuspended in a liquid and volume appropriate for subsequent manipulation.
  • the present invention concerns miRNA that are labeled. It is contemplated that miRNA may first be isolated and/or purified prior to labeling. This may achieve a reaction that more efficiently labels the miRNA, as opposed to other RNA in a sample in which the miRNA is not isolated or purified prior to labeling.
  • the label is non-radioactive.
  • nucleic acids may be labeled by adding labeled nucleotides (one-step process) or adding nucleotides and labeling the added nucleotides (two-step process).
  • nucleic acids are labeled by catalytically adding to the nucleic acid an already labeled nucleotide or nucleotides.
  • One or more labeled nucleotides can be added to miRNA molecules. See U.S. Patent 6,723,509, which is hereby incorporated by reference.
  • an unlabeled nucleotide or nucleotides is catalytically added to a miRNA, and the unlabeled nucleotide is modified with a chemical moiety that enables it to be subsequently labeled.
  • the chemical moiety is a reactive amine such that the nucleotide is an amine-modified nucleotide. Examples of amine- modified nucleotides are well known to those of skill in the art, many being commercially available such as from Ambion, Sigma, Jena Bioscience, and TriLink.
  • the present invention concerns the use of an enzyme capable of using a di- or tri-phosphate ribonucleotide or deoxyribonucleotide as a substrate for its addition to a miRNA. Moreover, in specific embodiments, it involves using a modified di- or tri-phosphate ribonucleotide, which is added to the 3' end of a miRNA. Enzymes capable of adding such nucleotides include, but are not limited to, poly(A) polymerase, terminal transferase, and polynucleotide phosphorylase. Terminal transferase catalyzes the addition of nucleotides to the 3 ? terminus of a nucleic acid. Polynucleotide phosphorylase can polymerize nucleotide diphosphates without the need for a primer. 2. Labels
  • Labels on miRNA or miRNA probes may be colorimetric (includes visible and UV spectrum, including fluorescent), luminescent, enzymatic, or positron emitting (including radioactive).
  • the label may be detected directly or indirectly.
  • Radioactive labels include 125 I, 32 P, 33 P, and 35 S.
  • Examples of enzymatic labels include alkaline phosphatase, luciferase, horseradish peroxidase, and ⁇ -galactosidase. Labels can also be proteins with luminescent properties, e.g., green fluorescent protein and phicoerythrin.
  • the colorimetric and fluorescent labels contemplated for use as conjugates include, but are not limited to, Alexa Fluor dyes, BODIPY dyes, such as BODIPY FL; Cascade Blue; Cascade Yellow; coumarin and its derivatives, such as 7-amino-4-methylcoumarin, aminocoumarin and hydroxycoumarin; cyanine dyes, such as Cy3 and Cy5; eosins and erythrosins; fluorescein and its derivatives, such as fluorescein isothiocyanate; macrocyclic chelates of lanthanide ions, such as Quantum DyeTM; Marina Blue; Oregon Green; rhodamine dyes, such as rhodamine red, tetramethylrhodamine and rhodamine 6G; Texas Red; fluorescent energy transfer dyes, such as thiazole orange-ethidium heterodimer; and TOTAB.
  • Alexa Fluor dyes such as BODIPY FL
  • Cascade Blue Ca
  • dyes include, but are not limited to, those identified above and the following: Alexa Fluor 350, Alexa Fluor 405, Alexa Fluor 430, Alexa Fluor 488, Alexa Fluor 500. Alexa Fluor 514, Alexa Fluor 532, Alexa Fluor 546, Alexa Fluor 555, Alexa Fluor 568, Alexa Fluor 594, Alexa Fluor 610, Alexa Fluor 633, Alexa Fluor 647, Alexa Fluor 660, Alexa Fluor 680, Alexa Fluor 700, and, Alexa Fluor 750; amine-reactive BODIPY dyes, such as BODIPY 493/503, BODIPY 530/550, BODIPY 558/568, BODIPY 564/570, BODIPY 576/589, BODIPY 581/591, BODIPY 630/650, BODIPY 650/655, BODIPY FL, BODIPY R6G, BODIPY TMR, and, BODIP
  • fluorescently labeled ribonucleotides are available from Molecular Probes, and these include, Alexa Fluor 488-5-UTP, Fluorescein- 12-UTP, BODIPY FL-14-UTP, BODIPY TMR-14-UTP, Tetramethylrhodamine-6-UTP, Alexa Fluor 546-14- UTP, Texas Red-5-UTP, and BODIPY TR-14-UTP.
  • Other fluorescent ribonucleotides are available from Amersham Biosciences, such as Cy3-UTP and Cy5-UTP.
  • Examples of fluorescently labeled deoxyribonucleotides include Dinitrophenyl (DNP)-11-dUTP, Cascade Blue-7-dUTP, Alexa Fluor 488-5-dUTP, Fluorescein- 12-dUTP, Oregon Green 488-5-dUTP, BODIPY FL-14-dUTP, Rhodamine Green-5-dUTP, Alexa Fluor 532-5-dUTP, BODIPY TMR-14-dUTP, Tetramethylrhodamine-6-dUTP, Alexa Fluor 546-14- dUTP, Alexa Fluor 568-5-dUTP, Texas Red-12-dUTP, Texas Red-5-dUTP, BODIPY TR- 14- dUTP, Alexa Fluor 594-5-dUTP, BODIPY 630/650-14-dUTP, BODIPY 650/665-14-dUTP; Alexa Fluor 488-7-OBEA-dCTP, Alexa Fluor 546
  • nucleic acids may be labeled with two different labels.
  • fluorescence resonance energy transfer FRET may be employed in methods of the invention (e.g., Kleinmeier et al, 2002; Emptage, 2001; Didenko, 2001, each incorporated by reference).
  • the label may not be detectable per se, but indirectly detectable or allowing for the isolation or separation of the targeted nucleic acid.
  • the label could be biotin, digoxigenin, polyvalent cations, chelator groups and the other ligands, include ligands for an antibody.
  • a number of techniques for visualizing or detecting labeled nucleic acids are readily available. Such techniques include, microscopy, arrays, Fluorometry, Light cyclers or other real time PCR machines, FACS analysis, scintillation counters, Phosphoimagers, Geiger counters, MRI, CAT, antibody-based detection methods (Westerns, immunofluorescence, immunohistochemistry), histochemical techniques, HPLC (Griffey et al, 1997), spectroscopy, capillary gel electrophoresis (Cummins et al., 1996), spectroscopy; mass spectroscopy; radiological techniques; and mass balance techniques.
  • FRET fluorescent resonance energy transfer
  • the present invention concerns the preparation and use of miRNA arrays or miRNA probe arrays.
  • the arrays can be ordered macroarrays or microarrays of nucleic acid molecules (probes) that are fully or nearly complementary or identical to a plurality of miRNA molecules or precursor miRNA molecules and are positioned on a support or support material in a spatially separated organization.
  • Macroarrays are typically a support ⁇ e.g., sheets of nitrocellulose or nylon) upon which probes have been spotted.
  • Microarrays position the nucleic acid probes more densely such that up to 10,000 nucleic acid molecules can be fit into a region typically 1 to 4 square centimeters.
  • Microarrays can be fabricated by spotting nucleic acid molecules, e.g., genes, oligonucleotides, etc., onto substrates or fabricating oligonucleotide sequences in situ on a substrate. Spotted or fabricated nucleic acid molecules can be applied in a high density matrix pattern of up to about 30 non-identical nucleic acid molecules per square centimeter or higher, e.g. up to about 100 or even 1000 per square centimeter. Microarrays typically use coated glass as the solid support, in contrast to the nitrocellulose-based material of filter arrays. By having an ordered array of miRNA- complementing nucleic acid samples, the position of each sample can be tracked and linked to the original sample.
  • nucleic acid molecules e.g., genes, oligonucleotides, etc.
  • array devices in which a plurality of distinct nucleic acid probes are stably associated with the surface of a solid support are known to those of skill in the art.
  • Useful substrates or supports for arrays include nylon, glass, metal, plastic, and silicon.
  • Such arrays may vary in a number of different ways, including average probe length, sequence or types of probes, nature of bond between the probe and the array surface, e.g. covalent or non-covalent, and the like.
  • the labeling and screening methods of the present invention and the arrays are not limited in its utility with respect to any parameter except that the probes detect miRNA; consequently, methods and compositions may be used with a variety of different types of miRNA arrays.
  • the arrays can be high density arrays, such that they contain 2, 20, 25, 50, 80, 100 or more different probes. It is contemplated that they may contain 1000, 16,000, 65,000, 250,000 or 1,000,000 or more different probes.
  • the probes can be directed to targets in one or more different organisms or cell types.
  • the oligonucleotide probes range from 5 to 50, 5 to 45, 10 to 40, 9 to 34, or 15 to 40 nucleotides in length in some embodiments. In certain embodiments, the oligonucleotide probes are 5, 10, 15, 20 to 20, 25, 30, 35, 40 nucleotides in length including all integers and ranges there between.
  • each different probe sequence in the array are generally known. Moreover, the large number of different probes can occupy a relatively small area providing a high density array having a probe density of generally greater than about 60, 100, 600, 1000, 5,000, 10,000, 40,000, 100,000, or 400,000 different oligonucleotide probes per cm 2 .
  • the surface area of the array can be about or less than about 1, 1.6, 2, 3, 4, 5, 6, 7, 8, 9, or 10 cm 2 .
  • miRNA of a wide variety of samples can be analyzed using the arrays, index of miRNA probes, or array technology described herein and known to the skilled artisan. While endogenous miRNA is contemplated for use with compositions and methods of the invention, recombinant miRNA - including nucleic acids that are complementary or identical to endogenous miRNA or precursor miRNA - can also be handled and analyzed as described herein. Samples may be biological samples, in which case, they can be from biopsy, fine needle aspirates, exfoliates, scrappings, blood, tissue, organs, or any sample containing or constituting biological cells of interest.
  • samples may be, but are not limited to, fresh, frozen, fixed, formalin fixed, paraffin embedded, or formalin fixed and paraffin embedded.
  • the sample may not be a biological sample, but be a chemical mixture, such as a cell-free reaction mixture (which may contain one or more biological enzymes).
  • the population of target nucleic acids is contacted with the array or probes under hybridization conditions, where such conditions can be adjusted, as desired, to provide for an optimum level of specificity in view of the particular assay being performed.
  • Suitable hybridization conditions are well known to those of skill in the art and reviewed in Sambrook et al. (2001) and WO 95/21944. Of particular interest in many embodiments is the use of stringent conditions during hybridization. Stringent conditions are known to those of skill in the art.
  • a single array or set of probes may be contacted with multiple samples.
  • the samples may be labeled with different labels to distinguish the samples.
  • a single array can be contacted with a tumor tissue sample labeled with Cy3, and normal tissue sample labeled with Cy5. Differences between the samples for particular miRNAs corresponding to probes on the array can be readily ascertained and quantified.
  • the small surface area of the array permits uniform hybridization conditions, such as temperature regulation and salt content. Moreover, because of the small area occupied by the high density arrays, hybridization may be carried out in extremely small fluid volumes ⁇ e.g., about 250 ⁇ l or less, including volumes of about or less than about 5, 10, 25, 50, 60, 70, 80 ,90, 100 ⁇ l, or any range derivable therein). In small volumes, hybridization may proceed very rapidly.
  • Arrays of the invention can be used to detect differences between two samples. Specifically contemplated applications include identifying and/or quantifying differences between miRNA from a sample that is normal and from a sample that is not normal or contains abnormal components, between a cancerous condition and a non-cancerous condition, or between two differently treated samples. Also, miRNA may be compared between a sample believed to be susceptible to a particular disease or condition and one believed to be not susceptible or resistant to that disease or condition. A sample that is not normal is one exhibiting phenotypic trait(s) of a disease or condition or one believed to be not normal with respect to that disease or condition. Phenotypic traits include symptoms of or susceptibility to a disease or condition of which a component is or may or may not be genetic or caused by a hyperproliferative or neoplastic cell or cells.
  • An array comprises a solid support with nucleic acid probes attached to the support.
  • Arrays typically comprise a plurality of different nucleic acid probes that are coupled to a surface of a substrate in different, known locations.
  • These arrays also described as “microarrays” or colloquially “chips” have been generally described in the art, for example, U.S. Pat. Nos. 5,143,854, 5,445,934, 5,744,305, 5,677,195, 6,040,193, 5,424,186 and Fodor et al, Science, 251:767-777 (1991), each of which is incorporated by reference in its entirety for all purposes.
  • arrays may generally be produced using mechanical synthesis methods or light directed synthesis methods which incorporate a combination of photolithographic methods and solid phase synthesis methods. Techniques for the synthesis of these arrays using mechanical synthesis methods are described in, e.g., U.S. Patent 5,384,261, incorporated herein by reference in its entirety for all purposes. Although a planar array surface is used in certain aspects, the array may be fabricated on a surface of virtually any shape or even a multiplicity of surfaces. Arrays may be nucleic acids on beads, gels, polymeric surfaces, fibers such as fiber optics, glass or any other appropriate substrate, see U.S.
  • Arrays may be packaged in such a manner as to allow for diagnostics or other manipulation of an all inclusive device, see for example, U.S. Patents 5,856,174 and 5,922,591 incorporated in their entirety by reference for all purposes. See also U.S. patent application Ser. No. 09/545,207, filed Apr. 7, 2000 for additional information concerning arrays, their manufacture, and their characteristics, which is incorporated by reference in its entirety for all purposes.
  • Particularly arrays can be used to evaluate samples with respect to diseases or conditions that include, but are not limited to pre-cancerous cervical lesion or cervical cancer.
  • Cancers that may be evaluated by methods and compositions of the invention include cancer cells particularly from the uterus or cervix, but may also include cells and cancer cells from the bladder, blood, bone, bone marrow, brain, breast, colon, esophagus, gastrointestine, gum, head, kidney, liver, lung, nasopharynx, neck, ovary, prostate, skin, stomach, testis, or tongue.
  • miRNA can be evaluated in precancers, such as metaplasia, dysplasia, and hyperplasia.
  • the invention can be used to evaluate differences between stages of disease, such as between hyperplasia, neoplasia, pre-cancer and cancer, or between a primary tumor and a metastasized tumor.
  • samples that have differences in the activity of certain pathways may also be compared. These pathways include those involving the following factors: antibody response, apoptosis, calcium/NFAT signaling, cell cycle, cell migration, cell adhesion, cell division, cytokines and cytokine receptors, drug metabolism, growth factors and growth factor receptors, inflammatory response, insulin signaling, NFK-B signaling, angiogenesis, adipogenesis, cell adhesion, viral infection, bacterial infection, senescence, motility, glucose transport, stress response, oxidation, aging, telomere extension, telomere shortening, neural transmission, blood clotting, stem cell differentiation, G-Protein Coupled Receptor (GPCR) signaling, and p53 misregulation.
  • GPCR G-Protein Coupled Receptor
  • Cellular pathways that may be profiled also include but are not limited to the following: any adhesion or motility pathway including but not limited to those involving cyclic AMP, protein kinase A, G-protein couple receptors, adenylyl cyclase, L-selectin, E- selectin, PECAM, VCAM-I, ⁇ -actinin, paxillin, cadherins, AKT, integrin- ⁇ , integrin- ⁇ , RAF-I, ERK, PI-3 kinase, vinculin, matrix metalloproteinases, Rho GTPases, p85, trefoil factors, profilin, FAK, MAP kinase, Ras, caveolin, calpain-1, calpain-2, epidermal growth factor receptor, ICAM-I, ICAM-2, cofilin, actin, gelsolin, RhoA, RACl, myosin light chain kinase, platelet-derived growth
  • nucleic acids molecules of the invention can be employed in diagnostic and therapeutic methods with respect to any of the above pathways or factors.
  • a miRNA may be differentially expressed with respect to one or more of the above pathways or factors.
  • the pathways or cellular elements involved or effected by HPV infection can be assessed, evaulated, and/or monitored, e.g., hTERT, E6TP1, MCM7, Bak, and PDZ domain-containing proteins such as Scribble, h-Dlg, MAGI-I, MAGI-3, and MUPPl.
  • Phenotypic traits also include characteristics such as susceptibility or receptivity to particular drugs or therapeutic treatments (drug efficacy), and risk of drug toxicity. Samples that differ in these phenotypic traits may also be evaluated using the arrays and methods described.
  • miRNA profiles may be generated to evaluate and correlate those profiles with pharmacokinetics.
  • miRNA profiles may be created and evaluated for patient tumor and blood samples prior to the patient's being treated or during treatment to determine if there are miRNAs whose expression correlates with the outcome of the patient. Identification of differential miRNAs can lead to a diagnostic assay involving them that can be used to evaluate tumor and/or blood samples to determine what drug regimen the patient should be provided. In addition, it can be used to identify or select patients suitable for a particular clinical trial. If a miRNA profile is determined to be correlated with drug efficacy or drug toxicity that may be relevant to whether that patient is an appropriate patient for receiving the drug or for a particular dosage of the drug.
  • samples from patients with a variety of diseases can be evaluated to determine if different diseases can be identified based on blood miRNA levels.
  • a diagnostic assay can be created based on the profiles that doctors can use to identify individuals with a disease or who are at risk to develop a disease.
  • treatments can be designed based on miRNA profiling. Examples of such methods and compositions are described in the U.S. Provisional Patent Application serial number 60/683,736 entitled “Methods and Compositions Involving miRNA and miRNA Inhibitor Molecules” filed on May 23, 2005, which is hereby incorporated by reference in its entirety.
  • RNAs include, but are not limited to, nucleic amplification, polymerase chain reaction, quantitative PCR, RT-PCR, in situ hybridization, Northern hybridization, hybridization protection assay (HPA)(GenProbe), branched DNA (bDNA) assay (Chiron), rolling circle amplification (RCA), single molecule hybridization detection (US Genomics), Invader assay (ThirdWave Technologies), and/or Bridge Litigation Assay (Genaco).
  • HPA hybridization protection assay
  • bDNA branched DNA
  • RCA rolling circle amplification
  • US Genomics Invader assay
  • GTWave Technologies Invader assay
  • Bridge Litigation Assay Geneaco
  • Methods of the invention include reducing or eliminating activity of one or more miRNAs in a cell comprising introducing into a cell an miRNA inhibitor; or supplying or enhancing the activity of one or more miRNAs in a cell.
  • the present invention also concerns inducing certain cellular characteristics by providing to a cell a particular nucleic acid, such as a specific synthetic miRNA molecule or a synthetic miRNA inhibitor molecule.
  • the miRNA molecule or miRNA inhibitor need not be synthetic. They may have a sequence that is identical to a naturally occurring miRNA or they may not have any design modifications, hi certain embodiments, the miRNA molecule and/or an miRNA inhibitor are synthetic, as discussed herein.
  • the particular nucleic acid molecule provided to the cell is understood to correspond to a particular miRNA in the cell, and thus, the miRNA in the cell is referred to as the "corresponding miRNA.”
  • the corresponding miRNA will be understood to be the induced miRNA. It is contemplated, however, that the miRNA molecule introduced into a cell is not a mature miRNA but is capable of becoming a mature miRNA under the appropriate physiological conditions, hi cases in which a particular corresponding miRNA is being inhibited by a miRNA inhibitor, the particular miRNA will be referred to as the targeted miRNA. It is contemplated that multiple corresponding miRNAs may be involved.
  • more than one miRNA molecule is introduced into a cell.
  • more than one miRNA inhibitor is introduced into a cell.
  • a combination of miRNA molecule(s) and miRNA inhibitor(s) may be introduced into a cell.
  • Methods include identifying a cell or patient in need of inducing those cellular characteristics. Also, it will be understood that an amount of a synthetic nucleic acid that is provided to a cell or organism is an "effective amount,” which refers to an amount needed to achieve a desired goal, such as inducing a particular cellular characteristic(s).
  • the methods include providing or introducing to a cell a nucleic acid molecule corresponding to a mature miRNA in the cell in an amount effective to achieve a desired physiological result.
  • methods can involve providing synthetic or nonsynthetic miRNA molecules. It is contemplated that in these embodiments, methods may or may not be limited to providing only one or more synthetic miRNA molecules or only on or more nonsynthetic miRNA molecules. Thus, in certain embodiments, methods may involve providing both synthetic and nonsynthetic miRNA molecules. In this situation, a cell or cells are most likely provided a synthetic miRNA molecule corresponding to a particular miRNA and a nonsynthetic miRNA molecule corresponding to a different miRNA. Furthermore, any method including a list of miRNAs using Markush group language may be articulated without the Markush group language and a disjunctive article (i.e., or) instead, and vice versa.
  • a method for reducing or inhibiting cell proliferation in a cell comprising introducing into or providing to the cell an effective amount of (i) an miRNA inhibitor molecule or (ii) a synthetic or nonsynthetic miRNA molecule that corresponds to an miRNA sequence.
  • the methods involves introducing into the cell an effective amount of (i) an miRNA inhibitor molecule having a 5' to 3' sequence that is at least 90% complementary to all or part of the 5' to 3' sequence of one or more mature miRNA of Table 1.
  • Certain embodiments of the invention include methods of treating a pre-cancerous cervical lesion or a cancerous cervical condition.
  • the method comprises contacting a cervical cell with one or more nucleic acid, synthetic miRNA, or miRNA comprising at least one nucleic acid segment having all or a portion of a miRNA sequence.
  • the segment may be 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30 or more nucleotides or nucleotide analog, including all integers there between.
  • An aspect of the invention includes the modulation of a miRNA or a mRNA within a target cell, such as a cervical cell.
  • an endogenous gene, miRNA or mRNA is modulated in the cell.
  • the nucleic acid sequence comprises at least one segment that is at least 70, 75, 80, 85, 90, 95, or 100% identical in nucleic acid sequence to one or more miRNA sequence listed in Table 1.
  • Modulation of the expression or processing of an endogenous gene, miRNA, or mRNA can be through modulation of the processing of an mRNA, such processing including transcription, transportation and/or translation with in a cell. Modulation may also be effected by the inhibition or enhancement of miRNA activity with a cell, tissue, or organ. Such processing may effect the expression of an encoded product or the stability of the mRNA.
  • a nucleic acid sequence can comprise a modified nucleic acid sequence.
  • the cervical cell is a cervical cancer cell.
  • Methods of the invention can further comprise administering a second therapy, such as chemotherapy, radiotherapy, surgery, or immunotherapy.
  • the nucleic acid can be transcribed from a nucleic acid vector, such as a plasmid vector or a viral vector.
  • Methods of treating a pre-cancerous or cancerous cervical condition include contacting or administering to a cervical cell one or more nucleic acid comprising a miRNA sequence, wherein expression of an endogenous miRNA is modulated in the cervical cell; where the miRNA sequence is at least 70, 75, 80, 85% or more identical to one or more of the sequences identified in Table 1.
  • the activity of those miRNAs indicated as having increased expression in a pre-cancerous or cancerous tissue is decreased.
  • the miRNA activity of those miRNA indicated as having decreased expression in a pre-cancerous or cancerous tissue is increased.
  • one or more miRNA sequence may include or comprise a modified nucleobase or nucleic acid sequence.
  • the methods may further comprise administering a second therapy.
  • the second therapy can be, but is not limited to chemotherapy, radiotherapy, surgery, or immunotherapy.
  • one or more miRNA are transcribed from a nucleic acid vector, such as a plasmid or viral vector.
  • a subject is administered: one or more nucleic acid possessing a function of an miRNA having a nucleic acid segment having at least 80, 85, 90, 95, 97, 98, 99, or 100% nucleic acid sequence identity to those miRNA decreased or down-regulated in a disease or condition to be treated.
  • a subject is administered: one or more miRNA inhibitors having a nucleic acid segment having at least 80, 85, 90, 95, 97, 98, 99, or 100% nucleic acid sequence identity to those miRNA increased or up-regulated in a disease or condition to be treated.
  • Synthetic nucleic acids can be administered to the subject or patient using modes of administration that are well known to those of skill in the art, particularly for therapeutic applications. It is particularly contemplated that a patient is human or any other mammal or animal.
  • a cell or other biological matter such as an organism (including patients) can be provided an miRNA or miRNA molecule corresponding to a particular miRNA by administering to the cell or organism a nucleic acid molecule that functions as the corresponding miRNA once inside the cell.
  • the form of the molecule provided to the cell may not be the form that acts as an miRNA once inside the cell.
  • biological matter is provided a synthetic miRNA or a nonsynthetic miRNA, such as one that becomes processed into a mature and active miRNA once it has access to the cell's miRNA processing machinery.
  • the miRNA molecule provided to the biological matter is not a mature miRNA molecule but a nucleic acid molecule that can be processed into the mature miRNA once it is accessible to miRNA processing machinery.
  • nonsynthetic in the context of miRNA means that the miRNA is not “synthetic,” as defined herein.
  • the use of corresponding nonsynthetic miRNAs is also considered an aspect of the invention, and vice versa.
  • the methods involve reducing cell viability comprising introducing into or providing to the cell an effective amount of (i) an miRNA inhibitor molecule or (ii) a synthetic or nonsynthetic miRNA molecule that corresponds to an miRNA sequence.
  • Methods for inducing apoptosis have a number of therapeutic applications including, but not limited to, the treatment of pre-cancer or cancer.
  • the present invention also concerns using miRNA compositions to treat diseases or conditions or to prepare therapeutics for the treatment of diseases or conditions. It is contemplated that 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 more miRNA (or any range derivable therein) may be used for these embodiments, hi certain embodiments, methods involve one or more miRNA inhibitors and/or an miRNA molecules corresponding to any of these miRNAs, particularly for the treatment or prevention of cancer. Cancer includes, but is not limited to, malignant cancers, tumors, metastatic cancers, unresectable cancers, chemo- and/or radiation-resistant cancers, and terminal cancers.
  • methods also include targeting an miRNA to modulate in a cell or organism.
  • targeting an miRNA to modulate or “targeting an miRNA” means a nucleic acid of the invention will be employed so as to modulate the selected miRNA.
  • the modulation is achieved with a synthetic or non-synthetic miRNA that corresponds to the targeted miRNA, which effectively provides the targeted miRNA to the cell or organism (positive modulation).
  • the modulation is achieved with an miRNA inhibitor, which effectively inhibits the targeted miRNA in the cell or organism (negative modulation).
  • the miRNA targeted to be modulated is an miRNA that affects a disease, condition, or pathway.
  • the miRNA is targeted because a treatment can be provided by negative modulation of the targeted miRNA.
  • the miRNA is targeted because a treatment can be provided by positive modulation of the targeted miRNA.
  • a further step of administering the selected miRNA modulator to a cell, tissue, organ, or organism in need of treatment related to modulation of the targeted miRNA or in need of the physiological or biological results discussed herein (such as with respect to a particular cellular pathway or result, such as a decrease in cell viability). Consequently, in some methods of the invention there is a step of identifying a patient in need of treatment that can be provided by the miRNA modulator(s). It is contemplated that an effective amount of an miRNA modulator can be administered in some embodiments.
  • a therapeutic benefit refers to an improvement in the one or more conditions or symptoms associated with a disease or condition or an improvement in the prognosis, duration, or status with respect to the disease. It is contemplated that a therapeutic benefit includes, but is not limited to, a decrease in pain, a decrease in morbidity, a decrease in a severity or duration of a symptom.
  • a therapeutic benefit can be inhibition of tumor growth, prevention of metastasis, reduction in number of metastases, inhibition of cancer cell proliferation, inhibition of cancer cell proliferation, induction of cell death in cancer cells, inhibition of angiogenesis near cancer cells, induction of apoptosis of cancer cells, reduction in pain, reduction in risk of recurrence, induction of chemo- or radiosensitivity in cancer cells, prolongation of life, palliation of symptoms related to the condition, and/or delay of death directly or indirectly related to a cancer.
  • the miRNA compositions may be provided as part of a therapy to a patient, in conjunction with traditional therapies or preventative agents.
  • any method discussed in the context of therapy may be applied as a preventative measure, particularly in a patient identified to be potentially in need of the therapy or at risk of the condition or disease for which a therapy is needed.
  • methods of the invention concern employing one or more nucleic acids corresponding to an miRNA and a therapeutic drug.
  • the nucleic acid can enhance the effect or efficacy of the drug, reduce any side effects or toxicity, modify its bioavailability, and/or decrease the dosage or frequency needed.
  • the therapeutic drug is a cancer therapeutic. Consequently, in some embodiments, there is a method of treating cancer in a patient comprising administering to the patient the cancer therapeutic and an effective amount of at least one miRNA molecule that improves the efficacy of the cancer therapeutic or protects non-cancer cells.
  • Cancer therapies also include a variety of combination therapies with both chemical and radiation based treatments.
  • Combination chemotherapies include but are not limited to, for example, bevacizumab, cisplatin (CDDP), carboplatin, EGFR inhibitors (gefitinib and cetuximab), procarbazine, mechlorethamine, cyclophosphamide, camptothecin, COX-2 inhibitors ⁇ e.g., celecoxib) ifosfamide, melphalan, chlorambucil, busulfan, nitrosurea, dactinomycin, daunorubicin, doxorubicin (adriamycin), bleomycin, plicomycin, mitomycin, etoposide (VP 16), tamoxifen, raloxifene, estrogen receptor binding agents, taxol, taxotere, gemcitabien, navelbine, farnesyl-protein transferase inhibitors, transplatinum, 5-fluorouracil, vincristin, vinblastin and methotrex
  • inhibitors of miRNAs can be given to achieve the opposite effect as compared to when nucleic acid molecules corresponding to the mature miRNA are given.
  • nucleic acid molecules corresponding to the mature miRNA can be given to achieve the opposite effect as compared to when inhibitors of the miRNA are given.
  • miRNA molecules that increase cell proliferation can be provided to cells to increase proliferation or inhibitors of such molecules can be provided to cells to decrease cell proliferation.
  • the present invention contemplates these embodiments in the context of the different physiological effects observed with the different miRNA molecules and miRNA inhibitors disclosed herein.
  • Methods of the invention are generally contemplated to include providing or introducing one or more different nucleic acid or mimetic molecules corresponding to one or more different miRNA molecules.
  • nucleic acid molecules may be provided or introduced: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, or any range derivable therein. This also applies to the number of different miRNA molecules that
  • compositions described herein may be comprised in a kit.
  • reagents for isolating miRNA, labeling miRNA, and/or evaluating a miRNA population using an array, nucleic acid amplification, and/or hybridization can be included in a kit, as well reagents for preparation of samples from cervical samples or other sample that have been, may have been, exposed to, or suspected of being infected with HPV.
  • the kit may further include reagents for creating or synthesizing miRNA probes.
  • the kits will thus comprise, in suitable container means, an enzyme for labeling the miRNA by incorporating labeled nucleotide or unlabeled nucleotides that are subsequently labeled.
  • the kit can include amplification reagents.
  • the kit may include various supports, such as glass, nylon, polymeric beads, and the like, and/or reagents for coupling any probes and/or target nucleic acids. It may also include one or more buffers, such as reaction buffer, labeling buffer, washing buffer, or a hybridization buffer, compounds for preparing the miRNA probes, and components for isolating miRNA.
  • buffers such as reaction buffer, labeling buffer, washing buffer, or a hybridization buffer, compounds for preparing the miRNA probes, and components for isolating miRNA.
  • Other kits of the invention may include components for making a nucleic acid array comprising miRNA, and thus, may include, for example, a solid support.
  • Kits for implementing methods of the invention described herein are specifically contemplated.
  • kits for preparing miRNA for multi-labeling and kits for preparing miRNA probes and/or miRNA arrays.
  • kit comprise, in suitable container means, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more of the following: (1) poly(A) polymerase; (2) unmodified nucleotides (G, A, T, C, and/or U); (3) a modified nucleotide (labeled or unlabeled); (4) poly(A) polymerase buffer; (5) at least one microfilter; (6) label that can be attached to a nucleotide; (7) at least one miRNA probe; (8) reaction buffer; (9) a miRNA array or components for making such an array; (10) acetic acid; (11) alcohol; and (12) solutions for preparing, isolating, enriching, and purifying miRNAs or miRNA probes or arrays.
  • Other reagents include those generally used
  • kits of the invention include an array containing miRNA probes, as described in the application.
  • An array may have probes corresponding to all known miRNAs of an organism or a particular tissue or organ in particular conditions, or to a subset of such probes.
  • the subset of probes on arrays of the invention may be or include those identified as relevant to a particular diagnostic, therapeutic, or prognostic application.
  • the array may contain one or more probes that is indicative or suggestive of (1) a disease or condition, (2) susceptibility or resistance to a particular drug or treatment; (3) susceptibility to toxicity from a drug or substance; (4) the stage of development or severity of a disease or condition (prognosis); and (5) genetic predisposition to a disease or condition.
  • kits embodiment including an array
  • nucleic acid molecules that contain or can be used to amplify a sequence that is a variant of, identical to or complementary to all or part of any of SEQ ID NOS: 1-562.
  • a kit or array of the invention can contain one or more probes for the miRNAs identified by SEQ ID NOS: 1-562. Any nucleic acid discussed above may be implemented as part of a kit.
  • kits may be packaged either in aqueous media or in lyophilized form.
  • the container means of the kits will generally include at least one vial, test tube, flask, bottle, syringe or other container, into which a component may be placed, and preferably, suitably aliquotted. Where there is more than one component in the kit (labeling reagent and label may be packaged together), the kit also will generally contain a second, third or other additional container into which the additional components may be separately placed. However, various combinations of components may be comprised in a vial.
  • the kits of the present invention also will typically include a means for containing the nucleic acids, and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow molded plastic containers into which the desired vials are retained.
  • the liquid solution is an aqueous solution, with a sterile aqueous solution being particularly preferred.
  • the components of the kit may be provided as dried powder(s).
  • the powder can be reconstituted by the addition of a suitable solvent. It is envisioned that the solvent may also be provided in another container means.
  • labeling dyes are provided as a dried power.
  • kits of the invention 10-20 30, 40, 50, 60, 70, 80, 90, 100, 120, 120, 130, 140, 150, 160, 170, 180, 190, 200, 300, 400, 500, 600, 700, 800, 900, 1000 ⁇ g or at least or at most those amounts of dried dye are provided in kits of the invention.
  • the dye may then be resuspended in any suitable solvent, such as DMSO.
  • the container means will generally include at least one vial, test tube, flask, bottle, syringe and/or other container means, into which the nucleic acid formulations are placed, preferably, suitably allocated.
  • the kits may also comprise a second container means for containing a sterile, pharmaceutically acceptable buffer and/or other diluent.
  • kits may also include components that facilitate isolation of the labeled miRNA. It may also include components that preserve or maintain the miRNA or that protect against its degradation. Such components may be RNAse-free or protect against RNAses. Such kits generally will comprise, in suitable means, distinct containers for each individual reagent or solution.
  • Kits of the invention may also include one or more of the following: Control RNA; nuclease- free water; RNase-free containers, such as 1.5 ml tubes; RNase-free elution tubes; PEG or dextran; ethanol; acetic acid; sodium acetate; ammonium acetate; guanidinium; detergent; nucleic acid size marker; RNase-free tube tips; and RNase or DNase inhibitors.
  • kits of the invention are embodiments of kits of the invention. Such kits, however, are not limited to the particular items identified above and may include any reagent used for the manipulation or characterization of miRNA.
  • RNA samples from nine squamous cell carcinomas (Ca) and nine normal adjacent regions (NAT) of the uterine cervix from the same patients and three squamous intraepithelial lesions (SIL) of the cervix were purchased from ProteoGenex (Culver City, CA, USA). Total RNA was extracted from these tissue samples using the mzWanaTM miRNA Isolation Kit (Ambion; Austin, TX, USA) according to the manufacturer's protocol. Additional samples used in the study included purified total RNA from sixteen normal cervical tissue specimens (NCX) (FirstChoice® Human Cervix Total RNA, Ambion).
  • the inventors first evaluated miRNA expression in normal cervical tissue samples. miRNA expression profiling was performed as previously described (Shingara et al, 2005) except that the miRNA fractions recovered from 20 ⁇ g total RNA were labeled with Cy5 (GE Healthcare Life Sciences; Piscataway, NJ, USA). To isolate miRNA fractions, total RNA samples were fractionated and purified using the flashPAGETM fractionator and reagents (catalog# - AMI 3100; Ambion) according to the manufacturer's recommendations. Labeled miRNAs were hybridized to mirVana. miRNA Bioarrays Vl (Ambion) according to the manufacturer's instructions.
  • the arrays contained 377 individual miRNA probes, including 281 human miRNAs from the mirBase Sequence Database (microrna.sanger.ac.uk) (Griffiths- Jones et al., 2006), 33 new human miRNAs (Ambi-miRs) and 63 mouse or rat miRNAs from the mirBase Sequence Database.
  • VSN variance stabilization method
  • the minimum observable threshold was determined by examining the foreground minus background median intensities for 'EMPTY' spots. The minimum threshold was defined as 5% symmetric trimmed mean plus 2 standard deviations across all 'EMPTY' spots on an individual array.
  • the inventors first characterized miRNA expression profiles from 16 normal cervix specimens (Table 5). Approximately 200 miRNAs were detected above background signal (Mean (NCX) > 2.87). These included 171 human miRNAs, 17 miRNAs previously identified in mouse or rat, and 15 new human miRNAs (Ambi-miR-7027, -7029, -7039, - 7058, -7068-1, -7070, -7075, -7076, -7079, -7081, -7083, -7085, -7100, -7101, and -7105).
  • miRNA expression in 33 different human tissues was determined using the same microarray platform described above.
  • the human reference set consisted of FirstChoice® Total RNA samples (Ambion) isolated from 33 distinct tissues: adipose, adrenal, aorta, bladder, bone marrow, brain, breast, colon, duodenum, esophagus, fallopian tube, heart, ileum, jejunum, kidney, liver, lung, lymph node, muscle, ovary, pancreas, pituitary, placenta, prostate, small intestine, spleen, stomach, testis, thymus, thyroid, trachea, uterus, and vena cava.
  • miRNAs potentially relevant to carcinogenesis frequently exhibit differential expression in cancer versus normal samples collected from the same tissue type.
  • miRNAs with differential expression in normal and cancerous samples may be used in the diagnosis of pre-cancerous and cancerous lesions.
  • the inventors used the microarray platform described in Example 2 to compare global miRNA expression in nine cervical squamous cell carcinomas and nine paired normal adjacent tissue samples from the same patients. The results are shown below in Table 7. On average, 244 miRNAs were detected above background (>2.32) in normal adjacent tissues from cancer patients and 208 miRNAs were detected above background (>2.64) in cervical tumors.
  • Table 7 Normalized Array Data for miRNA Expression in Nine Cervical Tumor Tissue Samples (CaI -Ca9) and Nine Cervical Normal Adjacent Tissue Samples (NATl -NAT-9). TV, threshold value. miRNAs > TV, miRNAs greater than threshold value. %, Percentage of miRNAs reater than threshold value.
  • these miRNAs 53 were down-regulated ( ⁇ H(NAT-Ca)>0.69), and 21 were up-regulated ( ⁇ H(NAT-Ca) ⁇ - 0.69) by at least 2-fold in Ca samples, when compared to their expression in NAT samples (Fold Change NAT vs Ca ⁇ 2.0).
  • miRNAs (hsa-miR-1, -133a, -204, -218, -143, -368, - 99a, -100, -195, -376a, and ambi-miR-7029) were down-regulated by more than 5-fold in Ca samples versus NAT samples ( ⁇ H(NAT-Ca)>1.6).
  • Three miRNAs (hsa-miR-205, -141, and - 182) were up-regulated more than 5-fold in Ca samples versus NAT samples ( ⁇ H(NAT-Ca) ⁇ - 1.6).
  • 70 were down-regulated ( ⁇ H(NCX-Ca)>0.69) and 27 were up-regulated ( ⁇ H(NCX-Ca) ⁇ -0.69), by at least 2-fold in Ca samples, when compared to their expression in NCX samples (Fold Change NCX vs Ca _ ⁇ .0).
  • 103 human miRNAs were significantly differentially expressed between the cancer samples (Ca) and both the normal cervix samples (NCX) and the normal adjacent tissue samples (NAT). Thirty human miRNAs were significantly differentially expressed between Ca and NCX samples that were not significantly differentially expressed between Ca and NAT samples. Four distinct human miRNAs were significantly differentially expressed between Ca and NAT samples that were not significantly differentially expressed between Ca and NCX samples.
  • the data described in this example demonstrate that miRNA expression analysis can be used to distinguish a cancerous cervical tissue sample from a normal, non-cancerous cervical tissue sample.
  • the inventors have shown that specific miRNAs are differentially expressed (up-regulated or down-regulated) in cancerous cervical tissue as opposed to normal cervical tissues. Comparing the expression levels of these specific miRNAs in a cervical tissue sample that is suspected of being cancerous with their expression levels in a reference non-cancerous cervical tissue sample can indicate whether or not the suspect tissue sample is cancerous.
  • Microarray profiling of cervical squamous cell carcinomas and normal primary tissues using the microarrays described in Example 2, identified 103 mis-regulated (differentially expressed) miRNAs between the cervical cancer samples and both normal sample types (Table 8).
  • the inventors identified 23 miRNAs that were differentially expressed, with at least a 5-fold change, between the Ca and NCX or NAT samples and a p- value ⁇ 0.0001.
  • These 23 miRNAs whose expression is significantly affected in cervical cancer represent novel biomarkers and therapeutic targets for cervical squamous cell carcinoma. Their identity and associated array data are summarized in Table 9.
  • the inventors performed real-time qRT-PCR reactions for 12 of the top 23 miRNAs in Table 9 with very distinct expression patterns between cervical squamous cell carcinomas (Ca) and normal adjacent tissues (NAT) or normal cervix (NCX) samples (hsa-miR-1, -15b, -133a, -143, -205, -21, -204, -195, -100, -99a, -368, and -183) and one miRNA with no significant change (hsa-miR-16), which was used for normalization.
  • qRT-PCR reactions were performed using TaqMan® MicroRNA Assays (Applied Biosystems; Foster City, CA, USA) according to the manufacturer's instructions.
  • Reactions included 15 ng of total RNA per reaction and were incubated in the 7900HT Fast Real-Time PCR System (Applied Biosystems). Initial data analysis was done using the 7500 Fast System SDS 2.3 software. The inventors analyzed the 34 samples previously profiled (16 NCX and 9 paired Ca and NAT samples).
  • the inventors sought to determine if the top 23 misregulated miRNAs in cervical cancer (Table 9) could distinguish precancerous cervical squamous epithelial lesions from normal cervical samples.
  • the inventors evaluated miRNA expression in three cervical squamous intraepithelial lesions (SILs) (also known as cervical intraepithelial neoplasias, CINs) (Table 10).
  • SILs cervical squamous intraepithelial lesions
  • CINs cervical intraepithelial neoplasias
  • Pathological analysis classified two lesions (SILl, SIL2) as low grade (LSIL) and one (SIL3) as high grade (HSIL).
  • Table 10 Histology and HPV (Human Papillomavirus) Status for Three Cervical Cancer Precursor Lesions. CIN, cervical intraepithelial neoplasia.
  • RNA expression analysis was carried out as described in Example 2, except that 7 ⁇ g of total RNA from each SIL sample were hybridized to the arrays.
  • the inventors used the non parametric "rank product" method (Breitling et al, 2004) for identifying differentially expressed genes.
  • Rank Product uses fold-change to assign ranks for all features in a dataset, providing reliable and sensitive results. Importantly, RP does not depend on the variance calculation for every gene and therefore is powerful when a small number of replicates is available. The robustness of this method has been demonstrated in previous studies where the list of genes generated from RP and a Student's T-test at a false discovery rate-corrected p-value of 5% were found in agreement (Breitling et al, 2004).
  • miRNAs down-regulated in SILs were among the miRNAs down-regulated by at least 2-fold in Ca versus NCX (miR-1, -133a, -187, -204, -145, -143, -125a, -376a, -505, -100, and -99a). It is also noteworthy that miR-1, which is the most significantly down-regulated miRNA in the cervical cancers of our series, was also the most down-regulated miRNA in SILs.
  • miRNAs up-regulated in SILs compared to the normal cervix samples
  • eight miRNAs (miR-141, -205, -146a, -200b, -182, -203, -21, and -31) were also up-regulated by at least 2-fold in the Ca samples compared to NCX samples.
  • SIL cervical squamous intraepithelial lesion
  • CIN cervical intraepithelial neoplasia
  • Specific miRNAs are differentially expressed (up-regulated or down-regulated) in cervical SILs (CINs). Comparing the expression levels of these specific miRNAs in a cervical tissue sample that is suspected of being pre-cancerous with their expression levels in a reference non-cancerous cervical tissue sample can indicate whether or not the suspect tissue is precancerous.
  • the inventors determined miRNA expression profiles of five cervical cancer-derived cell lines using the same microarray platform described in Example 2. To compare miRNA expression profiles, miRNA expression data from the five cervical cancer cell lines (CL) were normalized together with the data described in earlier examples from the nine pairs of cervical tumors (Ca) and normal adjacent tissue specimens (NAT) and from the sixteen normal cervix samples (NCX) (Table 12).
  • NCX normal cervix
  • Ca cervical cancer
  • NAT normal adjacent tissue
  • miRNA array expression analysis microRNA-containing fractions were recovered from 10 ⁇ g of total RNA isolated from each RNA sample, using the FlashPage fractionation system (Ambion, Inc.). Purified small RNAs were enzymatically biotinylated at the 3 '-termini and hybridized to a custom-made Affymetrix based microRNA array, Asuragen's Disco v Arrays V.I containing probes for 14,215 verified and candidate miRNAs from a number of sources including Sanger miRBase v9.2 database (http:microrna.sanger.ac.uk/sequences/) and published reports. Hybridization, washing, staining, imaging, and signal extraction were performed as recommended by the manufacturer, except that the 2OX GeneChip® Eukaryotic Hybridization Control cocktail was omitted.
  • Array data processing The signal processing implemented for the Ambion miRCHIP is a multi-step process involving probe specific signal detection calls, background estimate and correction, constant variance stabilization and either array scaling or global normalization.
  • miRNA processing and analysis see Davison et al. (2006). For more details, see supplementary information.
  • Probe specific signal detection calls Each probe on an array is assayed for detection based on a Wilcoxon rank-sum test of the miRNA probe signal compared to the distribution of signals from GC-content matched anti-genomic probes. If the resulting p- value for the probes is ⁇ 0.06 then it is considered "Detected above background.” Probes with p- values > 0.06 have insufficient signal to discriminate from the background and are thus considered not detected.
  • VSN is a global normalization process that stabilizes the variance evenly across the entire range of expression. Differences in VSN transformed expression are denoted by Log2 diff and were used for all subsequent data analyses. Differences in normalized expression values between samples (Log 2 diff) can be transformed to a generalized fold change via exponentiation base 2. These values will exhibit a compression for small differences in expression.
  • DiscovArrayTM miRNA Expression Profiling Service Asuragen.
  • the DiscovArrayTM microarray contains probes for 994 miRNAs from Sanger miRBase V9.2 (Griffiths- Jones et al, 2006) (467 human miRNAs, 234 rat miRNAs, 293 mouse miRNAs) and 12,894 predicted human miRNAs (see world wide web page at asuragen.com/services/discovarray.htmt).
  • One hundred forty four miRNAs were significantly differentially expressed between NCX and Ca samples by at least 2-fold (Table 14). Of those, 136 were down-regulated (Log2 Diff Ca vs NCX ⁇ -1.0) and 8 were up-regulated (Log2 Diff Ca vs NCX ⁇ l.O) in Ca samples compared to NCX samples. Sixty-three (63) miRNAs were down-regulated in Ca samples by more than 5-fold (Log2 Diff Ca vs NCX ⁇ -2.3) with a p-value ⁇ 0.0001. Seventeen of those miRNAs were down-regulated by over 10-fold in Ca samples (Log2 Diff Ca vs NCX ⁇ -3.3).
  • miRNA array expression analysis miRNA expression in six cervical cancer specimens (CaI -4, Ca6, and Ca8) and six normal cervix samples (NCXl -3, NCXlO, NCXl 3, and NCXl 6) were further evaluated using the Human miRNA Microarray (V2) (Agilent Technologies, Inc.; Santa Clara, CA, USA).
  • the Human miRNA Microarray contains probes for 723 human and 76 human viral microRNAs from the Sanger database v.10.1. Samples for miRNA profiling studies were processed by Asuragen Services (Asuragen, Inc.; Austin, TX, USA).
  • RNA from each sample 200 ng was dephosphorylated and the pCp-Cy3 labeling molecule was ligated to the 3' end of the RNA molecules.
  • the labeled RNA was purified using a Bio-Spin P-6 column (Bio-Rad Laboratories Inc.; Hercules, CA, USA). Array hybridization, washing, staining, imaging, and signal extraction were performed according to Agilent's recommended procedures.
  • miRNA array signal processing The signal processing implemented for the Agilent miRNA array is a multi-step process involving probe specific signal detection calls, background correction, and global normalization. For each probe, the contribution of signal due to background was estimated and removed by the Agilent Feature Extraction software as part of the data file output. Similarly, detection calls were based on the Agilent Feature Extraction software. Arrays within a specific analysis experiment were normalized together according to the VSN method described by Huber et al, 2002.
  • the “Total Gene Signal” is the total probe signal multiplied by the number of probes per gene and is calculated after the background effects have been accounted for.
  • the “Total Gene Error” is the square root of the square of the total probe error multiplied by the number of probes per gene.
  • the “Total Probe Error” is the robust average for each replicated probe multiplied by the total number of probe replicates.
  • the "Detection Call” is a binary number that indicates if the gene was detected on the miRNA microarray. Probes detected at least once across all samples in the experiment were considered for statistical analysis.
  • VSN Variance Stabilization Normalization
  • Generalized Iog 2 transformed The post-normalized data scale is reported as generalized log 2 data.
  • the distribution of microarray data is typically log normal (i.e, it tends to follow a normal distribution pattern after log transformation).
  • a normal distributed data is amendable to classical statistical treatments, including t-tests and one-way or two-way ANOVA.
  • a total of 382 human miRNAs were expressed above background level in the normal cervix samples, representing 53% of the miRNAs present on the arrays.
  • a total of 337 miRNAs were expressed above background level in the tumor samples, representing 46.6% of the miRNAs present on the arrays.
  • a total of 202 human miRNAs were significantly differentially expressed between NCX and Ca samples (Table 15). Among these, 129 were down-regulated (Log2 diff (NCX vs Ca) >1) and 41 were up-regulated (Log2 diff (NCX vs Ca) ⁇ 1) by more than 2-fold in Ca samples compared to NCX samples. Thirty-three miRNAs were down-regulated in Ca samples by more than 5-fold (Log2 diff (NCX vs Ca) >2.3) with a Student t-test p-value ⁇ 0.001.
  • NCX Six Cervical Cancer Samples (Ca) and Six Normal Cervix Tissue Samples (NCX) Following Expression Analysis on Agilent Human miRNA Microarrays. Positive Log2Diff (NCX vs Ca) values represent miRNAs down- regulated in cancer samples

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Hospice & Palliative Care (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The present invention concerns methods and compositions for identifying a miRNA profile for a particular condition, such as cervical disease, and using the profile in assessing the condition of a patient.

Description

DESCRIPTION
MICRORNAS DIFFERENTIALLY EXPRESSED IN CERVICAL CANCER
AND USES THEREOF
BACKGROUND OF THE INVENTION
[0001] This application claims priority to U.S. Provisional Application serial number 60/972,646 filed September 14, 2007, which is incorporated herein by reference in its entirety.
I. FIELD OF THE INVENTION
[0002] The present invention relates generally to the field of molecular biology. More particularly, it concerns methods and compositions involving microRNA (miRNAs) molecules. Certain aspects of the invention include applications for miRNAs in diagnostics, therapeutics, and prognostics of cervical cancer.
II. BACKGROUND
[0003] In 2001, several groups used a cloning method to isolate and identify a large group of "microRNAs" (miRNAs) from C. elegans, Drosophila, and humans (Lagos-Quintana et al, 2001; Lau et al, 2001; Lee and Ambros, 2001). Several hundred miRNAs have been identified in plants and animals — including humans — which do not appear to have endogenous siRNAs. Thus, while similar to siRNAs, miRNAs are distinct.
[0004] miRNAs thus far observed have been approximately 21-22 nucleotides in length, and they arise from longer precursors, which are transcribed from non-protein-encoding genes. See review of Caπϊngton et al (2003). The precursors form structures that fold back on themselves in self-complementary regions; they are then processed by the nuclease Dicer (in animals) or DCLl (in plants) to generate the short double-stranded miRNA. One of the miRNA strands is incorporated into a complex of proteins and miRNA called the RNA- induced silencing complex (RISC). The miRNA guides the RISC complex to a target mRNA, which is then cleaved or translationally silenced, depending on the degree of sequence complementarity of the miRNA to its target mRNA. Currently, it is believed that perfect or nearly perfect complementarity leads to mRNA degradation, as is most commonly observed in plants. In contrast, imperfect base pairing, as is primarily found in animals, leads to translational silencing. However, recent data suggest additional complexity (Bagga et al, 2005; Lim et al, 2005), and mechanisms of gene silencing by miRNAs remain under intense study.
[0005] Recent studies have shown that expression levels of numerous miRNAs are associated with various cancers (reviewed in Esquela-Kerscher and Slack, 2006; Calin and Croce, 2006). miRNAs have also been implicated in regulating cell growth and cell and tissue differentiation - cellular processes that are associated with the development of cancer.
[0006] Cervical cancer is the second most common cause of cancer in women worldwide (Pisani et al, 2002; Parkin et al, 2005). About 470,000 new cases are diagnosed and approximately 230,000 women die of cervical cancer every year (Pisani et al, 1999). While the majority (-80%) of these new cases and deaths occur in developing countries, it is estimated that approximately 3,700 women will die from invasive cervical cancer in the United States in 2007 (Jemal et al, 2007).
[0007] Epidemiological and molecular studies have demonstrated that human papillomaviruses (HPVs) are the etiological agents of the vast majority (99.7%) of cervical cancers and their intraepithelial precursors (Pisani, et al, 2002; Parkin et al, 2005; zur Hausen, 2002). Approximately, fifty HPV types infect the anogenital tract including the uterine cervix (Pisani, et al, 2002; Parkin et al, 2005). "High-risk" HPV types are associated with intraepithelial lesions that can progress into invasive carcinomas. Among these, HPV 16 and HPV 18 are associated with 50% and 20%, respectively, of cervical squamous cell carcinomas (Bosch and de Sanjose, 2002; zur Hausen, 2002; Clifford et al, 2003). Other high-risk HPV types (31, 33, 35, 39, and 45 among others) are found in 20-30% of cervical cancers (Bosch and de Sanjose, 2002; zur Hausen, 2002; Clifford et al, 2003). High-risk HPV types are also associated with 25% of head and neck tumors, in particular tumors of the mouth, tonsils, esophagus and larynx (Gillison et al, 2000; Rose et al, 2006).
[0008] Cytological examination of cervical smears with Papanicolaou staining (Pap smears) is the screening method universally accepted for early detection of cervical cancer and its precursors. Pap smear screening has been very effective in reducing cervical cancer incidence and mortality. Abnormal pap smear results include mild dysplasias referred to as low-grade squamous intraepithelial lesions (LSIL) and moderate to severe dysplasias referred to as high- grade squamous intraepithelial lesions (HSIL). A Pap smear with LSIL or HSIL indicates a need for further examination and possible treatment. In one study (ALTS Group, 2000), over 80% of LSILs were found to be positive for HPV; however, almost 50% of LSILs will regress to normal. HSILs are generally considered to be pre-cancerous in nature and indicate more aggressive treatment. In addition, as many as 3 million Pap smears are classified as inconclusive in the U.S. every year, and cervical cancer is still a significant public health problem. Pap smear screening is imperfect due, in part, to sampling and staining errors, resulting in false-negatives. It is estimated that -17% of cervical cancers develop in women with previous false-negative Pap smears. It is also estimated that approximately 9% of cervical cancers develop in women with previous true-negative Pap tests.
[0009] As an adjunct to cytology screening, The United States Food and Drug Administration has approved a nucleic acid hybridization test for HPV for all women over 30 years old and as triage for women with inconclusive Pap smears. However, several problems associated with this assay include variable assay sensitivity, the inability to genotype certain strains of HPV, and the inability to detect about half of the high risk HPV types associated with cervical cancer (Begeron et al, 2000; de Cremoux et al, 2006).
[0010] A need exists for additional diagnostic assays that can assess the condition of cervical tissue in general and accurately distinguish pre-cancerous or cancerous tissue from non-cancerous tissue in particular.
SUMMARY OF THE INVENTION
[0011] Embodiments of the invention include compositions and methods of identifying miRNAs that are differentially expressed or mis-regulated in various states of normal, precancerous, cancerous, and/or abnormal tissues, including but not limited to normal cervical tissue, pre-cancerous diseased cervical tissue {e.g., low-grade squamous intraepithelial lesions (LSIL) and high-grade squamous intraepithelial lesions (HSIL)), squamous cell carcinoma, cervical cancer {e.g., cervical squamous cell carcinoma), squamous cell carcinoma of the vulva and vagina, penile intraepithelial lesions, anal cancer, cancers of the digestive tract, oral cancers, a subset of head and neck cancers, and tonsil cancer. In certain aspects, the cancers are related to or a result of HPV infection. In other aspects a subject known or at risk of infection with HPV can be monitored. In still a further aspect, a subject can be monitored periodically, e.g. rm'RNA assays can be conducted in conjunction with pap smears or noninvasive procedures or procedures with limited invasiveness {e.g., swabbing or flushing and the like). The method of sampling is not intended to be a limiting factor and is at the discretion of the care giver. Further, the invention describes a method for diagnosing normal, pre-cancerous, and cancerous tissues or cells, including but not limited to cervical squamous intraepithelial lesions and cervical cancer based on determining levels (increased or decreased) of selected miRNAs in patient-derived samples. Samples may be obtained and/or analyzed from patients, including but not limited to patients having or suspected of having pre-cancerous cervical lesion or cervical cancer, or a patient suspected of having one or the other condition.
[0012] The term "miRNA" is used according to its ordinary and plain meaning and refers to a microRNA molecule found in eukaryotes that is involved in RNA-based gene regulation. See, e.g., Carrington et al, 2003, which is hereby incorporated by reference. Individual miRNAs in a variety of organisms have been identified, sequenced, and given names. Names of miRNAs and their sequences related to the present invention are provided herein.
[0013] It is understood that a "synthetic nucleic acid" of the invention means that the nucleic acid does not have a chemical structure or sequence of a naturally occurring nucleic acid or is made by non-natural processes. Consequently, it will be understood that the term "synthetic miRNA" refers to a "synthetic nucleic acid" that functions as or inhibits the functions of an miRNA, at least in part, in a cell or under physiological conditions.
[0014] While many of the embodiments of the invention involve synthetic miRNAs or synthetic nucleic acids, in some embodiments of the invention, the nucleic acid molecule(s) need not be "synthetic." hi certain embodiments, a non-synthetic miRNA employed in methods and compositions of the invention may have all or part of the sequence and structure of a naturally occurring miRNA precursor or the mature miRNA. For example, non-synthetic miRNAs used in methods and compositions of the invention may not have one or more modified nucleotides or nucleotide analogs. In these embodiments, the non-synthetic miRNA may or may not be recombinantly produced. In particular embodiments, the nucleic acid in methods and/or compositions of the invention is specifically a synthetic miRNA; though in other embodiments, the invention specifically involves a non-synthetic miRNA and not a synthetic miRNA. Any embodiments discussed with respect to the use of synthetic miRNAs can be applied with respect to non-synthetic miRNAs, and vice versa.
[0015] It will be understood that the term "naturally occurring" refers to something found in an organism without any intervention by a person; it could refer to a naturally-occurring wildtype or mutant molecule. In some embodiments a synthetic miRNA molecule does not have the sequence of a naturally occurring miRNA molecule, hi other embodiments, a synthetic miRNA molecule may have the sequence of a naturally occurring miRNA molecule, but the chemical structure of the molecule, particularly in the part unrelated specifically to the precise sequence (non-sequence chemical structure) differs from chemical structure of the naturally occurring miRNA molecule with that sequence. In some cases, the synthetic miRNA has both a sequence and non-sequence chemical structure that are not found in a naturally-occurring miRNA. Moreover, the sequence of the synthetic molecules will identify which miRNA is effectively being provided or inhibited; the endogenous miRNA will be referred to as the "corresponding miRNA." Corresponding miRNA sequences that can be used in the context of the invention include, but are not limited to, all or a portion of those sequences in SEQ ID NOs: 1-562, as well as any other of miRNA sequence, miRNA precursor sequence, or any complementary sequence. In some embodiments, the sequence is or is derived from or contains all or part of a sequence identified in Table 1 below to target a particular miRNA (or set of miRNAs) or mRNA (or set of mRNA).
Figure imgf000006_0001
Figure imgf000007_0001
Figure imgf000008_0001
Figure imgf000009_0001
Figure imgf000010_0001
Figure imgf000011_0001
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0001
(1) Bentwich et al, 2005; (2) miRBase; (3) Griffiths- Jones et al, 2006; (4) Berezikov et al., 2005; (5) Xie et al., 2005
[0016] In some embodiments, it may be useful to know whether a cell expresses a particular miRNA endogenously or whether such expression is affected under particular conditions or a particular disease state. Thus, in some embodiments of the invention, methods include assaying a cell or a sample containing a cell for the presence of one or more miRNA. In other aspects, a sample may comprise RNA or nucleic acid isolated from a tissue or cells of a patient or reference. Consequently, in some embodiments, methods include a step of generating an miRNA profile for a sample. The term "miRNA profile" refers to a set of data regarding the expression pattern for a plurality of miRNAs (e.g., one or more miRNA from Table 1) in the sample; it is contemplated that the miRNA profile can be obtained using a set of miRNAs, using for example nucleic acid amplification or hybridization techniques well known to one of ordinary skill in the art. It is contemplate the any one or subset of the miRNA listed in this application can be included or excluded from the claimed invention.
[0017] In some embodiments of the invention, an miRNA profile is generated by steps that include: (a) labeling miRNA in the sample; (b) hybridizing miRNA to a number of probes, or amplifying a number of miRNA, and (c) determining miRNA hybridization to the probes or detection of miRNA amplification products, wherein an miRNA profile is generated. See U.S. Provisional Patent Application 60/575,743 and the U.S. Provisional Patent Application 60/649,584, and U.S. Patent Application Serial No. 11/141,707, all of which are hereby incorporated by reference.
[0018] Methods of the invention involve diagnosing a patient based on an miRNA expression or expression profile. In certain embodiments, the presence, absence, elevation, or reduction in the level of expression of a particular miRNA or set of miRNA in a cell is correlated with a disease state compared to the expression level of that miRNA or set of miRNAs in a normal cell. This correlation allows for diagnostic methods to be carried out when the expression level of an miRNA is measured in a biological sample being assessed and then compared to the expression level of a normal cell. It is specifically contemplated that miRNA profiles for patients, particularly those suspected of having a particular disease or condition such as a pre-cancerous cervical lesion or a cervical cancer, can be generated by evaluating any miRNA or sets of the miRNAs discussed in this application. The miRNA profile that is generated from the patient will be one that provides information regarding the particular disease or condition. In many embodiments, the miRNA profile is generated using miRNA hybridization or amplification, (e.g., array hybridization or RT-PCR). In certain aspects, a miRNA profile can be used in conjunction with other diagnostic tests, such as serum protein profiles.
[0019] Embodiments of the invention include methods for diagnosing, prognosing, and/or assessing a condition in a patient comprising measuring an expression profile of one or more miRNAs in a sample from the patient. The difference in the expression profile in the sample from the patient and a reference expression profile, such as an expression profile from a normal or non-pathologic sample or a reference (e.g., a reference sample or a digital reference), is indicative of a pathologic, disease, or cancerous condition. An miRNA or probe set comprising or identifying a segment of a corresponding miRNA can include all or part of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 ,13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 125, 150, 175, 200, 250, 300, 350, 400, 500, 550 to 562 or any integer or range derivable there between, of a miRNA or a probe or its complement listed in Table 1. It is contemplate the any one or subset of the miRNA listed in this application can be included or excluded from the claimed invention.
[0020] In certain aspects, the methods for diagnosing a condition in a patient comprise measuring an expression profile of one or more miRNAs, from Table 1, in a sample from the patient, wherein the difference between the expression profile in the sample from the patient and an expression profile of a normal sample or a reference is indicative of a pathological condition.
[0021] In a further aspect, the methods for diagnosing a condition in a patient comprises measuring an expression profile of one or more miRNAs in a sample from the patient, wherein the difference between the expression profile of the sample from the patient and an expression profile of a reference is indicative of a cervical tissue disease, or condition; wherein the miRNA is one or more of hsa-let-7a, hsa-let-7b, hsa-let-7c, hsa-let-7i, hsa-miR- 100, hsa-miR-101, hsa-miR-106a, hsa-miR-125a, hsa-miR-125b, hsa-miR-126-AS, hsa-miR- 127, hsa-miR-130a, hsa-miR-134, hsa-miR-142-3p, hsa-miR-143, hsa-miR-145, hsa-miR- 148b, hsa-miR-149, hsa-miR-151, hsa-miR-152, hsa-miR-154, hsa-miR-15a, hsa-miR-15b, hsa-miR-16, hsa-miR-17-5p, hsa-miR-181a, hsa-miR-185, hsa-miR-186, hsa-miR-187, hsa- miR-192, hsa-miR-193a, hsa-miR-195, hsa-miR-196a, hsa-miR-196b, hsa-miR-199a, hsa- miR-199a-AS, hsa-miR-199b, hsa-miR-19a, hsa-miR-20a, hsa-miR-203, hsa-miR-205, hsa- miR-21, hsa-miR-214, hsa-miR-215, hsa-miR-22, hsa-miR-221, hsa-miR-223, hsa-miR-23b, hsa-miR-24, hsa-miR-25, hsa-miR-26a, hsa-miR-26b, hsa-miR-299-5p, hsa-miR-29b, hsa- miR-29c, hsa-miR-302c-AS, hsa-miR-30a-3p, hsa-miR-30a-5p, hsa-miR-30b, hsa-miR-30c, hsa-miR-30e-3p, hsa-miR-30e-5p, hsa-miR-31, hsa-miR-320, hsa-miR-324-3p, hsa-miR-335, hsa-miR-338, hsa-miR-342, hsa-miR-34a, hsa-miR-361, hsa-miR-365, hsa-miR-367, hsa- miR-368, hsa-miR-370, hsa-miR-374, hsa-miR-376a, hsa-miR-379, hsa-miR-381, hsa-miR- 423, hsa-miR-424, hsa-miR-450, hsa-miR-7, hsa-miR-93, hsa-miR-95, hsa-miR-96, hsa-miR- 99a, hsa-miR-99b, ambi-miR-7027, ambi-miR-7029, hsa-miR-509, hsa-miR-193b, ambi-miR- 7039, hsa-miR-526b, hsa-miR-498, hsa-miR-452, ambi-miR-7062, ambi-miR-7070, hsa-miR- 497, ambi-miR-7079, ambi-miR-7083, ambi-miR-7085, hsa-miR-503, or ambi-miR-7101 or complements thereof.
[0022] In a particular aspect, hsa-miR-205, hsa-miR-196b, hsa-miR-203, hsa-miR-503, hsa- miR-196a, hsa-miR-99a, hsa-miR-187, ambi-miR-7083 and/or ambi-miR-7101, or any combination or complement thereof being expressed or having an increased expression or upregulated expression when compared with expression in a non-cervical reference sample, is indicative of cervical tissue. In another aspect, a decrease in expression or down-regulation of hsa-miR-7 and/or hsa-miR-215 is also indicative of a cervical tissue.
[0023] In still further aspects of the invention, the methods for diagnosing a condition in a patient comprise measuring an expression profile of one or more miRNAs in a sample or a cervix sample from the patient believed to be pre-cancerous or cancerous or contain precancerous or cancerous cells, wherein the difference between the expression profile in the sample from the patient and an expression profile of normal adjacent tissue (NAT) or a reference tissue is indicative of a disease or condition; wherein the miRNA is one or more miRNA listed in Table 1. It is contemplate the any one or subset of the miRNA listed in this application can be included or excluded from the claimed invention.
[0024] In still further aspects of the invention, the methods for diagnosing a condition in a patient comprise measuring an expression profile of one or more miRNAs in a cervix sample from a patient believed to be precancerous or cancerous, or contain precancerous or cancerous cells, or from a patient suspected of having or at risk of developing a pre-cancerous or cancerous condition, wherein the difference between the expression profile in the sample from the patient and an expression profile of normal cervix (NCX) or normal adjacent tissue (NAT) or a reference tissue is indicative of a disease or condition; wherein the miRNA is one or more of hsa-let-7a, hsa-let-7b, hsa-let-7c, hsa-let-7d, hsa-let-7e, hsa-let-7f, hsa-let-7g, hsa-let-7i, hsa-miR-1, hsa-miR-100, hsa-miR-101, hsa-miR-106a, hsa-miR-106b, hsa-miR-10a, hsa- miR-lOb, hsa-miR-124a, hsa-miR-125a, hsa-miR-125b, hsa-miR-126, hsa-miR-126-AS, hsa- miR-127, hsa-miR-130a, hsa-miR-130b, hsa-miR-133a, hsa-miR-134, hsa-miR-135b, hsa- miR-139, hsa-miR-140, hsa-miR-141, hsa-miR-142-5p, hsa-miR-143, hsa-miR-145, hsa-miR- 146a, hsa-miR-149, hsa-miR-150, hsa-miR-152, hsa-miR-153, hsa-miR-154, hsa-miR-155, hsa-miR-15b, hsa-miR-16, hsa-miR-17-5p, hsa-miR-18a, hsa-miR-181a, hsa-miR-181b, hsa- miR-182, hsa-miR-183, hsa-miR-185, hsa-miR-186, hsa-miR-187, hsa-miR-189, hsa-miR- 190, hsa-miR-195, hsa-miR-196a, hsa-miR-196b, hsa-miR-199a, hsa-miR-199a- AS, hsa-miR- 199b, hsa-miR-20a, hsa-miR-200a, hsa-miR-200b, hsa-miR-200c, hsa-miR-203, hsa-miR- 204, hsa-miR-205, hsa-miR-21, hsa-miR-210, hsa-miR-214, hsa-miR-215, hsa-miR-218, hsa- miR-223, hsa-miR-224, hsa-miR-23b, hsa-miR-24, hsa-miR-25, hsa-miR-26a, hsa-miR-26b, hsa-miR-27b, hsa-miR-28, hsa-miR-296, hsa-miR-299-5p, hsa-miR-29a, hsa-miR-29b, hsa- miR-29c, hsa-miR-302d, hsa-miR-30a-3p, hsa-miR-30a-5ρ, hsa-miR-30b, hsa-miR-30d, hsa- miR-31, hsa-miR-320, hsa-miR-324-3p, hsa-miR-325, hsa-miR-328, hsa-miR-330, hsa-miR- 335, hsa-miR-339, hsa-miR-361, hsa-miR-365, hsa-miR-368, hsa-miR-370, hsa-miR-373-AS, hsa-miR-376a, hsa-miR-377, hsa-miR-379, hsa-miR-381, hsa-miR-382, hsa-miR-423, hsa- miR-424, hsa-miR-429, hsa-miR-450, hsa-miR-92, hsa-miR-93, hsa-miR-95, hsa-miR-98, hsa-miR-99a, hsa-miR-99b, hsa-miR-520d, hsa-miR-518b, ambi-miR-7029, hsa-miR-491, hsa-miR-515-5p, hsa-miR-498, ambi-miR-7062, hsa-miR-432, hsa-miR-495, ambi-miR-7066, ambi-miR-7068-1, ambi-miR-7070, hsa-miR-492, hsa-miR-497, ambi-miR-7074, ambi-miR- 7075, hsa-miR-501, ambi-miR-7079, ambi-miR-7083, ambi-miR-7085, hsa-miR-500, hsa- miR-513, hsa-miR-505, ambi-miR-7100, or ambi-miR-7101, hsa-miR-133b, hsa-miR-455, hsa-asg-5021_stl, hsa-asg-13254_stl, hsa-asg-14176_stl, hsa-miR-487b, hsa-miR-411, hsa- miR-574, hsa-miR-542-5p, hsa-asg-5617_stl, hsa-asg-14172_stl, hsa-asg-13304_st2, hsa- asg-13297_stl, hsa-miR157_st2, hsa-cand317_stl, hsa-miR-329, hsa-asg-10202_st2, hsa- miR-369-5p, hsa-asg-13284_stl, hsa-asg-9687_stl, hsa-miR-433, hsa-miR-565, hsa-asg- 562_stl, hsa-asg-279_st2, hsa-asg-8411_st2, hsa-asg-7472_st2, hsa-asg-13279_stl, hsa-miR- 487a, hsa-cand206_stl, hsa-cand345_stl, hsa-asg-9696_stl, hsa-miR-485-3p, hsa-asg- 13166_st2, hsa-miR-594_st2, hsa-miR-299-3p, hsa-asg-924_stl, hsa-miR-539, hsa-asg- 10883_stl, hsa-miR-585, hsa-miR-493-5p, hsa-asg-12964_st2, hsa-asg-4557_st2, hsa-asg- 10674_stl, hsa-asg-14230_stl, hsa-asg-9681_stl, hsa-miR-628, hsa-asg-13237_stl, hsa-asg- 13230_st2, hsa-miR-493-3ρ, hsa-miR-654, hsa-asg-2919_stl, hsa-asg-8067_st2, hsa-asg- 11199_st2, hsa-asg-12346_st2, hsa-asg-11883_stl, hsa-miR102_st2, hsa-asg-2027_stl, hsa- asg-3711_st2, hsa-asg-3145_stl, hsa-asg-7465_st2, hsa-asg-3376_stl, hsa-cand283_stl, hsa- asg-2301_st2, hsa-cand207_stl, hsa-miR-598, hsa-asg-10278_st2, hsa-miR-18b, hsa-asg- 11181_stl, hsa-asg-7023_st2, hsa-asg-3597_st2, hsa-asg-5304_stl, hsa-asg-11688_stl, hsa- cand720_stl, hsa-asg-13308_st2, hsa-asg-3038_st2, hsa-asg-13966_st2, hsa-asg-13189_stl, hsa-asg-11938_stl, hsa-asg-5740_st2, hsa-asg-8477_stl, hsa-asg-12325_st2, hsa-asg- 12356_stl, hsa-asg-6758_stl, hsa-asg-522_stl, hsa-asg-4564_st2, hsa-asg-6951_st2, hsa-asg- 9920_stl, hsa-asg-13613_st2, hsa-miR-184, hsa-miR-503, hsa-miR-485-5p, hsa-miR-494, hsa-miR-504, hsa-miR-211, hsa-miR-99b, hsa-miR-499, hsa-miR-422b, hsa-miR-338, hsa- miR-422a, hsa-miR-331, hsa-miR-489, hsa-miR-324-5p, hsa-miR-151, hsa-miR-17-3p, hsa- miR-340, hsa-miR-194, hsa-let-7d*, hsa-let-7e*, hsa-miR-100*, hsa-miR-101*, hsa-miR- 106b*, hsa-miR-lOb*, hsa-miR-125a-5p, hsa-miR-125b-2*, hsa-miR-126*, hsa-miR-127-3p, hsa-miR-129-3p, hsa-miR-135a, hsa-miR-136, hsa-miR-136*, hsa-miR-139-5p, hsa-miR-140- 3ρ, hsa-miR-140-5p, hsa-miR-141*, hsa-miR-142-3p, hsa-miR-143*, hsa-miR-144, hsa-miR- 144*, hsa-miR-145*, hsa-miR-146b-5p, hsa-miR-151-3p, hsa-miR-154*, hsa-miR-15b*, hsa- miR-17, hsa-miR-181c, hsa-miR-181d, hsa-miR-183*, hsa-miR-193a-5p, hsa-miR-195*, hsa- miR-197, hsa-miR-199a-5p, hsa-miR-199b-3p, hsa-miR-199b-5p, hsa-miR-20a*, hsa-miR- 21*, hsa-miR-212, hsa-miR-214*, hsa-miR-24-1*, hsa-miR-26b*, hsa-miR-28-3p, hsa-miR- 28-5p, hsa-miR-29a*, hsa-miR-29b-l*, hsa-miR-29b-2*, hsa-miR-29c*, hsa-miR-30a, hsa- miR-30a*, hsa-miR-30b*, hsa-miR-30c-2*, hsa-miR-31*, hsa-miR-32, hsa-miR-330-3ρ, hsa- miR-337-3p, hsa-miR-337-5p, hsa-miR-345, hsa-miR-361-5p, hsa-miR-374a, hsa-miR-374b, hsa-miR-374b*, hsa-miR-375, hsa-miR-376b, hsa-miR-376c, hsa-miR-377*, hsa-miR-410, hsa-miR-424*, hsa-miR-425, hsa-miR-431*, hsa-miR-450a, hsa-miR-451, hsa-miR-455-3p, hsa-miR-455-5p, hsa-miR-483-3p, hsa-miR-486-5p, hsa-miR-488, hsa-miR-490-3p, hsa-miR- 499-5p, hsa-miR-501-5p, hsa-miR-505*, hsa-miR-512-3ρ, hsa-miR-513c, hsa-miR-517*, hsa- miR-542-3p, hsa-miR-543, hsa-miR-574-3p, hsa-miR-602, hsa-miR-628-5p, hsa-miR-629*, hsa-miR-630, hsa-miR-650, hsa-miR-654-3p, hsa-miR-654-5p, hsa-miR-655, hsa-miR-656, hsa-miR-744, hsa-miR-766, hsa-miR-768-3p, hsa-miR-873, hsa-miR-885-5p, hsa-miR-886- 3p, hsa-miR-886-5p, hsa-miR-889, hsa-miR-93*, hsa-miR-940, hsa-miR-944, hsa-miR-96, hsa-miR-99a*, hsa-miR-99b*, or complements thereof.
[0025] In certain aspects, the reduced expression or down regulation of hsa-miR-1, hsa- miR-100, hsa-miR-125b, hsa-miR-125b-2*, hsa-miR-133a, hsa-miR-133b, hsa-miR-134, hsa- miR-135a, hsa-miR-139-5p, hsa-miR-136*, hsa-miR-143*, hsa-miR-154, hsa-miR-204, hsa- miR-211, hsa-miR-218, hsa-miR-224, hsa-miR-143, hsa-miR-145, hsa-miR-145*, hsa-miR- 299-3p, hsa-miR-299-5ρ, hsa-miR-368, hsa-miR-99a, hsa-miR-195, hsa-miR-337-3ρ, hsa- miR-337-5p, hsa-miR-375, hsa-miR-376a,hsa-miR-376c, hsa-miR-379, hsa-miR-381, hsa- miR-410, hsa-miR-411, hsa-miR-424, hsa-miR-451, hsa-miR-455-5p, hsa-miR-495, hsa-miR- 497, hsa-miR-517*, hsa-miR-654-3ρ, hsa-miR-885-5p, hsa-miR-886-5p, hsa-miR-99a*, ambi-miR-7101 and/or ambi-miR-7029 or a complement thereof relative to normal adjacent tissue or a normal cervix tissue reference is indicative of a cancerous condition or tissue, hi other aspects, the increased or upregulation of hsa-miR-21, hsa-miR-21*hsa-miR-31, hsa- miR-31*, hsa-miR-135b, hsa-miR-141, hsa-miR-182, hsa-miR-183, hsa-miR-203, hsa-miR- 205, hsa-miR-224, hsa-miR-141, hsa-miR-944, hsa-miR-96, and/or hsa-miR-182 or complement thereof relative to normal adjacent tissue or a normal cervix tissue reference is indicative of a cancerous condition or cancerous tissue.
[0026] In a still further aspect, the reduced expression or down regulation of hsa-miR-1, hsa-miR-133a, hsa-miR-204, hsa-miR-218, hsa-miR-143, hsa-miR-368, hsa-miR-99a, hsa- miR-100, hsa-miR-195, hsa-miR-376a, hsa-miR-424, hsa-miR-497, hsa-miR-299_5p, hsa- miR-154, hsa-miR-134, ambi-miR-7101 and/or ambi-miR-7029 or complement thereof relative to normal cervix tissue is indicative of a cancerous condition or tissue. In other aspects, the increased or upregulation of hsa-miR-205, hsa-miR-183, hsa-miR-31, hsa-miR- 224, hsa-miR-182, hsa-miR-21 and/or hsa-miR-203 or complement thereof relative to normal cervix tissue is indicative of a cancerous condition or cancerous tissue.
[0027] In yet still further aspects of the invention, the reduced expression or down regulation of hsa-miR-133b, hsa-miR-455, hsa-asg-5021_stl, hsa-asg-13254_stl, hsa-asg- 14176_stl, hsa-miR-487b, hsa-miR-411, hsa-miR-574, hsa-miR-542-5p, hsa-asg-5617_stl, hsa-asg-14172_stl, hsa-asg-13304_st2, hsa-asg-13297_stl, hsa-miR157_st2, hsa-miR-329, hsa-asg-10202_st2, hsa-miR-369-5p, hsa-asg-13284_stl, hsa-asg-9687_stl, hsa-miR-433, hsa-miR-565, hsa-asg-562_stl, hsa-asg-279_st2, hsa-asg-8411_st2, hsa-asg-7472_st2, hsa- asg-13279_stl, hsa-miR-487a, hsa-cand206_stl, hsa-cand345_stl, hsa-asg-9696_stl, hsa- miR-485-3p, hsa-asg-13166_st2, hsa-miR-594_st2, hsa-miR-299-3p, hsa-miR-539, hsa-asg- 10883_stl, hsa-miR-585, hsa-miR-493-5ρ, hsa-asg-12964_st2, hsa-asg-4557_st2, hsa-asg- 10674_stl, hsa-asg-14230_stl, hsa-asg-9681_stl, hsa-miR-628, hsa-asg-13237_stl, hsa-asg- 13230_st2, hsa-miR-493-3p, hsa-miR-654, hsa-asg-33_stl, hsa-asg-8067_st2, hsa-asg- 11199_st2, hsa-asg-12346_st2, hsa-asg-11883_stl, hsa-miR102_st2, hsa-asg-2027_stl, hsa- asg-3145_stl, hsa-asg-7465_st2, hsa-asg-3376_stl, hsa-cand283_stl, hsa-asg-2301_st2, hsa- cand207_stl, hsa-miR-598, hsa-asg-10278_st2, hsa-asg-11181_stl, hsa-asg-7023_st2, hsa- asg-3597_st2, hsa-asg-5304_stl, hsa-cand720_stl, hsa-asg-13308_st2, hsa-asg-3038_st2, hsa- asg-13966_st2, hsa-asg-13189_stl, hsa-asg-11938_stl, hsa-asg-5740_st2, hsa-asg-8477_stl, hsa-asg-12325_st2, hsa-asg-12356_stl, hsa-asg-6758_stl, hsa-asg-522_stl, hsa-asg-4564_st2, hsa-asg-6951_st2, hsa-asg-9920_stl, hsa-asg-13613_st2, hsa-miR-184, hsa-miR-503, hsa- miR-485-5p, hsa-miR-494, hsa-miR-504, hsa-miR-211, hsa-miR-99b, hsa-miR-499, hsa-miR- 422b, hsa-miR-338, hsa-miR-422a, hsa-miR-331, hsa-miR-489, hsa-miR-324-5p, hsa-miR- 151, hsa-miR-17-3p, hsa-miR-340, and/or hsa-miR-194 or complement thereof relative to normal cervix tissue is indicative of a cancerous condition or tissue. In other aspects, the increased or upregulation of hsa-asg-11688_stl, hsa-miR-18b, hsa-miR-18a, hsa-asg- 3711_st2, hsa-miR-183, hsa-asg-2919_stl, hsa-asg-924_stl, and/or hsa-cand317_stl, or complement thereof relative to normal cervix tissue is indicative of a cancerous condition or cancerous tissue.
[0028] In yet still further aspects of the invention, the reduced expression or down regulation of hsa-let-7d*, hsa-let-7e*, hsa-miR-100*, hsa-miR-101*, hsa-miR-106b*, hsa- miR-10b*, hsa-miR-125a-5p, hsa-miR-125b-2*, hsa-miR-126*, hsa-miR-127-3p, hsa-miR- 129-3p, hsa-miR-135a, hsa-miR-136, hsa-miR-136*, hsa-miR-139-5p, hsa-miR-140-3p, hsa- miR-140-5p, hsa-miR-141 *, hsa-miR-142-3p, hsa-miR-143*, hsa-miR-144, hsa-miR-144*, hsa-miR-145*, hsa-miR-146b-5p, hsa-miR-151-3ρ, hsa-miR-154*, hsa-miR-15b*, hsa-miR- 17, hsa-miR-181c, hsa-miR-181d, hsa-miR-183*, hsa-miR-193a-5p, hsa-miR-195*, hsa-miR- 197, hsa-miR-199a-5p, hsa-miR-199b-3ρ, hsa-miR-199b-5ρ, hsa-miR-20a*, hsa-miR-212, hsa-miR-214*, hsa-miR-24-1*, hsa-miR-26b*, hsa-miR-28-3p, hsa-miR-28-5p, hsa-miR- 29a*, hsa-miR-29b-l*, hsa-miR-29b-2*, hsa-miR-29c*, hsa-miR-30a, hsa-miR-30a*, hsa- miR-30b*, hsa-miR-30c-2*, hsa-miR-32, hsa-miR-330-3p, hsa-miR-337-3p, hsa-miR-337-5p, hsa-miR-345, hsa-miR-361-5p, hsa-miR-374a, hsa-miR-374b, hsa-miR-374b*, hsa-miR-375, hsa-miR-376b, hsa-miR-376c, hsa-miR-377*, hsa-miR-410, hsa-miR-424*, hsa-miR-425, hsa-miR-431*, hsa-miR-450a, hsa-miR-451, hsa-miR-455-3p, hsa-miR-455-5p, hsa-miR-483- 3p, hsa-miR-486-5p, hsa-miR-488, hsa-miR-490-3p, hsa-miR-499-5p, hsa-miR-501-5p, hsa- miR-505*, hsa-miR-512-3ρ, hsa-miR-513c, hsa-miR-517*, hsa-miR-542-3p, hsa-miR-543, hsa-miR-574-3p, hsa-miR-602, hsa-miR-628-5p, hsa-miR-629*, hsa-miR-630, hsa-miR-650, hsa-miR-654-3p, hsa-miR-654-5p, hsa-miR-655, hsa-miR-656, hsa-miR-744, hsa-miR-766, i hsa-miR-768-3p, hsa-miR-873, hsa-miR-885-5p, hsa-miR-886-3p, hsa-miR-886-5p, hsa-miR-
889, hsa-miR-93*, hsa-miR-940, hsa-miR-99a*, and/or hsa-miR-99b* or complement thereof relative to normal cervix tissue is indicative of a cancerous condition or tissue. In other aspects, the increased or upregulation of hsa-miR-31*, hsa-miR-96, hsa-miR-21*, and/or hsa- miR-944 or complement thereof relative to normal cervix tissue is indicative of a cancerous condition or cancerous cells or cancerous tissue.
[0029] In certain aspects, the expression of one or more of hsa-miR-1, hsa-miR-15b, hsa- miR-133a, hsa-miR-143, hsa-miR-205, hsa-miR-21, hsa-miR-204, hsa-miR-195, hsa-miR- 100, hsa-miR-99a, hsa-miR-368, and/or hsa-miR-183 or complement thereof, including various combinations thereof, are assessed to determine if a target sample is cancerous. In other aspects, hsa-miR-16 can be used as reference. Embodiments of the invention include analysis of miR expression by amplification assays. In certain aspects, the PCR assay is quantitative PCR and in particular real time quantitative reverse transcription PCR (qRT- PCR).
[0030] In yet still further aspects of the invention, the methods for diagnosing a condition in a patient comprise measuring an expression profile of one or more miRNAs in a cervix sample from the patient suspected of having a cancerous condition, (e.g. , cervical squamous cell carcinoma) wherein the difference between the expression profile in the sample from the patient and an expression profile of normal tissue or a reference tissue is indicative of a cancerous disease or condition; wherein the miRNA is one or more of hsa-miR-1, hsa-miR- 100, hsa-miR-133a, hsa-miR-134, hsa-miR-143, hsa-miR-154, hsa-miR-182, hsa-miR-183, hsa-miR-195, hsa-miR-204, hsa-miR-205, hsa-miR-21, hsa-miR-218, hsa-miR-224, hsa-miR- 299-5p, hsa-miR-31, hsa-miR-368, hsa-miR-376a, hsa-miR-424, hsa-miR-99a, ambi-miR- 7029, hsa-miR-497, ambi-miR-7101, hsa-miR-133b, hsa-miR-455, hsa-asg-5021_stl, hsa- asg-13254_stl, hsa-asg-14176_stl, hsa-miR-487b, hsa-miR-411, hsa-miR-574, hsa-miR-542- 5p, hsa-asg-5617_stl, hsa-asg-14172_stl, hsa-asg-13304_st2, hsa-asg-13297_stl, hsa- miR157_st2, hsa-cand317_stl, hsa-miR-329, hsa-asg-10202_st2, hsa-miR-369-5p, hsa-asg- 13284_stl, hsa-asg-9687_stl, hsa-miR-433, hsa-miR-565, hsa-asg-562_stl, hsa-asg-279_st2, hsa-asg-8411_st2, hsa-asg-7472_st2, hsa-asg-13279_stl, hsa-miR-487a, hsa-cand206_stl, hsa-cand345_stl, hsa-asg-9696_stl, hsa-miR-485-3p, hsa-asg-13166_st2, hsa-miR-594_st2, hsa-miR-299-3p, hsa-asg-924_stl, hsa-miR-539, hsa-asg-10883_stl, hsa-miR-184, hsa-miR- 503, hsa-miR-485-5p, hsa-miR-494, hsa-miR-504, hsa-miR-211, hsa-miR-99b, or a complement thereof.
[0031] In certain aspects of the invention, the methods for diagnosing a condition in a patient comprise measuring an expression profile of one or more miRNAs in a cervix sample from the patient suspected of having a precancerous condition (e.g., cervical squamous intraepithelial lesion (SIL), also known as cervical intraepithelial neoplasias), wherein the difference between the expression profile in the sample from the patient and an expression profile of normal tissue or a reference is indicative of a disease or condition; wherein the miRNA is one or more of hsa-miR-1, hsa-miR-133a, hsa-miR-124a, hsa-miR-187, hsa-miR- 204, hsa-miR-145, hsa-miR-143, hsa-miR-325, hsa-miR-500, hsa-miR-196a, hsa-miR-125a, hsa-miR-376a, hsa-miR-505, hsa-miR-100, hsa-miR-99a, hsa-miR-141, hsa-miR-200a, ambi- miR-7029, hsa-miR-223, hsa-miR-205, hsa-miR-146a, hsa-miR-429, hsa-miR-200b, hsa- miR-182, hsa-miR-142-5p, hsa-miR-203, hsa-miR-21, hsa-miR-31, or hsa-miR-513 or complement thereof. In certain aspects, decreased expression, in a patient sample, of hsa- miR-1, hsa-miR-133a, hsa-miR-124a, hsa-miR-187, hsa-miR-204, hsa-miR-145, hsa-miR- 143, hsa-miR-325, hsa-miR-500, hsa-miR-196a, hsa-miR-125a, hsa-miR-376a, hsa-miR-505, hsa-miR-100, and/or hsa-miR-99a or complements thereof are indicative of SIL. Increased expression of hsa-miR-141, hsa-miR-200a, ambi-miR-7029, hsa-miR-233, hsa-miR-205, hsa- miR-146a, hsa-miR-429, hsa-miR-200b, hsa-miR-182, hsa-miR-142-5p, hsa-miR-203, hsa- miR-21, hsa-miR-513, and/or hsa-miR-31 can be indicative of SIL.
[0032] Other embodiments of the invention use the cycle threshold (Ct) values to distinguish between pre-cancerous or cancerous tissue and normal tissues. Ct values may be determined for one or more of the miRNA listed in Table 1 or their complements. In certain aspects, the Ct values for miR-1, miR-21, or both miR-1 and miR-21 can be used to distinguish between pre-cancerous or cancerous cervical samples or tissues and normal or non-cancerous tissues. [0033] A sample may be taken from a patient having or suspected of having a disease or pathological condition. A sample may also comprise nucleic acids or RNA isolated from a tissue or cell sample from a patient. In certain aspects, the sample can be, but is not limited to tissue (e.g., biopsy, particularly fine needle biopsy), blood, serum, plasma, or cervical samples (e.g., pap smear or punch biopsy). The sample can be fresh, frozen, fixed (e.g., formalin fixed), or embedded (e.g., paraffin embedded) tissues or cells. In a particular aspect, the sample is a cervical sample or nucleic acid or RNA isolated therefrom.
[0034] Methods of the invention can be used to diagnose or assess a pathological condition. In certain aspect, the condition is a non-cancerous condition, such as pre-cancerous cervical lesion. In other aspects the condition is a cancerous condition, such as cervical cancer.
[0035] The methods can further comprise one or more steps including: (a) obtaining a sample from the patient, (b) isolating nucleic acids from the sample, (c) labeling the nucleic acids isolated from the sample, and (d) hybridizing the labeled nucleic acids to one or more probes or primers. Nucleic acids of the invention include one or more nucleic acid comprising at least one segment having a sequence or complementary sequence of one or more of the miRNA sequences in Table 1. In certain aspects, the nucleic acids identify one or more miRNAs listed in Table 1. Nucleic acids of the invention are typically coupled to a support. Such supports are well known to those of ordinary skill in the art and include, but are not limited to glass, plastic, metal, or latex. In particular aspects of the invention, the support can be planar or in the form of a bead or other geometric shapes or configurations.
[0036] Certain embodiments of the invention include determining expression of one or more miRNA by using an amplification assay or a hybridization assay, a variety of which are well known to one of ordinary skill in the art. In certain aspects, an amplification assay can be a quantitative amplification assay, such as quantitative RT-PCR or the like. In still further aspects, a hybridization assay can include in situ hybridization, array hybridization assays or solution hybridization assays.
[0037] Aspects of the invention can be used to diagnose or assess a patient's condition. For example, the methods can be used to screen for a pathological condition, assess prognosis of a pathological condition, stage a pathological condition, or assess response of a pathological condition to therapy. [0038] Embodiments of the invention concern nucleic acids that perform the activities of or inhibit endogenous miRNAs when introduced into cells. In certain aspects, nucleic acids are synthetic or non-synthetic miRNA. Sequence-specific miRNA inhibitors can be used to inhibit sequentially or in combination the activities of one or more endogenous miRNAs in cells, as well those genes and associated pathways modulated by the endogenous miRNA.
[0039] The present invention concerns, in some embodiments, short nucleic acid molecules that function as miRNAs or as inhibitors of miRNA in a cell. The term "short" refers to a length of a single polynucleotide that is 5, 10, 15, 20, 25, 50, 100, or 150 nucleotides or fewer, including all integers or range derivable there between.
[0040] The present invention also concerns kits containing compositions of the invention or compositions to implement methods of the invention. In some embodiments, kits can be used to evaluate one or more miRNA molecules. In certain embodiments, a kit contains, contains at least or contains at most 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 100, 200, 300, 400, 500 or more miRNA probes, miRNA molecules or miRNA inhibitors, or any range and combination derivable therein. In some embodiments, there are kits for evaluating or modulating miRNA activity in a cell.
[0041] Kits may comprise components, which may be individually packaged or placed in a container, such as a tube, bottle, vial, syringe, or other suitable container means.
[0042] Individual components may also be provided in a kit in concentrated amounts; in some embodiments, a component is provided individually in the same concentration as it would be in a solution with other components. Concentrations of components may be provided as Ix, 2x, 5x, 10x, or 2Ox or more.
[0043] Kits for using miRNA probes or primers, synthetic miRNAs, nonsynthetic miRNAs, and/or miRNA inhibitors of the invention for therapeutic, prognostic, or diagnostic applications are also included as part of the invention. Specifically contemplated are any such molecules corresponding to any miRNA reported to influence biological activity, such as those discussed herein. [0044] In certain aspects, negative and/or positive control synthetic miRNAs and/or miRNA inhibitors are included in some kit embodiments. The control molecules can be used to verify transfection efficiency and/or control for transfection-induced changes in cells.
[0045] It is contemplated that any method or composition described herein can be implemented with respect to any other method or composition described herein and that different embodiments may be combined. It is specifically contemplated that any methods and compositions discussed herein with respect to miRNA molecules or miRNA may be implemented with respect to synthetic miRNAs to the extent the synthetic miRNA is exposed to the proper conditions to allow it to become a mature miRNA under physiological circumstances. The claims originally filed are contemplated to cover claims that are multiply dependent on any filed claim or combination of filed claims.
[0046] It is also contemplated that any one or more of the miRNA listed, particularly in Table 1 , may be specifically excluded from any particular set or subset of miRNA or nucleic acid.
[0047] Any embodiment of the invention involving specific miRNAs by name is contemplated also to cover embodiments involving miRNAs whose sequences are at least 70, 75, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98% identical to the mature sequence of the specified miRNA. This also includes the various fragments of these miRNA or nucleic acid sequences.
[0048] Embodiments of the invention include kits for analysis of a pathological sample by assessing miRNA profile for a sample comprising, in suitable container means, two or more miRNA probes, wherein the miRNA probes detect one or more of the miRNAs described in Table 1. The kit can further comprise reagents for labeling miRNA in the sample. The kit may also include the labeling reagents include at least one amine-modified nucleotide, poly(A) polymerase, and poly(A) polymerase buffer. Labeling reagents can include an amine-reactive dye.
[0049] Other embodiments of the invention are discussed throughout this application. Any embodiment discussed with respect to one aspect of the invention applies to other aspects of the invention as well and vice versa. The embodiments in the Example section are understood to be embodiments of the invention that are applicable to all aspects of the invention. [0050] It will be understood that shorthand notations are employed such that a generic description of an miRNA refers to any of its gene family members (distinguished by a number or sequence similarity), unless otherwise indicated. It is understood by those of skill in the art that a "gene family" refers to a group of genes having the same or similar miRNA coding sequence. Typically, members of a gene family are identified by a number following the initial designation; however some family members are identified by sequence similarity, for example see the various miRNA databases. For example, miR-16-1 and miR-16-2 are members of the miR-16 gene family and "mir-7" refers to miR-7-1, miR-7-2 and miR-7-3. Moreover, unless otherwise indicated, a shorthand notation refers to related miRNAs (distinguished by a letter). Thus, "let-7," for example, refers to let-7a-l, let7-a-2, let-7b, let- 7c, let-7d, let-7e, let-7f-l, and let-7f-2." Exceptions to this shorthand notations will be otherwise identified.
[0051] It will be understand that the term "providing" an agent is used to include "administering" the agent to a patient.
[0052] The terms "inhibiting," "reducing," or "prevention," or any variation of these terms, when used in the claims and/or the specification includes any measurable decrease or complete inhibition to achieve a desired result.
[0053] The use of the word "a" or "an" when used in conjunction with the term "comprising" in the claims and/or the specification may mean "one," but it is also consistent with the meaning of "one or more," "at least one," and "one or more than one."
[0054] It is contemplated that any embodiment discussed herein can be implemented with respect to any method or composition of the invention, and vice versa. Furthermore, compositions and kits of the invention can be used to achieve methods of the invention.
[0055] Throughout this application, the term "about" is used to indicate that a value includes the standard deviation of error for the device or method being employed to determine the value.
[0056] The use of the term "or" in the claims is used to mean "and/or" unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and "and/or." [0057] As used in this specification and claim(s), the words "comprising" (and any form of comprising, such as "comprise" and "comprises"), "having" (and any form of having, such as "have" and "has"), "including" (and any form of including, such as "includes" and "include") or "containing" (and any form of containing, such as "contains" and "contain") are inclusive or open-ended and do not exclude additional, unrecited elements or method steps.
[0058] Other objects, features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.
DESCRIPTION OF THE DRAWINGS
[0059] The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present invention. The invention may be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein.
[0060] FIG. 1 Principal Component Analysis of miRNAs expressed in normal cervix sample (NCX), cancerous cervix samples (Ca), and paired normal adjacent cervix samples (NAT).
[0061] FIGs. 2A-2B Comparison between array and qRT-PCR data. Real time RT-PCR were performed using 15 ng of total RNA input from the 34 samples previously profiled (16 NCX and 9 paired Ca and NAT samples). miRNA expression data obtained with primer sets specific for the indicated miRNAs were normalized to miR-16 expression level for each sample (miRNA Ct - miR-16 Ct). The graphs show the individual normalized miRNA expression levels determined by array or qRT-PCR and associated p-values (p (ANOVA) and *p (pairwise t-test)).
[0062] FIG. 3 Principal Component Analysis of miRNAs expressed in normal cervix sample (NCX), cancerous cervix samples (Ca) and paired normal adjacent cervix samples (NAT), and cervical cancer cell lines (CL). [0063] FIG. 4 Expression of miR-21 and miR-1 classifies normal and cancerous cervical tissues. Real time RT-PCR were performed with primer sets specific for miR-1 and miR-21 using 15 ng total RNA from 16 normal cervix (NCX), 9 cancerous cervix (Ca), or 9 normal adjacent cervix (NAT) samples. Raw Ct values were directly used to calculate the ratio of miR-21 to miR-1 expression, i.e., miR-21 Ct - miRl Ct in the logarithmic space.
DETAILED DESCRIPTION OF THE INVENTION
[0064] The present invention is directed to compositions and methods relating to preparation and characterization of miRNAs, as well as use of miRNAs for therapeutic, prognostic, and diagnostic applications, particularly those methods and compositions related to assessing and/or identifying cervical diseases and conditions.
I. CANCEROUS AND PRECANCEROUS CONDITIONS
[0065] As mentioned above, cervical cancer is the second most common cause of cancer in women worldwide (Pisani et al, 2002; Parkin et al, 2005). About 470,000 new cases are diagnosed and approximately 230,000 women die of cervical cancer every year (Pisani et al, 1999). While the majority (-80%) of these new cases and deaths occur in developing countries, it is estimated that approximately 3,700 women will die from invasive cervical cancer in the United States in 2007 (Jemal et al, 2007).
[0066] Cytological examination of cervical smears with Papanicolaou staining (Pap smear) is the screening method universally accepted for early detection of cervical cancer and its precursors. Pap smear screening programs have been very effective in reducing cervical cancer incidence and have reduced mortality rates by 60% among women aged 30 and over. However, even in those countries where these programs are routinely used, cervical cancer is still a significant public health problem. The Bethesda system categorizes Pap smear results as negative, ASC-US (atypical squamous cells of undetermined significance), ASCU-H (atypical squamous cells, cannot exclude high grade lesion), LSIL (low grade squamous intraepithelial lesion), HSIL (high grade intraepithelial lesion) and carcinoma. While guidelines and protocols for the management of women diagnosed with ASC-H, LSIL, HSIL and cancer are well established, the ASC-US entity is a significant problem for clinicians. The morphological criteria for ASC-US are suggestive, resulting in great variations in reported rates for ASC-US between laboratories. Approximately 3 million Pap smears are classified as ASC-US every year in the US, and the predictive value is very low. Although most Pap tests indicating ASC-US will revert spontaneously, it is estimated that in 5 to 15% of cases ASC-US Pap tests might already correspond to high grade SIL at histology. The ASC-US category is thus a source of confusion to both physicians and patients due to uncertainties that can lead to either the risk of false-positive diagnosis and unnecessary treatment or the risk of missing bona-fide lesions.
[0067] Epidemiological and molecular studies have demonstrated that human papillomaviruses (HPVs) are the etiological agents of the vast majority (99.7%) of cervical cancers and their intraepithelial precursors (Pisani, et al, 2002; Parkin et al, 2005; zur Hausen, 2002). About a dozen HPV types (including types 16, 18, 31 and 45) are called "high-risk" types because they can lead to cervical cancer, as well as anal cancer, vulvar cancer, and penile cancer. Several types of HPV, particularly type 16, have been found to be associated with oropharyngeal squamous-cell carcinoma, a form of head and neck cancer. HPV-induced cancers often have viral sequences integrated into the cellular DNA. Some of the HPV "early" genes, such as E6 and E7, are known to act as oncogenes that promote tumor growth and malignant transformation.
II. miRNA MOLECULES
[0068] MicroRNA molecules ("miRNAs") are generally 21 to 22 nucleotides in length, though lengths of 19 and up to 23 nucleotides have been reported. The miRNAs are each processed from a longer precursor RNA molecule ("precursor miRNA"). Precursor miRNAs are transcribed from non-protein-encoding genes. The precursor miRNAs have two regions of complementarity that enables them to form a stem-loop- or fold-back-like structure, which is cleaved in animals by a ribonuclease Ill-like nuclease enzyme called Dicer. The processed miRNA is typically a portion of the stem.
[0069] The processed miRNA (also referred to as "mature miRNA") become part of a large complex to down-regulate a particular target gene. Examples of animal miRNAs include those that imperfectly basepair with the target, which halts translation (Olsen et al, 1999; Seggerson et al, 2002). siRNA molecules also are processed by Dicer, but from a long, double-stranded RNA molecule. siRNAs are not naturally found in animal cells, but they can direct the sequence-specific cleavage of an mRNA target through a RNA-induced silencing complex (RISC) (Denli et al, 2003). [0070] The nucleic acid molecules are typically synthetic. The term "synthetic" means the nucleic acid molecule is isolated and not identical in sequence and/or chemical structure to a naturally-occurring nucleic acid molecule, such as an endogenous precursor miRNA or miRNA molecule. While in some embodiments, nucleic acids of the invention do not have an entire sequence that is identical to a sequence of a naturally-occurring nucleic acid, such molecules may encompass all or part of a naturally-occurring sequence. It is contemplated, however, that a synthetic nucleic acid administered to a cell may subsequently be modified or altered in the cell such that its structure or sequence is the same or similar as non-synthetic or naturally occurring nucleic acid, such as a mature miRNA sequence. For example, a synthetic nucleic acid may have a sequence that differs from the sequence of a precursor miRNA, but that sequence may be altered once in a cell to be the same as an endogenous, processed miRNA. The term "isolated" means that the nucleic acid molecules of the invention are initially separated from different (in terms of sequence or structure) and unwanted nucleic acid molecules such that a population of isolated nucleic acids is at least about 90% homogenous, and may be at least about 95, 96, 97, 98, 99, or 100% homogenous with respect to other polynucleotide molecules. In many embodiments of the invention, a nucleic acid is isolated by virtue of it having been synthesized in vitro separate from endogenous nucleic acids in a cell. It will be understood, however, that isolated nucleic acids may be subsequently mixed or pooled together in a variety of combinations.
[0071] In certain aspects, synthetic miRNA of the invention are RNA or RNA analogs. miRNA inhibitors may be DNA or RNA, or analogs thereof. miRNA and miRNA inhibitors of the invention are typically "synthetic nucleic acids."
[0072] In some embodiments, there is a recombinant or synthetic miRNA having a length of between 17 and 130 residues. The present invention concerns miRNA molecules that are, are at least, or are at most 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 61, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 140, 145, 150, 160, 170, 180, 190, 200 or more residues in length, including any integer or any range derivable therein, be it synthetic or non-synthetic. [0073] In certain embodiments, synthetic miRNA have (a) an "miRNA region" whose sequence from 5' to 3' is identical to all or a segment of a mature miRNA sequence, and (b) a "complementary region" whose sequence from 5' to 3' is between 60% and 100% complementary to the miRNA sequence. The term "miRNA region" refers to a region on the synthetic miRNA that is at least 75, 80, 85, 90, 95, or 100% identical, including all integers there between, to all or part of the sequence of a mature, naturally occurring miRNA sequence. In certain embodiments, the miRNA region is or is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9 or 100% identical to the sequence of a naturally-occurring miRNA.
[0074] The term "complementary region" refers to a region of a synthetic miRNA that is or is at least 60% complementary to a corresponding naturally occurring miRNA sequence. The complementary region is or is at least 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9 or 100% complementary to its corresponding naturally occurring miRNA, or any range derivable therein. With single polynucleotide sequences, there can be a hairpin loop structure as a result of chemical bonding between the miRNA region and the complementary region. Li other embodiments, the complementary region is on a different nucleic acid molecule than the miRNA region, in which case the complementary region is on the complementary strand and the miRNA region is on the active or functional strand.
[0075] In other embodiments of the invention, there are synthetic nucleic acids that are miRNA inhibitors. An miRNA inhibitor is between about 17 to 25 nucleotides in length and comprises a 5' to 3' sequence that is at least 90% complementary to the 5' to 3' sequence of a mature miRNA. In certain embodiments, an miRNA inhibitor molecule is 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleotides in length, or any range derivable therein. Moreover, an miRNA inhibitor has a sequence (from 5' to 3') that is or is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9 or 100% complementary, or any range derivable therein, to the 5' to 3' sequence of a mature miRNA, particularly a mature, naturally occurring miRNA. One of skill in the art could use a portion of a sequence that is complementary to the sequence of a mature miRNA as the sequence for an miRNA inhibitor. Moreover, that portion of a sequence can be altered so that it is still 90% complementary to the sequence of a mature miRNA. [0076] In some embodiments of the invention, a synthetic miRNA contains one or more design elements. These design elements include, but are not limited to: (i) a replacement group for the phosphate or hydroxyl of the nucleotide at the 5' terminus of the complementary region; (ii) one or more sugar modifications in the first or last 1 to 6 residues of the complementary region; or (iii) noncomplementarity between one or more nucleotides in the last 1 to 5 residues at the 3' end of the complementary region and the corresponding nucleotides of the miRNA region.
[0077] In certain embodiments, a synthetic miRNA has a nucleotide at its 5' end of the complementary region in which the phosphate and/or hydroxyl group has been replaced with another chemical group (referred to as the "replacement design"). In some cases, the phosphate group is replaced, while in others, the hydroxyl group has been replaced. In particular embodiments, the replacement group is biotin, an amine group, a lower alkylamine group, an acetyl group, 2'0-Me (2 Oxygen-methyl), DMTO (4,4'-dimethoxytrityl with oxygen), fluorescein, a thiol, or acridine, though other replacement groups are well known to those of skill in the art and can be used as well. This design element can also be used with an miRNA inhibitor.
[0078] Additional embodiments concern a synthetic miRNA having one or more sugar modifications in the first or last 1 to 6 residues of the complementary region (referred to as the "sugar replacement design"). In certain cases, there is one or more sugar modifications in the first 1, 2, 3, 4, 5, 6 or more residues of the complementary region, or any range derivable therein. In additional cases, there can be one or more sugar modifications in the last 1, 2, 3, 4, 5, 6 or more residues of the complementary region, or any range derivable therein. It will be understood that the terms "first" and "last" are with respect to the order of residues from the 5' end to the 3' end of the region. In particular embodiments, the sugar modification is a 2'O- Me modification. In further embodiments, there is one or more sugar modifications in the first or last 2 to 4 residues of the complementary region or the first or last 4 to 6 residues of the complementary region. This design element can also be used with an miRNA inhibitor. Thus, an miRNA inhibitor can have this design element and/or a replacement group on the nucleotide at the 5' terminus, as discussed above.
[0079] In other embodiments of the invention, there is a synthetic miRNA in which one or more nucleotides in the last 1 to 5 residues at the 3' end of the complementary region are not complementary to the corresponding nucleotides of the miRNA region ("noncomplementarity") (referred to as the "noncomplementarity design"). The noncomplementarity may be in the last 1, 2, 3, 4, and/or 5 residues of the complementary miRNA. In certain embodiments, there is noncomplementarity with at least 2 nucleotides in the complementary region.
[0080] It is contemplated that synthetic miRNA of the invention have one or more of the replacement, sugar modification, or noncomplementarity designs. In certain cases, synthetic RNA molecules have two of them, while in others these molecules have all three designs in place.
[0081] The miRNA region and the complementary region may be on the same or separate polynucleotides. In cases in which they are contained on or in the same polynucleotide, the miRNA molecule will be considered a single polynucleotide. In embodiments in which the different regions are on separate polynucleotides, the synthetic miRNA will be considered to be comprised of two polynucleotides.
[0082] When the RNA molecule is a single polynucleotide, there is a linker region between the miRNA region and the complementary region. In some embodiments, the single polynucleotide is capable of forming a hairpin loop structure as a result of bonding between the miRNA region and the complementary region. The linker constitutes the hairpin loop. It is contemplated that in some embodiments, the linker region is, is at least, or is at most 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 residues in length, or any range derivable therein. In certain embodiments, the linker is between 3 and 30 residues (inclusive) in length.
[0083] In addition to having an miRNA region and a complementary region, there may be flanking sequences as well at either the 5' or 3' end of the region. In some embodiments, there is or is at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 nucleotides or more, or any range derivable therein, flanking one or both sides of these regions.
A. Nucleic Acids
[0084] The present invention concerns miRNAs that can be labeled or amplified, used in array analysis, or employed in diagnostic, therapeutic, or prognostic applications, particularly those related to diseases and conditions of the cervix. The RNA may have been endogenously produced by a cell, or been synthesized or produced chemically or recombinantly. They may be isolated and/or purified. The term "miRNA," unless otherwise indicated, refers to the processed RNA, after it has been cleaved from its precursor. Table 1 indicates which SEQ ID NO correspond to a mature miRNA sequence. The name of the miRNA is often abbreviated and referred to without a hsa-, mmu-, or rno- prefix and will be understood as such, depending on the context. Unless otherwise indicated, miRNAs referred to in the application are human sequences identified as mir-X or let-X, where X is a number and/or letter.
[0085] In certain embodiments, a miRNA is designated with a "5P" or "3P" suffix. "5P" indicates that the mature miRNA derives from the 5' end of the precursor and a corresponding "3P" indicates that it derives from the 3' end of the precursor, as described on the world wide web at sanger.ac.uk. Moreover, in some embodiments, a miRNA is used that does not correspond to a known human miRNA. It is contemplated that these non-human miRNA probes may be used in embodiments of the invention or that there may exist a human miRNA that is homologous to the non-human miRNA. While the invention is not limited to human miRNA, in certain embodiments, miRNA from human cells or a human biological sample is evaluated. In other embodiments, any mammalian cell, biological sample, or preparation thereof may be employed.
[0086] In some embodiments of the invention, methods and compositions involving miRNA may concern miRNA and/or other nucleic acids. Nucleic acids may be, be at least, or be at most 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 441, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990, or 1000 nucleotides, or any range derivable therein, in length. Such lengths cover the lengths of processed miRNA, miRNA probes, precursor miRNA, miRNA containing vectors, control nucleic acids, and other probes and primers. In many embodiments, miRNA sequences are 19-24 nucleotides in length, while miRNA probes are 19-35 nucleotides in length, depending on the length of the processed miRNA and any flanking regions added. miRNA precursors are generally between 62 and 110 nucleotides in humans. [0087] Nucleic acids, and mimetics thereof, of the invention may have regions of identity or complementarity to another nucleic acid. It is contemplated that the region of complementarity or identity can be at least 5 contiguous residues, though it is specifically contemplated that the region is, is at least, or is at most 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 441, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990, or 1000 contiguous nucleotides. It is further understood that the length of complementarity within a precursor miRNA or between a miRNA probe and a miRNA or a miRNA gene are such lengths. Moreover, the complementarity may be expressed as a percentage, meaning that the complementarity between a nucleic acid and its target is 90% or greater over the length of the nucleic acid. In some embodiments, complementarity is or is at least 90%, 95% or 100%. In particular, such lengths may be applied to any nucleic acid comprising a nucleic acid sequence identified in any of SEQ ID NO:1 through SEQ ID NO:562 or any other sequence disclosed herein. Each of these SEQ ID NOs is disclosed herein. The commonly used name of the miRNA is given (with its identifying source in the prefix, for example, "hsa" for human sequences) and the processed miRNA sequence. Moreover, a lowercase letter in the table below may or may not be lowercase; for example, hsa-mir-130b can also be referred to as miR-130B. The term "miRNA probe" refers to a nucleic acid probe that can identify a particular miRNA or structurally related miRNAs.
[0088] It is understood that a miRNA is derived from genomic sequences or a gene. In this respect, the term "gene" is used for simplicity to refer to the genomic sequence encoding the precursor miRNA for a given miRNA. However, embodiments of the invention may involve genomic sequences of a miRNA that are involved in its expression, such as a promoter or other regulatory sequences.
[0089] The term "recombinant" may be used and this generally refers to a molecule that has been manipulated in vitro or that is a replicated or expressed product of such a molecule. [0090] The term "nucleic acid" is well known in the art. A "nucleic acid" as used herein will generally refer to a molecule (one or more strands) of DNA, RNA or a derivative or analog thereof, comprising a nucleobase. A nucleobase includes, for example, a naturally occurring purine or pyrimidine base found in DNA (e.g., an adenine "A," a guanine "G," a thymine "T" or a cytosine "C") or RNA (e.g., an A, a G, an uracil "U" or a C). The term "nucleic acid" encompasses the terms "oligonucleotide" and "polynucleotide," each as a subgenus of the term "nucleic acid."
[0091] The term "miRNA" generally refers to a single-stranded molecule, but in specific embodiments, molecules implemented in the invention will also encompass a region or an additional strand that is partially (between 10 and 50% complementary across length of strand), substantially (greater than 50% but less than 100% complementary across length of strand) or fully complementary to another region of the same single-stranded molecule or to another nucleic acid. Thus, nucleic acids may encompass a molecule that comprises one or more complementary or self-complementary strand(s) or "complement(s)" of a particular sequence comprising a molecule. For example, precursor miRNA may have a self- complementary region, which is up to 100% complementary. miRNA probes or nucleic acids of the invention can include, can be or can be at least 60, 65, 70, 75, 80, 85, 90, 95, 96, 97, 98, 99 or 100% complementary to their target.
[0092] As used herein, "hybridization," "hybridizes," or "capable of hybridizing" is understood to mean the forming of a double or triple stranded molecule or a molecule with partial double or triple stranded nature. The term "anneal" as used herein is synonymous with "hybridize." The term "hybridization," "hybridize(s)," or "capable of hybridizing" encompasses hybridization under "stringent condition(s)" or "high stringency" and "low stringency" or "low stringency condition(s)."
[0093] As used herein "stringent condition(s)" or "high stringency" are those conditions that allow hybridization between or within one or more nucleic acid strand(s) containing complementary sequence(s), but preclude hybridization of random sequences. Stringent conditions tolerate little, if any, mismatch between a nucleic acid and a target strand. Such conditions are well known to those of ordinary skill in the art, and are preferred for applications requiring high selectivity. Non-limiting applications include isolating a nucleic acid, such as a gene or a nucleic acid segment thereof, or detecting at least one specific mRNA transcript or a nucleic acid segment thereof, and the like. [0094] Stringent conditions may comprise low salt and/or high temperature conditions, such as provided by about 0.02 M to about 0.5 M NaCl at temperatures of about 42°C to about 70°C. It is understood that the temperature and ionic strength of a desired stringency are determined in part by the length of the particular nucleic acid(s), the length and nucleobase content of the target sequence(s), the charge composition of the nucleic acid(s), and to the presence or concentration of formamide, tetramethylammonium chloride or other solvent(s) in a hybridization mixture.
[0095] It is also understood that these ranges, compositions and conditions for hybridization are mentioned by way of non-limiting examples only, and that the desired stringency for a particular hybridization reaction is often determined empirically by comparison to one or more positive or negative controls. Depending on the application envisioned it is preferred to employ varying conditions of hybridization to achieve varying degrees of selectivity of a nucleic acid towards a target sequence. In a non-limiting example, identification or isolation of a related target nucleic acid that does not hybridize to a nucleic acid under stringent conditions may be achieved by hybridization at low temperature and/or high ionic strength. Such conditions are termed "low stringency" or "low stringency conditions," and non-limiting examples of low stringency include hybridization performed at about 0.15 M to about 0.9 M NaCl at a temperature range of about 20°C to about 50°C. Of course, it is within the skill of one in the art to further modify the low or high stringency conditions to suite a particular application.
1. Nucleobases
[0096] As used herein a "nucleobase" refers to a heterocyclic base, such as for example a naturally occurring nucleobase (i.e., an A, T, G, C or U) found in at least one naturally occurring nucleic acid (i.e., DNA and RNA), and naturally or non-naturally occurring derivative(s) and analogs of such a nucleobase. A nucleobase generally can form one or more hydrogen bonds ("anneal" or "hybridize") with at least one naturally occurring nucleobase in manner that may substitute for naturally occurring nucleobase pairing (e.g., the hydrogen bonding between A and T, G and C, and A and U).
[0097] "Purine" and/or "pyrimidine" nucleobase(s) encompass naturally occurring purine and/or pyrimidine nucleobases and also derivative(s) and analog(s) thereof, including but not limited to, those a purine or pyrimidine substituted by one or more of an alkyl, carboxyalkyl, amino, hydroxyl, halogen (i.e., fluoro, chloro, bromo, or iodo), thiol or alkylthiol moiety. Preferred alkyl (e.g., alkyl, carboxyalkyl, etc.) moieties comprise of from about 1, about 2, about 3, about 4, about 5, to about 6 carbon atoms. Other non-limiting examples of a purine or pyrimidine include a deazapurine, a 2,6-diaminopurine, a 5-fluorouracil, a xanthine, a hypoxanthine, a 8-bromoguanine, a 8-chloroguanine, a bromothymine, a 8-aminoguanine, a 8- hydroxyguanine, a 8-methylguanine, a 8-thioguanine, an azaguanine, a 2-aminopurine, a 5- ethylcytosine, a 5-methylcyosine, a 5-bromouracil, a 5-ethyluracil, a 5-iodouracil, a 5- chlorouracil, a 5-propyluracil, a thiouracil, a 2-methyladenine, a methylthioadenine, a N5N- diemethyladenine, an azaadenines, a 8-bromoadenine, a 8-hydroxyadenine, a 6- hydroxyaminopurine, a 6-thiopurine, a 4-(6-aminohexyl/cytosine), and the like. Other examples are well known to those of skill in the art.
[0098] A nucleobase may be comprised in a nucleoside or nucleotide, using any chemical or natural synthesis method described herein or known to one of ordinary skill in the art. Such nucleobase may be labeled or it may be part of a molecule that is labeled and contains the nucleobase.
2. Nucleosides
[0099] As used herein, a "nucleoside" refers to an individual chemical unit comprising a nucleobase covalently attached to a nucleobase linker moiety. A non-limiting example of a "nucleobase linker moiety" is a sugar comprising 5-carbon atoms (i.e., a "5-carbon sugar"), including but not limited to a deoxyribose, a ribose, an arabinose, or a derivative or an analog of a 5-carbon sugar. Non-limiting examples of a derivative or an analog of a 5-carbon sugar include a 2'-fluoro-2'-deoxyribose or a carbocyclic sugar where a carbon is substituted for an oxygen atom in the sugar ring.
[00100] Different types of covalent attachment(s) of a nucleobase to a nucleobase linker moiety are known in the art. By way of non-limiting example, a nucleoside comprising a purine (i.e., A or G) or a 7-deazapurine nucleobase typically covalently attaches the 9 position of a purine or a 7-deazapurine to the 1 '-position of a 5-carbon sugar. In another non-limiting example, a nucleoside comprising a pyrimidine nucleobase (i.e., C, T or U) typically covalently attaches a 1 position of a pyrimidine to a 1 '-position of a 5-carbon sugar (Kornberg and Baker, 1992). 3. Nucleotides
[00101] As used herein, a "nucleotide" refers to a nucleoside further comprising a "backbone moiety." A backbone moiety generally covalently attaches a nucleotide to another molecule comprising a nucleotide, or to another nucleotide to form a nucleic acid. The "backbone moiety" in naturally occurring nucleotides typically comprises a phosphorus moiety, which is covalently attached to a 5-carbon sugar. The attachment of the backbone moiety typically occurs at either the 3'- or 5'-position of the 5-carbon sugar. However, other types of attachments are known in the art, particularly when a nucleotide comprises derivatives or analogs of a naturally occurring 5-carbon sugar or phosphorus moiety.
4. Nucleic Acid Analogs
[00102] A nucleic acid may comprise, or be composed entirely of, a derivative or analog of a nucleobase, a nucleobase linker moiety and/or backbone moiety that may be present in a naturally occurring nucleic acid. RNA with nucleic acid analogs may also be labeled according to methods of the invention. As used herein a "derivative" refers to a chemically modified or altered form of a naturally occurring molecule, while the terms "mimic" or "analog" refer to a molecule that may or may not structurally resemble a naturally occurring molecule or moiety, but possesses similar functions. As used herein, a "moiety" generally refers to a smaller chemical or molecular component of a larger chemical or molecular structure. Nucleobase, nucleoside and nucleotide analogs or derivatives are well known in the art, and have been described (see for example, Scheit, 1980, incorporated herein by reference).
[00103] Additional non-limiting examples of nucleosides, nucleotides or nucleic acids comprising 5-carbon sugar and/or backbone moiety derivatives or analogs, include those in: U.S. Patent 5,681,947, which describes oligonucleotides comprising purine derivatives that form triple helixes with and/or prevent expression of dsDNA; U.S. Patents 5,652,099 and 5,763,167, which describe nucleic acids incorporating fluorescent analogs of nucleosides found in DNA or RNA, particularly for use as fluorescent nucleic acids probes; U.S. Patent 5,614,617, which describes oligonucleotide analogs with substitutions on pyrimidine rings that possess enhanced nuclease stability; U.S. Patents 5,670,663, 5,872,232 and 5,859,221, which describe oligonucleotide analogs with modified 5-carbon sugars (i.e., modified T- deoxyfuranosyl moieties) used in nucleic acid detection; U.S. Patent 5,446,137, which describes oligonucleotides comprising at least one 5-carbon sugar moiety substituted at the 4' position with a substituent other than hydrogen that can be used in hybridization assays; U.S. Patent 5,886,165, which describes oligonucleotides with both deoxyribonucleotides with 3'-5' internucleotide linkages and ribonucleotides with 2'-5' internucleotide linkages; U.S. Patent 5,714,606, which describes a modified internucleotide linkage wherein a 3'-position oxygen of the internucleotide linkage is replaced by a carbon to enhance the nuclease resistance of nucleic acids; U.S. Patent 5,672,697, which describes oligonucleotides containing one or more 5' methylene phosphonate internucleotide linkages that enhance nuclease resistance; U.S. Patents 5,466,786 and 5,792,847, which describe the linkage of a substituent moiety which may comprise a drug or label to the 2' carbon of an oligonucleotide to provide enhanced nuclease stability and ability to deliver drugs or detection moieties; U.S. Patent 5,223,618, which describes oligonucleotide analogs with a 2 or 3 carbon backbone linkage attaching the 4' position and 3' position of adjacent 5-carbon sugar moiety to enhanced cellular uptake, resistance to nucleases and hybridization to target RNA; U.S. Patent 5,470,967, which describes oligonucleotides comprising at least one sulfamate or sulfamide internucleotide linkage that are useful as nucleic acid hybridization probe; U.S. Patents 5,378,825, 5,777,092, 5,623,070, 5,610,289 and 5,602,240, which describe oligonucleotides with three or four atom linker moiety replacing phosphodiester backbone moiety used for improved nuclease resistance, cellular uptake, and regulating RNA expression; U.S. Patent 5,858,988, which describes hydrophobic carrier agent attached to the 2'-O position of oligonucleotides to enhanced their membrane permeability and stability; U.S. Patent 5,214,136, which describes oligonucleotides conjugated to anthraquinone at the 5' terminus that possess enhanced hybridization to DNA or RNA; enhanced stability to nucleases; U.S. Patent 5,700,922, which describes PNA-DNA-PNA chimeras wherein the DNA comprises T- deoxy-erythro-pentofuranosyl nucleotides for enhanced nuclease resistance, binding affinity, and ability to activate RNase H; and U.S. Patent 5,708,154, which describes RNA linked to a DNA to form a DNA-RNA hybrid; U.S. Patent 5,728,525, which describes the labeling of nucleoside analogs with a universal fluorescent label.
[00104] Additional teachings for nucleoside analogs and nucleic acid analogs are U.S. Patent 5,728,525, which describes nucleoside analogs that are end-labeled; U.S. Patent 5,637,683, 6,251,666 (L-nucleotide substitutions), and 5,480,980 (7-deaza-2'deoxyguanosine nucleotides and nucleic acid analogs thereof). 5. Modified Nucleotides
[00105] Labeling methods and kits of the invention specifically contemplate the use of nucleotides that are both modified for attachment of a label and can be incorporated into a miRNA molecule. Such nucleotides include those that can be labeled with a dye, including a fluorescent dye, or with a molecule such as biotin. Labeled nucleotides are readily available; they can be acquired commercially or they can be synthesized by reactions known to those of skill in the art.
[00106] Modified nucleotides for use in the invention are not naturally occurring nucleotides, but instead, refer to prepared nucleotides that have a reactive moiety on them. Specific reactive functionalities of interest include: amino, sulfhydryl, sulfoxyl, aminosulfhydryl, azido, epoxide, isothiocyanate, isocyanate, anhydride, monochlorotriazine, dichlorotriazine, mono-or dihalogen substituted pyridine, mono- or disubstituted diazine, maleimide, epoxide, aziridine, sulfonyl halide, acid halide, alkyl halide, aryl halide, alkylsulfonate, N- hydroxysuccinimide ester, imido ester, hydrazine, azidonitrophenyl, azide, 3-(2-pyridyl dithio)-propionamide, glyoxal, aldehyde, iodoacetyl, cyanomethyl ester, p-nitrophenyl ester, o-nitrophenyl ester, hydroxypyridine ester, carbonyl imidazole, and the other such chemical groups. In some embodiments, the reactive functionality may be bonded directly to a nucleotide, or it may be bonded to the nucleotide through a linking group. The functional moiety and any linker cannot substantially impair the ability of the nucleotide to be added to the miRNA or to be labeled. Representative linking groups include carbon containing linking groups, typically ranging from about 2 to 18, usually from about 2 to 8 carbon atoms, where the carbon containing linking groups may or may not include one or more heteroatoms, e.g. S, O, N etc., and may or may not include one or more sites of unsaturation. Of particular interest in many embodiments, are alkyl linking groups, typically lower alkyl linking groups of 1 to 16, usually 1 to 4 carbon atoms, where the linking groups may include one or more sites of unsaturation. The functionalized nucleotides (or primers) used in the above methods of functionalized target generation may be fabricated using known protocols or purchased from commercial vendors, e.g., Sigma, Roche, Ambion, Biosearch Technologies and NEN. Functional groups may be prepared according to ways known to those of skill in the art, including the representative information found in U.S. Patents 4,404,289; 4,405,711; 4,337,063 and 5,268,486, and U.K. Patent 1,529,202, which are all incorporated by reference. [00107] Amine-modified nucleotides are used in several embodiments of the invention. The amine-modified nucleotide is a nucleotide that has a reactive amine group for attachment of the label. It is contemplated that any ribonucleotide (G, A, U, or C) or deoxyribonucleotide (G, A, T, or C) can be modified for labeling. Examples include, but are not limited to, the following modified ribo- and deoxyribo-nucleotides: 5-(3-aminoallyl)-UTP; 8-[(4- amino)butyl]-amino-ATP and 8-[(6-amino)butyl]-amino-ATP; N6-(4-amino)butyl-ATP, N6- (6-amino)butyl-ATP, N4-[2,2-oxy-bis-(ethylamine)]-CTP; N6-(6-Amino)hexyl-ATP; 8-[(6- Amino)hexyl] -amino- ATP; 5-propargylamino-CTP, 5-propargylamino-UTP; 5-(3- aminoallyl)-dUTP; 8-[(4-amino)butyl]-amino-dATP and 8-[(6-amino)butyl]-amino-dATP; N6-(4-amino)butyl-dATP, N6-(6-amino)butyl-dATP, N4-[2,2-oxy-bis-(ethylamine)]-dCTP; N6-(6-Amino)hexyl-dATP; 8-[(6-Amino)hexyl]-amino-dATP; 5-propargylamino-dCTP, and 5-propargylamino-dUTP. Such nucleotides can be prepared according to methods known to those of skill in the art. Moreover, a person of ordinary skill in the art could prepare other nucleotide entities with the same amine-modification, such as a 5-(3-aminoallyl)-CTP, GTP, ATP, dCTP, dGTP, dTTP, or dUTP in place of a 5-(3-aminoallyl)-UTP.
B. Preparation of Nucleic Acids
[00108] A nucleic acid may be made or prepared by any technique known to one of ordinary skill in the art, such as for example, chemical synthesis, enzymatic production or biological production. It is specifically contemplated that nucleic acids of the invention are chemically synthesized.
[00109] In some embodiments of the invention, miRNAs are recovered or isolated from a biological sample. The miRNA may be recombinant or it may be natural or endogenous to the cell (produced from the cell's genome). It is contemplated that a biological sample may be treated in a way so as to enhance the recovery of small RNA molecules such as miRNA. U.S. Patent Application Serial No. 10/667,126 describes such methods and it is specifically incorporated by reference herein. Generally, methods involve lysing cells with a solution having guanidinium and a detergent.
[00110] Alternatively, nucleic acid synthesis is performed according to standard methods. See, for example, Itakura and Riggs (1980). Additionally, U.S. Patents 4,704,362, 5,221,619, and 5,583,013 each describe various methods of preparing synthetic nucleic acids. Non- limiting examples of a synthetic nucleic acid (e.g., a synthetic oligonucleotide), include a nucleic acid made by in vitro chemically synthesis using phosphotriester, phosphite, or phosphoramidite chemistry and solid phase techniques such as described in EP 266,032, incorporated herein by reference, or via deoxynucleoside H-phosphonate intermediates as described by Froehler et al, 1986 and U.S. Patent 5,705,629, each incorporated herein by reference. In the methods of the present invention, one or more oligonucleotide may be used. Various different mechanisms of oligonucleotide synthesis have been disclosed in for example, U.S. Patents 4,659,774, 4,816,571, 5,141,813, 5,264,566, 4,959,463, 5,428,148, 5,554,744, 5,574,146, 5,602,244, each of which is incorporated herein by reference.
[00111] A non-limiting example of an enzymatically produced nucleic acid include one produced by enzymes in amplification reactions such as PCR™ (see for example, U.S. Patents 4,683,202 and 4,682,195, each incorporated herein by reference), or the synthesis of an oligonucleotide described in U.S. Patent 5,645,897, incorporated herein by reference. A non-limiting example of a biologically produced nucleic acid includes a recombinant nucleic acid produced (i.e., replicated) in a living cell, such as a recombinant DNA vector replicated in bacteria (see for example, Sambrook et al., 2001, incorporated herein by reference).
[00112] Oligonucleotide synthesis is well known to those of skill in the art. Various different mechanisms of oligonucleotide synthesis have been disclosed in for example, U.S. Patents 4,659,774, 4,816,571, 5,141,813, 5,264,566, 4,959,463, 5,428,148, 5,554,744, 5,574,146, 5,602,244, each of which is incorporated herein by reference.
[00113] Basically, chemical synthesis can be achieved by the diester method, the triester method polynucleotides phosphorylase method and by solid-phase chemistry. The diester method was the first to be developed to a usable state, primarily by Khorana and co-workers. (Khorana, 1979). The basic step is the joining of two suitably protected deoxynucleotides to form a dideoxynucleotide containing a phosphodi ester bond.
[00114] The main difference between the diester and triester methods is the presence in the latter of an extra protecting group on the phosphate atoms of the reactants and products (Itakura et al., 1975). Purification's are typically done in chloroform solutions. Other improvements in the method include (i) the block coupling of trimers and larger oligomers, (ii) the extensive use of high-performance liquid chromatography for the purification of both intermediate and final products, and (iii) solid-phase synthesis.
[00115] Polynucleotide phosphorylase method is an enzymatic method of DNA synthesis that can be used to synthesize many useful oligonucleotides (Gillam et al., 1978; Gillam et al, 1979). Under controlled conditions, polynucleotide phosphorylase adds predominantly a single nucleotide to a short oligonucleotide. Chromatographic purification allows the desired single adduct to be obtained. At least a trimer is required to start the procedure, and this primer must be obtained by some other method. The polynucleotide phosphorylase method works and has the advantage that the procedures involved are familiar to most biochemists.
[00116] Solid-phase methods draw on technology developed for the solid-phase synthesis of polypeptides, it has been possible to attach the initial nucleotide to solid support material and proceed with the stepwise addition of nucleotides. All mixing and washing steps are simplified, and the procedure becomes amenable to automation. These syntheses are now routinely carried out using automatic nucleic acid synthesizers.
[00117] Phosphoramidite chemistry (Beaucage and Lyer, 1992) has become by far the most widely used coupling chemistry for the synthesis of oligonucleotides. Phosphoramidite synthesis of oligonucleotides involves activation of nucleoside phosphoramidite monomer precursors by reaction with an activating agent to form activated intermediates, followed by sequential addition of the activated intermediates to the growing oligonucleotide chain (generally anchored at one end to a suitable solid support) to form the oligonucleotide product.
[00118] Recombinant methods for producing nucleic acids in a cell are well known to those of skill in the art. These include the use of vectors (viral and non- viral), plasmids, cosmids, and other vehicles for delivering a nucleic acid to a cell, which may be the target cell (e.g., a cancer cell) or simply a host cell (to produce large quantities of the desired RNA molecule). Alternatively, such vehicles can be used in the context of a cell free system so long as the reagents for generating the RNA molecule are present. Such methods include those described in Sambrook, 2003, Sambrook, 2001 and Sambrook, 1989, which are hereby incorporated by reference.
[00119] In certain embodiments, the present invention concerns nucleic acid molecules that are not synthetic. In some embodiments, the nucleic acid molecule has a chemical structure of a naturally occurring nucleic acid and a sequence of a naturally occurring nucleic acid, such as the exact and entire sequence of a single stranded primary miRNA (see Lee 2002), a single-stranded precursor miRNA, or a single-stranded mature miRNA. In addition to the use of recombinant technology, such non-synthetic nucleic acids may be generated chemically, such as by employing technology used for creating oligonucleotides.
C. Isolation of Nucleic Acids
[00120] Nucleic acids may be isolated using techniques well known to those of skill in the art, though in particular embodiments, methods for isolating small nucleic acid molecules, and/or isolating RNA molecules can be employed. Chromatography is a process often used to separate or isolate nucleic acids from protein or from other nucleic acids. Such methods can involve electrophoresis with a gel matrix, filter columns, alcohol precipitation, and/or other chromatography. If miRNA from cells is to be used or evaluated, methods generally involve lysing the cells with a chaotropic (e.g., guanidinium isothiocyanate) and/or detergent (e.g., N- lauroyl sarcosine) prior to implementing processes for isolating particular populations of RNA.
[00121] Methods may involve the use of organic solvents and/or alcohol to isolate nucleic acids, particularly miRNA used in methods and compositions of the invention. Some embodiments are described in U.S. Patent Application Serial No. 10/667,126, which is hereby incorporated by reference. Generally, this disclosure provides methods for efficiently isolating small RNA molecules from cells comprising: adding an alcohol solution to a cell lysate and applying the alcohol/lysate mixture to a solid support before eluting the RNA molecules from the solid support. In some embodiments, the amount of alcohol added to a cell lysate achieves an alcohol concentration of about 55% to 60%. While different alcohols can be employed, ethanol works well. A solid support may be any structure, and it includes beads, filters, and columns, which may include a mineral or polymer support with electronegative groups. A glass fiber filter or column has worked particularly well for such isolation procedures.
[00122] In specific embodiments, miRNA isolation processes include: a) lysing cells in the sample with a lysing solution comprising guanidinium, wherein a lysate with a concentration of at least about 1 M guanidinium is produced; b) extracting miRNA molecules from the lysate with an extraction solution comprising phenol; c) adding to the lysate an alcohol solution for form a lysate/alcohol mixture, wherein the concentration of alcohol in the mixture is between about 35% to about 70%; d) applying the lysate/alcohol mixture to a solid support; e) eluting the miRNA molecules from the solid support with an ionic solution; and, f) capturing the miRNA molecules. Typically the sample is dried down and resuspended in a liquid and volume appropriate for subsequent manipulation.
D. Labels and Labeling Techniques
[00123] In some embodiments, the present invention concerns miRNA that are labeled. It is contemplated that miRNA may first be isolated and/or purified prior to labeling. This may achieve a reaction that more efficiently labels the miRNA, as opposed to other RNA in a sample in which the miRNA is not isolated or purified prior to labeling. In many embodiments of the invention, the label is non-radioactive. Generally, nucleic acids may be labeled by adding labeled nucleotides (one-step process) or adding nucleotides and labeling the added nucleotides (two-step process).
1. Labeling Techniques
[00124] In some embodiments, nucleic acids are labeled by catalytically adding to the nucleic acid an already labeled nucleotide or nucleotides. One or more labeled nucleotides can be added to miRNA molecules. See U.S. Patent 6,723,509, which is hereby incorporated by reference.
[00125] In other embodiments, an unlabeled nucleotide or nucleotides is catalytically added to a miRNA, and the unlabeled nucleotide is modified with a chemical moiety that enables it to be subsequently labeled. In embodiments of the invention, the chemical moiety is a reactive amine such that the nucleotide is an amine-modified nucleotide. Examples of amine- modified nucleotides are well known to those of skill in the art, many being commercially available such as from Ambion, Sigma, Jena Bioscience, and TriLink.
[00126] One issue for labeling miRNA is how to label the already existing molecule. The present invention concerns the use of an enzyme capable of using a di- or tri-phosphate ribonucleotide or deoxyribonucleotide as a substrate for its addition to a miRNA. Moreover, in specific embodiments, it involves using a modified di- or tri-phosphate ribonucleotide, which is added to the 3' end of a miRNA. Enzymes capable of adding such nucleotides include, but are not limited to, poly(A) polymerase, terminal transferase, and polynucleotide phosphorylase. Terminal transferase catalyzes the addition of nucleotides to the 3? terminus of a nucleic acid. Polynucleotide phosphorylase can polymerize nucleotide diphosphates without the need for a primer. 2. Labels
[00127] Labels on miRNA or miRNA probes may be colorimetric (includes visible and UV spectrum, including fluorescent), luminescent, enzymatic, or positron emitting (including radioactive). The label may be detected directly or indirectly. Radioactive labels include 125I, 32P, 33P, and 35S. Examples of enzymatic labels include alkaline phosphatase, luciferase, horseradish peroxidase, and β-galactosidase. Labels can also be proteins with luminescent properties, e.g., green fluorescent protein and phicoerythrin.
[00128] The colorimetric and fluorescent labels contemplated for use as conjugates include, but are not limited to, Alexa Fluor dyes, BODIPY dyes, such as BODIPY FL; Cascade Blue; Cascade Yellow; coumarin and its derivatives, such as 7-amino-4-methylcoumarin, aminocoumarin and hydroxycoumarin; cyanine dyes, such as Cy3 and Cy5; eosins and erythrosins; fluorescein and its derivatives, such as fluorescein isothiocyanate; macrocyclic chelates of lanthanide ions, such as Quantum Dye™; Marina Blue; Oregon Green; rhodamine dyes, such as rhodamine red, tetramethylrhodamine and rhodamine 6G; Texas Red; fluorescent energy transfer dyes, such as thiazole orange-ethidium heterodimer; and TOTAB. Specific examples of dyes include, but are not limited to, those identified above and the following: Alexa Fluor 350, Alexa Fluor 405, Alexa Fluor 430, Alexa Fluor 488, Alexa Fluor 500. Alexa Fluor 514, Alexa Fluor 532, Alexa Fluor 546, Alexa Fluor 555, Alexa Fluor 568, Alexa Fluor 594, Alexa Fluor 610, Alexa Fluor 633, Alexa Fluor 647, Alexa Fluor 660, Alexa Fluor 680, Alexa Fluor 700, and, Alexa Fluor 750; amine-reactive BODIPY dyes, such as BODIPY 493/503, BODIPY 530/550, BODIPY 558/568, BODIPY 564/570, BODIPY 576/589, BODIPY 581/591, BODIPY 630/650, BODIPY 650/655, BODIPY FL, BODIPY R6G, BODIPY TMR, and, BODIPY-TR; Cy3, Cy5, 6-FAM, Fluorescein Isothiocyanate, HEX, 6-JOE, Oregon Green 488, Oregon Green 500, Oregon Green 514, Pacific Blue, REG, Rhodamine Green, Rhodamine Red, Renographin, ROX, SYPRO, TAMRA, 2',4',5',7'- Tetrabromosulfonefluorescein, and TET.
[00129] Specific examples of fluorescently labeled ribonucleotides are available from Molecular Probes, and these include, Alexa Fluor 488-5-UTP, Fluorescein- 12-UTP, BODIPY FL-14-UTP, BODIPY TMR-14-UTP, Tetramethylrhodamine-6-UTP, Alexa Fluor 546-14- UTP, Texas Red-5-UTP, and BODIPY TR-14-UTP. Other fluorescent ribonucleotides are available from Amersham Biosciences, such as Cy3-UTP and Cy5-UTP. [00130] Examples of fluorescently labeled deoxyribonucleotides include Dinitrophenyl (DNP)-11-dUTP, Cascade Blue-7-dUTP, Alexa Fluor 488-5-dUTP, Fluorescein- 12-dUTP, Oregon Green 488-5-dUTP, BODIPY FL-14-dUTP, Rhodamine Green-5-dUTP, Alexa Fluor 532-5-dUTP, BODIPY TMR-14-dUTP, Tetramethylrhodamine-6-dUTP, Alexa Fluor 546-14- dUTP, Alexa Fluor 568-5-dUTP, Texas Red-12-dUTP, Texas Red-5-dUTP, BODIPY TR- 14- dUTP, Alexa Fluor 594-5-dUTP, BODIPY 630/650-14-dUTP, BODIPY 650/665-14-dUTP; Alexa Fluor 488-7-OBEA-dCTP, Alexa Fluor 546-16-OBEA-dCTP, Alexa Fluor 594-7- OBEA-dCTP, Alexa Fluor 647-12-OBEA-dCTP.
[00131] It is contemplated that nucleic acids may be labeled with two different labels. Furthermore, fluorescence resonance energy transfer (FRET) may be employed in methods of the invention (e.g., Klostermeier et al, 2002; Emptage, 2001; Didenko, 2001, each incorporated by reference).
[00132] Alternatively, the label may not be detectable per se, but indirectly detectable or allowing for the isolation or separation of the targeted nucleic acid. For example, the label could be biotin, digoxigenin, polyvalent cations, chelator groups and the other ligands, include ligands for an antibody.
3. Visualization Techniques
[00133] A number of techniques for visualizing or detecting labeled nucleic acids are readily available. Such techniques include, microscopy, arrays, Fluorometry, Light cyclers or other real time PCR machines, FACS analysis, scintillation counters, Phosphoimagers, Geiger counters, MRI, CAT, antibody-based detection methods (Westerns, immunofluorescence, immunohistochemistry), histochemical techniques, HPLC (Griffey et al, 1997), spectroscopy, capillary gel electrophoresis (Cummins et al., 1996), spectroscopy; mass spectroscopy; radiological techniques; and mass balance techniques.
[00134] When two or more differentially detectable labels are employed, fluorescent resonance energy transfer (FRET) techniques may be employed to characterize association of one or more nucleic acid. Furthermore, a person of ordinary skill in the art is well aware of ways of visualizing, identifying, and characterizing labeled nucleic acids, and accordingly, such protocols may be used as part of the invention. Examples of tools that may be used also include fluorescent microscopy, a BioAnalyzer, a plate reader, Storm (Molecular Dynamics), Array Scanner, FACS (fluorescent activated cell sorter), or any instrument that has the ability to excite and detect a fluorescent molecule.
III. ARRAY PREPARATION AND SCREENING A. Array Preparation
[00135] The present invention concerns the preparation and use of miRNA arrays or miRNA probe arrays. The arrays can be ordered macroarrays or microarrays of nucleic acid molecules (probes) that are fully or nearly complementary or identical to a plurality of miRNA molecules or precursor miRNA molecules and are positioned on a support or support material in a spatially separated organization. Macroarrays are typically a support {e.g., sheets of nitrocellulose or nylon) upon which probes have been spotted. Microarrays position the nucleic acid probes more densely such that up to 10,000 nucleic acid molecules can be fit into a region typically 1 to 4 square centimeters. Microarrays can be fabricated by spotting nucleic acid molecules, e.g., genes, oligonucleotides, etc., onto substrates or fabricating oligonucleotide sequences in situ on a substrate. Spotted or fabricated nucleic acid molecules can be applied in a high density matrix pattern of up to about 30 non-identical nucleic acid molecules per square centimeter or higher, e.g. up to about 100 or even 1000 per square centimeter. Microarrays typically use coated glass as the solid support, in contrast to the nitrocellulose-based material of filter arrays. By having an ordered array of miRNA- complementing nucleic acid samples, the position of each sample can be tracked and linked to the original sample. A variety of different array devices in which a plurality of distinct nucleic acid probes are stably associated with the surface of a solid support are known to those of skill in the art. Useful substrates or supports for arrays include nylon, glass, metal, plastic, and silicon. Such arrays may vary in a number of different ways, including average probe length, sequence or types of probes, nature of bond between the probe and the array surface, e.g. covalent or non-covalent, and the like. The labeling and screening methods of the present invention and the arrays are not limited in its utility with respect to any parameter except that the probes detect miRNA; consequently, methods and compositions may be used with a variety of different types of miRNA arrays.
[00136] Representative methods and apparatus for preparing a microarray have been described, for example, in U.S. Patents 5,143,854; 5,202,231; 5,242,974; 5,288,644; 5,324,633; 5,384,261; 5,405,783; 5,412,087; 5,424,186; 5,429,807; 5,432,049; 5,436,327; 5,445,934; 5,468,613; 5,470,710; 5,472,672; 5,492,806; 5,525,464; 5,503,980; 5,510,270; 5,525,464; 5,527,681; 5,529,756; 5,532,128; 5,545,531; 5,547,839; 5,554,501; 5,556,752; 5,561,071; 5,571,639; 5,580,726; 5,580,732; 5,593,839; 5,599,695; 5,599,672; 5,610;287; 5,624,711; 5,631,134; 5,639,603; 5,654,413; 5,658,734; 5,661,028; 5,665,547; 5,667,972; 5,695,940; 5,700,637; 5,744,305; 5,800,992; 5,807,522; 5,830,645; 5,837,196; 5,871,928; 5,847,219; 5,876,932; 5,919,626; 6,004,755; 6,087,102; 6,368,799; 6,383,749; 6,617,112; 6,638,717; 6,720,138, as well as WO 93/17126; WO 95/11995; WO 95/21265; WO 95/21944; WO 95/35505; WO 96/31622; WO 97/10365; WO 97/27317; WO 99/35505; WO 09923256; WO 09936760; WO0138580; WO 0168255; WO 03020898; WO 03040410; WO 03053586; WO 03087297; WO 03091426; WO03100012; WO 04020085; WO 04027093; EP 373 203; EP 785 280; EP 799 897 and UK 8 803 000; the disclosures of which are all herein incorporated by reference.
[00137] It is contemplated that the arrays can be high density arrays, such that they contain 2, 20, 25, 50, 80, 100 or more different probes. It is contemplated that they may contain 1000, 16,000, 65,000, 250,000 or 1,000,000 or more different probes. The probes can be directed to targets in one or more different organisms or cell types. The oligonucleotide probes range from 5 to 50, 5 to 45, 10 to 40, 9 to 34, or 15 to 40 nucleotides in length in some embodiments. In certain embodiments, the oligonucleotide probes are 5, 10, 15, 20 to 20, 25, 30, 35, 40 nucleotides in length including all integers and ranges there between.
[00138] The location and sequence of each different probe sequence in the array are generally known. Moreover, the large number of different probes can occupy a relatively small area providing a high density array having a probe density of generally greater than about 60, 100, 600, 1000, 5,000, 10,000, 40,000, 100,000, or 400,000 different oligonucleotide probes per cm2. The surface area of the array can be about or less than about 1, 1.6, 2, 3, 4, 5, 6, 7, 8, 9, or 10 cm2.
[00139] Moreover, a person of ordinary skill in the art could readily analyze data generated using an array. Such protocols are disclosed above, and include information found in WO 9743450; WO 03023058; WO 03022421; WO 03029485; WO 03067217; WO 03066906; WO 03076928; WO 03093810; WO 03100448A1, all of which are specifically incorporated by reference. B. Sample Preparation
[00140] It is contemplated that the miRNA of a wide variety of samples can be analyzed using the arrays, index of miRNA probes, or array technology described herein and known to the skilled artisan. While endogenous miRNA is contemplated for use with compositions and methods of the invention, recombinant miRNA - including nucleic acids that are complementary or identical to endogenous miRNA or precursor miRNA - can also be handled and analyzed as described herein. Samples may be biological samples, in which case, they can be from biopsy, fine needle aspirates, exfoliates, scrappings, blood, tissue, organs, or any sample containing or constituting biological cells of interest. In certain embodiments, samples may be, but are not limited to, fresh, frozen, fixed, formalin fixed, paraffin embedded, or formalin fixed and paraffin embedded. Alternatively, the sample may not be a biological sample, but be a chemical mixture, such as a cell-free reaction mixture (which may contain one or more biological enzymes).
C. Hybridization
[00141] After an array or a set of miRNA probes is prepared and the miRNA in the sample is labeled, the population of target nucleic acids is contacted with the array or probes under hybridization conditions, where such conditions can be adjusted, as desired, to provide for an optimum level of specificity in view of the particular assay being performed. Suitable hybridization conditions are well known to those of skill in the art and reviewed in Sambrook et al. (2001) and WO 95/21944. Of particular interest in many embodiments is the use of stringent conditions during hybridization. Stringent conditions are known to those of skill in the art.
[00142] It is specifically contemplated that a single array or set of probes may be contacted with multiple samples. The samples may be labeled with different labels to distinguish the samples. For example, a single array can be contacted with a tumor tissue sample labeled with Cy3, and normal tissue sample labeled with Cy5. Differences between the samples for particular miRNAs corresponding to probes on the array can be readily ascertained and quantified.
[00143] The small surface area of the array permits uniform hybridization conditions, such as temperature regulation and salt content. Moreover, because of the small area occupied by the high density arrays, hybridization may be carried out in extremely small fluid volumes {e.g., about 250 μl or less, including volumes of about or less than about 5, 10, 25, 50, 60, 70, 80 ,90, 100 μl, or any range derivable therein). In small volumes, hybridization may proceed very rapidly.
D. Differential Expression Analyses
[00144] Arrays of the invention can be used to detect differences between two samples. Specifically contemplated applications include identifying and/or quantifying differences between miRNA from a sample that is normal and from a sample that is not normal or contains abnormal components, between a cancerous condition and a non-cancerous condition, or between two differently treated samples. Also, miRNA may be compared between a sample believed to be susceptible to a particular disease or condition and one believed to be not susceptible or resistant to that disease or condition. A sample that is not normal is one exhibiting phenotypic trait(s) of a disease or condition or one believed to be not normal with respect to that disease or condition. Phenotypic traits include symptoms of or susceptibility to a disease or condition of which a component is or may or may not be genetic or caused by a hyperproliferative or neoplastic cell or cells.
[00145] An array comprises a solid support with nucleic acid probes attached to the support. Arrays typically comprise a plurality of different nucleic acid probes that are coupled to a surface of a substrate in different, known locations. These arrays, also described as "microarrays" or colloquially "chips" have been generally described in the art, for example, U.S. Pat. Nos. 5,143,854, 5,445,934, 5,744,305, 5,677,195, 6,040,193, 5,424,186 and Fodor et al, Science, 251:767-777 (1991), each of which is incorporated by reference in its entirety for all purposes. These arrays may generally be produced using mechanical synthesis methods or light directed synthesis methods which incorporate a combination of photolithographic methods and solid phase synthesis methods. Techniques for the synthesis of these arrays using mechanical synthesis methods are described in, e.g., U.S. Patent 5,384,261, incorporated herein by reference in its entirety for all purposes. Although a planar array surface is used in certain aspects, the array may be fabricated on a surface of virtually any shape or even a multiplicity of surfaces. Arrays may be nucleic acids on beads, gels, polymeric surfaces, fibers such as fiber optics, glass or any other appropriate substrate, see U.S. Patents 5,770,358, 5,789,162, 5,708,153, 6,040,193 and 5,800,992, which are hereby incorporated in their entirety for all purposes. Arrays may be packaged in such a manner as to allow for diagnostics or other manipulation of an all inclusive device, see for example, U.S. Patents 5,856,174 and 5,922,591 incorporated in their entirety by reference for all purposes. See also U.S. patent application Ser. No. 09/545,207, filed Apr. 7, 2000 for additional information concerning arrays, their manufacture, and their characteristics, which is incorporated by reference in its entirety for all purposes.
[00146] Particularly arrays can be used to evaluate samples with respect to diseases or conditions that include, but are not limited to pre-cancerous cervical lesion or cervical cancer.
[00147] Cancers that may be evaluated by methods and compositions of the invention include cancer cells particularly from the uterus or cervix, but may also include cells and cancer cells from the bladder, blood, bone, bone marrow, brain, breast, colon, esophagus, gastrointestine, gum, head, kidney, liver, lung, nasopharynx, neck, ovary, prostate, skin, stomach, testis, or tongue. miRNA can be evaluated in precancers, such as metaplasia, dysplasia, and hyperplasia.
[00148] It is specifically contemplated that the invention can be used to evaluate differences between stages of disease, such as between hyperplasia, neoplasia, pre-cancer and cancer, or between a primary tumor and a metastasized tumor.
[00149] Moreover, it is contemplated that samples that have differences in the activity of certain pathways may also be compared. These pathways include those involving the following factors: antibody response, apoptosis, calcium/NFAT signaling, cell cycle, cell migration, cell adhesion, cell division, cytokines and cytokine receptors, drug metabolism, growth factors and growth factor receptors, inflammatory response, insulin signaling, NFK-B signaling, angiogenesis, adipogenesis, cell adhesion, viral infection, bacterial infection, senescence, motility, glucose transport, stress response, oxidation, aging, telomere extension, telomere shortening, neural transmission, blood clotting, stem cell differentiation, G-Protein Coupled Receptor (GPCR) signaling, and p53 misregulation.
[00150] Cellular pathways that may be profiled also include but are not limited to the following: any adhesion or motility pathway including but not limited to those involving cyclic AMP, protein kinase A, G-protein couple receptors, adenylyl cyclase, L-selectin, E- selectin, PECAM, VCAM-I, α-actinin, paxillin, cadherins, AKT, integrin-α, integrin-β, RAF-I, ERK, PI-3 kinase, vinculin, matrix metalloproteinases, Rho GTPases, p85, trefoil factors, profilin, FAK, MAP kinase, Ras, caveolin, calpain-1, calpain-2, epidermal growth factor receptor, ICAM-I, ICAM-2, cofilin, actin, gelsolin, RhoA, RACl, myosin light chain kinase, platelet-derived growth factor receptor or ezrin; any apoptosis pathway including but not limited to those involving AKT, Fas ligand, NFKB, caspase-9, PB kinase, caspase-3, caspase-7, ICAD, CAD, EndoG, Granzyme B, Bad, Bax, Bid, Bak, APAF-I, cytochrome C, p53, ATM, Bcl-2, PARP, Chkl, Chk2, p21, c-Jun, p73, Rad51, Mdm2, Rad50, c-Abl, BRCA- 1, perforin, caspase-4, caspase-8, caspase-6, caspase-1, caspase-2, caspase-10, Rho, Jun kinase, Jun kinase kinase, Rip2, lamin-A, lamin-Bl, lamin-B2, Fas receptor, H2O2, Granzyme A, NADPH oxidase, HMG2, CD4, CD28, CD3, TRADD, IKK, FADD, GADD45, DR3 death receptor, DR4/5 death receptor, FLIPs, APO-3, GRB2, SHC, ERK, MEK, RAF-I, cyclic AMP, protein kinase A, E2F, retinoblastoma protein, Smac/Diablo, ACH receptor, 14-3-3, FAK, SODD, TNF receptor, RIP, cyclin-Dl, PCNA, BcI-XL, PIP2, PIP3, PTEN, ATM, Cdc2, protein kinase C, calcineurin, IKKα, IKKβ, IKKγ, SOS-I, c-FOS, Traf-1, Traf-2, IκBβor the proteasome; any cell activation pathway including but not limited to those involving protein kinase A, nitric oxide, caveolin-1, actin, calcium, protein kinase C, Cdc2, cyclin B, Cdc25, GRB2, SRC protein kinase, ADP-ribosylation factors (ARFs), phospholipase D, AKAP95, p68, Aurora B, CDKl, Eg7, histone H3, PKAc, CD80, PI3 kinase, WASP, Arp2, Arp3, plό, p34, p20, PP2A, angiotensin, angiotensin-converting enzyme, protease-activated receptor- 1, protease-activated receptor-4, Ras, RAF-I, PLCβ, PLCγ, COX-I, G-protein-couρled receptors, phospholipase A2, IP3, SUMOl, SUMO 2/3, ubiquitin, Ran, Ran-GAP, Ran-GEF, p53, glucocorticoids, glucocorticoid receptor, components of the SWI/SNF complex, RanBPl, RanBP2, importins, exportins, RCCl, CD40, CD40 ligand, p38, IKKα, IKKβ, NFKB, TRAF2, TRAF3, TRAF5, TRAF6, IL-4, IL-4 receptor, CDK5, AP-I transcription factor, CD45, CD4, T cell receptors, MAP kinase, nerve growth factor, nerve growth factor receptor, c-Jun, c-Fos, Jun kinase, GRB2, SOS-I, ERK-I, ERK, JAK2, STAT4, IL- 12, IL- 12 receptor, nitric oxide synthase, TYK2, IFNγ, elastase, IL- 8, epithelins, IL-2, IL-2 receptor, CD28, SMAD3, SMAD4, TGFβ or TGFβ receptor; any cell cycle regulation, signaling or differentiation pathway including but not limited to those involving TNFs, SRC protein kinase, Cdc2, cyclin B, Grb2, Sos-1, SHC, p68, Aurora kinases, protein kinase A, protein kinase C, Eg7, p53, cyclins, cyclin-dependent kinases, neural growth factor, epidermal growth factor, retinoblastoma protein, ATF-2, ATM, ATR, AKT, CHKl, CHK2, 14-3-3, WEEl, CDC25 CDC6, Origin Recognition Complex proteins, pl5, pl6, p27, p21, ABL, c-ABL, SMADs, ubiquitin, SUMO, heat shock proteins, Wnt, GSK-3, angiotensin, p73 any PPAR, TGFα, TGFβ, p300, E6-AP, Hect-E3s, MDM2, GADD45, Notch, cdc34, BRCA-I, BRCA-2, SKPl, the proteasome, CULl, E2F, plO7, steroid hormones, steroid hormone receptors, IκBα, IκBβ, Sin3A, heat shock proteins, Ras, Rho, ERKs, IKKs, PI3 kinase, Bcl-2, Bax, PCNA, MAP kinases, dynein, RhoA, PKAc, cyclin AMP, FAK, PIP2, PIP3, integrins, thrombopoietin, Fas, Fas ligand, PLK3, MEKs, JAKs, STATs, acetylcholine, paxillin calcineurin, p38, importins, exportins, Ran, Rad50, Rad51, DNA polymerase, RNA polymerase, Ran-GAP, Ran-GEF, NuMA, Tρx2, RCCl, Sonic Hedgehog, Crml, Patched (Ptc-1), MPF, CaM kinases, tubulin, actin, kinetochore-associated proteins, centromere- binding proteins, telomerase, TERT, PP2A, c-MYC, insulin, T cell receptors, B cell receptors, CBP, IKβ, NFKB, RACl, RAFl, EPO, diacylglycerol, c-Jun, c-Fos, Jun kinase, hypoxia- inducible factors, GATA4, β-catenin, α-catenin, calcium, arrestin, survivin, caspases, procaspases, CREB, CREM, cadherins, PECAMs, corticosteroids, colony-stimulating factors, calpains, adenylyl cyclase, growth factors, nitric oxide, transmembrane receptors, retinoids, G-proteins, ion channels, transcriptional activators, transcriptional coactivators, transcriptional repressors, interleukins, vitamins, interferons, transcriptional corepressors, the nuclear pore, nitrogen, toxins, proteolysis, or phosphorylation; or any metabolic pathway including but not limited to those involving the biosynthesis of amino acids, oxidation of fatty acids, biosynthesis of neurotransmitters and other cell signaling molecules, biosynthesis of polyamines, biosynthesis of lipids and sphingolipids, catabolism of amino acids and nutrients, nucleotide synthesis, eicosanoids, electron transport reactions, ER-associated degradation, glycolysis, fibrinolysis, formation of ketone bodies, formation of phagosomes, cholesterol metabolism, regulation of food intake, energy homeostasis, prothrombin activation, synthesis of lactose and other sugars, multi-drug resistance, biosynthesis of phosphatidylcholine, the proteasome, amyloid precursor protein, Rab GTPases, starch synthesis, glycosylation, synthesis of phoshoglycerides, vitamins, the citric acid cycle, IGF-I receptor, the urea cycle, vesicular transport, or salvage pathways. It is further contemplated that nucleic acids molecules of the invention can be employed in diagnostic and therapeutic methods with respect to any of the above pathways or factors. Thus, in some embodiments of the invention, a miRNA may be differentially expressed with respect to one or more of the above pathways or factors. In certain aspects, the pathways or cellular elements involved or effected by HPV infection can be assessed, evaulated, and/or monitored, e.g., hTERT, E6TP1, MCM7, Bak, and PDZ domain-containing proteins such as Scribble, h-Dlg, MAGI-I, MAGI-3, and MUPPl.
[00151] Phenotypic traits also include characteristics such as susceptibility or receptivity to particular drugs or therapeutic treatments (drug efficacy), and risk of drug toxicity. Samples that differ in these phenotypic traits may also be evaluated using the arrays and methods described.
[00152] hi certain embodiments, miRNA profiles may be generated to evaluate and correlate those profiles with pharmacokinetics. For example, miRNA profiles may be created and evaluated for patient tumor and blood samples prior to the patient's being treated or during treatment to determine if there are miRNAs whose expression correlates with the outcome of the patient. Identification of differential miRNAs can lead to a diagnostic assay involving them that can be used to evaluate tumor and/or blood samples to determine what drug regimen the patient should be provided. In addition, it can be used to identify or select patients suitable for a particular clinical trial. If a miRNA profile is determined to be correlated with drug efficacy or drug toxicity that may be relevant to whether that patient is an appropriate patient for receiving the drug or for a particular dosage of the drug.
[00153] In addition to prognostic assays, samples from patients with a variety of diseases can be evaluated to determine if different diseases can be identified based on blood miRNA levels. A diagnostic assay can be created based on the profiles that doctors can use to identify individuals with a disease or who are at risk to develop a disease. Alternatively, treatments can be designed based on miRNA profiling. Examples of such methods and compositions are described in the U.S. Provisional Patent Application serial number 60/683,736 entitled "Methods and Compositions Involving miRNA and miRNA Inhibitor Molecules" filed on May 23, 2005, which is hereby incorporated by reference in its entirety.
E. Other Assays
[00154] In addition to the use of arrays and microarrays, it is contemplated that a number of difference assays could be employed to analyze miRNAs, their activities, and their effects. Such assays include, but are not limited to, nucleic amplification, polymerase chain reaction, quantitative PCR, RT-PCR, in situ hybridization, Northern hybridization, hybridization protection assay (HPA)(GenProbe), branched DNA (bDNA) assay (Chiron), rolling circle amplification (RCA), single molecule hybridization detection (US Genomics), Invader assay (ThirdWave Technologies), and/or Bridge Litigation Assay (Genaco).
IV. THERAPEUTIC METHODS
[00155] Methods of the invention include reducing or eliminating activity of one or more miRNAs in a cell comprising introducing into a cell an miRNA inhibitor; or supplying or enhancing the activity of one or more miRNAs in a cell. The present invention also concerns inducing certain cellular characteristics by providing to a cell a particular nucleic acid, such as a specific synthetic miRNA molecule or a synthetic miRNA inhibitor molecule. However, in methods of the invention, the miRNA molecule or miRNA inhibitor need not be synthetic. They may have a sequence that is identical to a naturally occurring miRNA or they may not have any design modifications, hi certain embodiments, the miRNA molecule and/or an miRNA inhibitor are synthetic, as discussed herein.
[00156] The particular nucleic acid molecule provided to the cell is understood to correspond to a particular miRNA in the cell, and thus, the miRNA in the cell is referred to as the "corresponding miRNA." In situations in which a named miRNA molecule is introduced into a cell, the corresponding miRNA will be understood to be the induced miRNA. It is contemplated, however, that the miRNA molecule introduced into a cell is not a mature miRNA but is capable of becoming a mature miRNA under the appropriate physiological conditions, hi cases in which a particular corresponding miRNA is being inhibited by a miRNA inhibitor, the particular miRNA will be referred to as the targeted miRNA. It is contemplated that multiple corresponding miRNAs may be involved. In particular embodiments, more than one miRNA molecule is introduced into a cell. Moreover, in other embodiments, more than one miRNA inhibitor is introduced into a cell. Furthermore, a combination of miRNA molecule(s) and miRNA inhibitor(s) may be introduced into a cell.
[00157] Methods include identifying a cell or patient in need of inducing those cellular characteristics. Also, it will be understood that an amount of a synthetic nucleic acid that is provided to a cell or organism is an "effective amount," which refers to an amount needed to achieve a desired goal, such as inducing a particular cellular characteristic(s).
[00158] In certain embodiments of the methods include providing or introducing to a cell a nucleic acid molecule corresponding to a mature miRNA in the cell in an amount effective to achieve a desired physiological result.
[00159] Moreover, methods can involve providing synthetic or nonsynthetic miRNA molecules. It is contemplated that in these embodiments, methods may or may not be limited to providing only one or more synthetic miRNA molecules or only on or more nonsynthetic miRNA molecules. Thus, in certain embodiments, methods may involve providing both synthetic and nonsynthetic miRNA molecules. In this situation, a cell or cells are most likely provided a synthetic miRNA molecule corresponding to a particular miRNA and a nonsynthetic miRNA molecule corresponding to a different miRNA. Furthermore, any method including a list of miRNAs using Markush group language may be articulated without the Markush group language and a disjunctive article (i.e., or) instead, and vice versa.
[00160] In some embodiments, there is a method for reducing or inhibiting cell proliferation in a cell comprising introducing into or providing to the cell an effective amount of (i) an miRNA inhibitor molecule or (ii) a synthetic or nonsynthetic miRNA molecule that corresponds to an miRNA sequence. In certain embodiments the methods involves introducing into the cell an effective amount of (i) an miRNA inhibitor molecule having a 5' to 3' sequence that is at least 90% complementary to all or part of the 5' to 3' sequence of one or more mature miRNA of Table 1.
[00161] Certain embodiments of the invention include methods of treating a pre-cancerous cervical lesion or a cancerous cervical condition. In one aspect, the method comprises contacting a cervical cell with one or more nucleic acid, synthetic miRNA, or miRNA comprising at least one nucleic acid segment having all or a portion of a miRNA sequence. The segment may be 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30 or more nucleotides or nucleotide analog, including all integers there between. An aspect of the invention includes the modulation of a miRNA or a mRNA within a target cell, such as a cervical cell.
[00162] Typically, an endogenous gene, miRNA or mRNA is modulated in the cell. In particular embodiments, the nucleic acid sequence comprises at least one segment that is at least 70, 75, 80, 85, 90, 95, or 100% identical in nucleic acid sequence to one or more miRNA sequence listed in Table 1. Modulation of the expression or processing of an endogenous gene, miRNA, or mRNA can be through modulation of the processing of an mRNA, such processing including transcription, transportation and/or translation with in a cell. Modulation may also be effected by the inhibition or enhancement of miRNA activity with a cell, tissue, or organ. Such processing may effect the expression of an encoded product or the stability of the mRNA. In still other embodiments, a nucleic acid sequence can comprise a modified nucleic acid sequence.
[00163] In particular embodiments, the cervical cell is a cervical cancer cell. Methods of the invention can further comprise administering a second therapy, such as chemotherapy, radiotherapy, surgery, or immunotherapy. The nucleic acid can be transcribed from a nucleic acid vector, such as a plasmid vector or a viral vector.
[00164] Methods of treating a pre-cancerous or cancerous cervical condition include contacting or administering to a cervical cell one or more nucleic acid comprising a miRNA sequence, wherein expression of an endogenous miRNA is modulated in the cervical cell; where the miRNA sequence is at least 70, 75, 80, 85% or more identical to one or more of the sequences identified in Table 1. In certain aspects, the activity of those miRNAs indicated as having increased expression in a pre-cancerous or cancerous tissue is decreased. In a further aspect, the miRNA activity of those miRNA indicated as having decreased expression in a pre-cancerous or cancerous tissue is increased.
[00165] In certain aspects, one or more miRNA sequence may include or comprise a modified nucleobase or nucleic acid sequence.
[00166] The methods may further comprise administering a second therapy. The second therapy can be, but is not limited to chemotherapy, radiotherapy, surgery, or immunotherapy.
[00167] In still further aspects, one or more miRNA are transcribed from a nucleic acid vector, such as a plasmid or viral vector.
[00168] In certain aspects, a subject is administered: one or more nucleic acid possessing a function of an miRNA having a nucleic acid segment having at least 80, 85, 90, 95, 97, 98, 99, or 100% nucleic acid sequence identity to those miRNA decreased or down-regulated in a disease or condition to be treated.
[00169] In certain aspects, a subject is administered: one or more miRNA inhibitors having a nucleic acid segment having at least 80, 85, 90, 95, 97, 98, 99, or 100% nucleic acid sequence identity to those miRNA increased or up-regulated in a disease or condition to be treated.
[00170] Synthetic nucleic acids can be administered to the subject or patient using modes of administration that are well known to those of skill in the art, particularly for therapeutic applications. It is particularly contemplated that a patient is human or any other mammal or animal.
[00171] It will be understood in methods of the invention that a cell or other biological matter such as an organism (including patients) can be provided an miRNA or miRNA molecule corresponding to a particular miRNA by administering to the cell or organism a nucleic acid molecule that functions as the corresponding miRNA once inside the cell. The form of the molecule provided to the cell may not be the form that acts as an miRNA once inside the cell. Thus, it is contemplated that in some embodiments, biological matter is provided a synthetic miRNA or a nonsynthetic miRNA, such as one that becomes processed into a mature and active miRNA once it has access to the cell's miRNA processing machinery. In certain embodiments, it is specifically contemplated that the miRNA molecule provided to the biological matter is not a mature miRNA molecule but a nucleic acid molecule that can be processed into the mature miRNA once it is accessible to miRNA processing machinery. The term "nonsynthetic" in the context of miRNA means that the miRNA is not "synthetic," as defined herein. Furthermore, it is contemplated that in embodiments of the invention that concern the use of synthetic miRNAs, the use of corresponding nonsynthetic miRNAs is also considered an aspect of the invention, and vice versa.
[00172] In other embodiments, the methods involve reducing cell viability comprising introducing into or providing to the cell an effective amount of (i) an miRNA inhibitor molecule or (ii) a synthetic or nonsynthetic miRNA molecule that corresponds to an miRNA sequence. Methods for inducing apoptosis have a number of therapeutic applications including, but not limited to, the treatment of pre-cancer or cancer.
[00173] The present invention also concerns using miRNA compositions to treat diseases or conditions or to prepare therapeutics for the treatment of diseases or conditions. It is contemplated that 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 more miRNA (or any range derivable therein) may be used for these embodiments, hi certain embodiments, methods involve one or more miRNA inhibitors and/or an miRNA molecules corresponding to any of these miRNAs, particularly for the treatment or prevention of cancer. Cancer includes, but is not limited to, malignant cancers, tumors, metastatic cancers, unresectable cancers, chemo- and/or radiation-resistant cancers, and terminal cancers.
[00174] In certain embodiments, methods also include targeting an miRNA to modulate in a cell or organism. The term "targeting an miRNA to modulate" or "targeting an miRNA" means a nucleic acid of the invention will be employed so as to modulate the selected miRNA. In some embodiments the modulation is achieved with a synthetic or non-synthetic miRNA that corresponds to the targeted miRNA, which effectively provides the targeted miRNA to the cell or organism (positive modulation). In other embodiments, the modulation is achieved with an miRNA inhibitor, which effectively inhibits the targeted miRNA in the cell or organism (negative modulation).
[00175] In some embodiments, the miRNA targeted to be modulated is an miRNA that affects a disease, condition, or pathway. In certain embodiments, the miRNA is targeted because a treatment can be provided by negative modulation of the targeted miRNA. hi other embodiments, the miRNA is targeted because a treatment can be provided by positive modulation of the targeted miRNA.
[00176] In certain methods of the invention, there is a further step of administering the selected miRNA modulator to a cell, tissue, organ, or organism (collectively "biological matter") in need of treatment related to modulation of the targeted miRNA or in need of the physiological or biological results discussed herein (such as with respect to a particular cellular pathway or result, such as a decrease in cell viability). Consequently, in some methods of the invention there is a step of identifying a patient in need of treatment that can be provided by the miRNA modulator(s). It is contemplated that an effective amount of an miRNA modulator can be administered in some embodiments. In particular embodiments, there is a therapeutic benefit conferred on the biological matter, where a "therapeutic benefit" refers to an improvement in the one or more conditions or symptoms associated with a disease or condition or an improvement in the prognosis, duration, or status with respect to the disease. It is contemplated that a therapeutic benefit includes, but is not limited to, a decrease in pain, a decrease in morbidity, a decrease in a severity or duration of a symptom. For example, with respect to cancer, it is contemplated that a therapeutic benefit can be inhibition of tumor growth, prevention of metastasis, reduction in number of metastases, inhibition of cancer cell proliferation, inhibition of cancer cell proliferation, induction of cell death in cancer cells, inhibition of angiogenesis near cancer cells, induction of apoptosis of cancer cells, reduction in pain, reduction in risk of recurrence, induction of chemo- or radiosensitivity in cancer cells, prolongation of life, palliation of symptoms related to the condition, and/or delay of death directly or indirectly related to a cancer.
[00177] Furthermore, it is contemplated that the miRNA compositions may be provided as part of a therapy to a patient, in conjunction with traditional therapies or preventative agents. Moreover, it is contemplated that any method discussed in the context of therapy may be applied as a preventative measure, particularly in a patient identified to be potentially in need of the therapy or at risk of the condition or disease for which a therapy is needed.
[00178] In addition, methods of the invention concern employing one or more nucleic acids corresponding to an miRNA and a therapeutic drug. The nucleic acid can enhance the effect or efficacy of the drug, reduce any side effects or toxicity, modify its bioavailability, and/or decrease the dosage or frequency needed. In certain embodiments, the therapeutic drug is a cancer therapeutic. Consequently, in some embodiments, there is a method of treating cancer in a patient comprising administering to the patient the cancer therapeutic and an effective amount of at least one miRNA molecule that improves the efficacy of the cancer therapeutic or protects non-cancer cells. Cancer therapies also include a variety of combination therapies with both chemical and radiation based treatments. Combination chemotherapies include but are not limited to, for example, bevacizumab, cisplatin (CDDP), carboplatin, EGFR inhibitors (gefitinib and cetuximab), procarbazine, mechlorethamine, cyclophosphamide, camptothecin, COX-2 inhibitors {e.g., celecoxib) ifosfamide, melphalan, chlorambucil, busulfan, nitrosurea, dactinomycin, daunorubicin, doxorubicin (adriamycin), bleomycin, plicomycin, mitomycin, etoposide (VP 16), tamoxifen, raloxifene, estrogen receptor binding agents, taxol, taxotere, gemcitabien, navelbine, farnesyl-protein transferase inhibitors, transplatinum, 5-fluorouracil, vincristin, vinblastin and methotrexate, or any analog or derivative variant of the foregoing.
[00179] Generally, inhibitors of miRNAs can be given to achieve the opposite effect as compared to when nucleic acid molecules corresponding to the mature miRNA are given. Similarly, nucleic acid molecules corresponding to the mature miRNA can be given to achieve the opposite effect as compared to when inhibitors of the miRNA are given. For example, miRNA molecules that increase cell proliferation can be provided to cells to increase proliferation or inhibitors of such molecules can be provided to cells to decrease cell proliferation. The present invention contemplates these embodiments in the context of the different physiological effects observed with the different miRNA molecules and miRNA inhibitors disclosed herein. These include, but are not limited to, the following physiological effects: increase and decreasing cell proliferation; increasing or decreasing apoptosis; increasing or decreasing transformation; increasing or decreasing cell viability; activating, stimulating or suppressing cellular pathways; reduce or increase viable cell number; and increase or decrease number of cells at a particular phase of the cell cycle. Methods of the invention are generally contemplated to include providing or introducing one or more different nucleic acid or mimetic molecules corresponding to one or more different miRNA molecules. It is contemplated that the following, at least the following, or at most the following number of different nucleic acid molecules may be provided or introduced: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, or any range derivable therein. This also applies to the number of different miRNA molecules that can be provided or introduced into a cell.
V. KITS
[00180] Any of the compositions described herein may be comprised in a kit. In a non- limiting example, reagents for isolating miRNA, labeling miRNA, and/or evaluating a miRNA population using an array, nucleic acid amplification, and/or hybridization can be included in a kit, as well reagents for preparation of samples from cervical samples or other sample that have been, may have been, exposed to, or suspected of being infected with HPV. The kit may further include reagents for creating or synthesizing miRNA probes. The kits will thus comprise, in suitable container means, an enzyme for labeling the miRNA by incorporating labeled nucleotide or unlabeled nucleotides that are subsequently labeled. In certain aspects, the kit can include amplification reagents. In other aspects, the kit may include various supports, such as glass, nylon, polymeric beads, and the like, and/or reagents for coupling any probes and/or target nucleic acids. It may also include one or more buffers, such as reaction buffer, labeling buffer, washing buffer, or a hybridization buffer, compounds for preparing the miRNA probes, and components for isolating miRNA. Other kits of the invention may include components for making a nucleic acid array comprising miRNA, and thus, may include, for example, a solid support.
[00181] Kits for implementing methods of the invention described herein are specifically contemplated. In some embodiments, there are kits for preparing miRNA for multi-labeling and kits for preparing miRNA probes and/or miRNA arrays. In these embodiments, kit comprise, in suitable container means, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more of the following: (1) poly(A) polymerase; (2) unmodified nucleotides (G, A, T, C, and/or U); (3) a modified nucleotide (labeled or unlabeled); (4) poly(A) polymerase buffer; (5) at least one microfilter; (6) label that can be attached to a nucleotide; (7) at least one miRNA probe; (8) reaction buffer; (9) a miRNA array or components for making such an array; (10) acetic acid; (11) alcohol; and (12) solutions for preparing, isolating, enriching, and purifying miRNAs or miRNA probes or arrays. Other reagents include those generally used for manipulating RNA, such as formamide, loading dye, ribonuclease inhibitors, and DNase.
[00182] In specific embodiments, kits of the invention include an array containing miRNA probes, as described in the application. An array may have probes corresponding to all known miRNAs of an organism or a particular tissue or organ in particular conditions, or to a subset of such probes. The subset of probes on arrays of the invention may be or include those identified as relevant to a particular diagnostic, therapeutic, or prognostic application. For example, the array may contain one or more probes that is indicative or suggestive of (1) a disease or condition, (2) susceptibility or resistance to a particular drug or treatment; (3) susceptibility to toxicity from a drug or substance; (4) the stage of development or severity of a disease or condition (prognosis); and (5) genetic predisposition to a disease or condition.
[00183] For any kit embodiment, including an array, there can be nucleic acid molecules that contain or can be used to amplify a sequence that is a variant of, identical to or complementary to all or part of any of SEQ ID NOS: 1-562. In certain embodiments, a kit or array of the invention can contain one or more probes for the miRNAs identified by SEQ ID NOS: 1-562. Any nucleic acid discussed above may be implemented as part of a kit.
[00184] The components of the kits may be packaged either in aqueous media or in lyophilized form. The container means of the kits will generally include at least one vial, test tube, flask, bottle, syringe or other container, into which a component may be placed, and preferably, suitably aliquotted. Where there is more than one component in the kit (labeling reagent and label may be packaged together), the kit also will generally contain a second, third or other additional container into which the additional components may be separately placed. However, various combinations of components may be comprised in a vial. The kits of the present invention also will typically include a means for containing the nucleic acids, and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow molded plastic containers into which the desired vials are retained.
[00185] When the components of the kit are provided in one and/or more liquid solutions, the liquid solution is an aqueous solution, with a sterile aqueous solution being particularly preferred. [00186] However, the components of the kit may be provided as dried powder(s). When reagents and/or components are provided as a dry powder, the powder can be reconstituted by the addition of a suitable solvent. It is envisioned that the solvent may also be provided in another container means. In some embodiments, labeling dyes are provided as a dried power. It is contemplated that 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 120, 120, 130, 140, 150, 160, 170, 180, 190, 200, 300, 400, 500, 600, 700, 800, 900, 1000 μg or at least or at most those amounts of dried dye are provided in kits of the invention. The dye may then be resuspended in any suitable solvent, such as DMSO.
[00187] The container means will generally include at least one vial, test tube, flask, bottle, syringe and/or other container means, into which the nucleic acid formulations are placed, preferably, suitably allocated. The kits may also comprise a second container means for containing a sterile, pharmaceutically acceptable buffer and/or other diluent.
[00188] Such kits may also include components that facilitate isolation of the labeled miRNA. It may also include components that preserve or maintain the miRNA or that protect against its degradation. Such components may be RNAse-free or protect against RNAses. Such kits generally will comprise, in suitable means, distinct containers for each individual reagent or solution.
[00189] Kits of the invention may also include one or more of the following: Control RNA; nuclease- free water; RNase-free containers, such as 1.5 ml tubes; RNase-free elution tubes; PEG or dextran; ethanol; acetic acid; sodium acetate; ammonium acetate; guanidinium; detergent; nucleic acid size marker; RNase-free tube tips; and RNase or DNase inhibitors.
[00190] It is contemplated that such reagents are embodiments of kits of the invention. Such kits, however, are not limited to the particular items identified above and may include any reagent used for the manipulation or characterization of miRNA.
VI. EXAMPLES
[00191] The following examples are given for the purpose of illustrating various embodiments of the invention and are not meant to limit the present invention in any fashion. One skilled in the art will appreciate readily that the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those objects, ends and advantages inherent herein. The present examples, along with the methods described herein are presently representative of preferred embodiments, are exemplary, and are not intended as limitations on the scope of the invention. Changes therein and other uses which are encompassed within the spirit of the invention as defined by the scope of the claims will occur to those skilled in the art.
EXAMPLE 1 RNA FROM CERVICAL SPECIMENS AND CELL LINES
[00192] Paired tissue samples from nine squamous cell carcinomas (Ca) and nine normal adjacent regions (NAT) of the uterine cervix from the same patients and three squamous intraepithelial lesions (SIL) of the cervix were purchased from ProteoGenex (Culver City, CA, USA). Total RNA was extracted from these tissue samples using the mzWana™ miRNA Isolation Kit (Ambion; Austin, TX, USA) according to the manufacturer's protocol. Additional samples used in the study included purified total RNA from sixteen normal cervical tissue specimens (NCX) (FirstChoice® Human Cervix Total RNA, Ambion). In addition, total RNA was purified from cells of four squamous cell carcinoma-derived cervical cell lines (SW756, C4-1, CaSki, SiHa) and from an adenocarcinoma-derived cervical cell line (HeLa), using the mzVVana™ miRNA Isolation Kit (Ambion) according to the manufacturer's protocol. In all cases, purified total RNA was quantified using a NanoDrop® ND- 1000 spectrophotometer (NanoDrop Technologies; Wilmington, DE, USA). Tables 2, 3, and 4, below, show information regarding the nine matched Ca/NAT cervical samples (Table 2), the sixteen normal cervical samples (NCX, Table 3), and the cervical cell lines (CL) (Table 4)
Table 2. Tissue Sample Pathology Report for Nine Pairs of Cancerous (Ca) and Normal Adjacent Cervical Tissue (NAT Sam les. HPV; human a illomavirus.
Figure imgf000069_0001
Figure imgf000070_0001
Table 3. Patient Information for Sixteen Normal Cervical Tissue (NCX) Samples. HPV;
Figure imgf000070_0002
Table 4. Histology and HPV (human papillomavirus) Status for Five Cervical Cancer- Derived Cell Lines. SCC, s uamous cell carcinoma.
Figure imgf000070_0003
EXAMPLE 2: MIRNA EXPRESSION IN THE NORMAL CERVIX
[00193] The inventors first evaluated miRNA expression in normal cervical tissue samples. miRNA expression profiling was performed as previously described (Shingara et al, 2005) except that the miRNA fractions recovered from 20 μg total RNA were labeled with Cy5 (GE Healthcare Life Sciences; Piscataway, NJ, USA). To isolate miRNA fractions, total RNA samples were fractionated and purified using the flashPAGE™ fractionator and reagents (catalog# - AMI 3100; Ambion) according to the manufacturer's recommendations. Labeled miRNAs were hybridized to mirVana. miRNA Bioarrays Vl (Ambion) according to the manufacturer's instructions. The arrays contained 377 individual miRNA probes, including 281 human miRNAs from the mirBase Sequence Database (microrna.sanger.ac.uk) (Griffiths- Jones et al., 2006), 33 new human miRNAs (Ambi-miRs) and 63 mouse or rat miRNAs from the mirBase Sequence Database.
[00194] Following hybridization, the arrays were scanned using the Axon® GenePix 400B scanner and associated GenePix software (Molecular Devices Corporation; Sunnyvale, CA, USA). Raw array data were normalized with the variance stabilization method (VSN) (Huber et al., 2002). VSN is a global normalization process that stabilizes the variance evenly across the entire range of expression and utilizes calibration of signal followed by transformation of data to a generalized natural logarithmic space in lieu of the traditional logarithm base 2 transformation. Absolute values and differences in VSN transformed expression are denoted by H and ΔH, respectively, and were used for all subsequent data analyses. Differences in normalized expression values between samples (ΔH) were transformed to a generalized fold change via exponentiation base e. These values exhibit a compression for small differences in expression. For each array, the minimum observable threshold was determined by examining the foreground minus background median intensities for 'EMPTY' spots. The minimum threshold was defined as 5% symmetric trimmed mean plus 2 standard deviations across all 'EMPTY' spots on an individual array. For an overview of miRNA processing and analysis, see Davison et al, (2006).
[00195] The inventors first characterized miRNA expression profiles from 16 normal cervix specimens (Table 5). Approximately 200 miRNAs were detected above background signal (Mean (NCX) > 2.87). These included 171 human miRNAs, 17 miRNAs previously identified in mouse or rat, and 15 new human miRNAs (Ambi-miR-7027, -7029, -7039, - 7058, -7068-1, -7070, -7075, -7076, -7079, -7081, -7083, -7085, -7100, -7101, and -7105). Pair-wise t-test comparison analyses revealed no statistically significant differences in cervical miRNA expression between Caucasian (n=l l) and African- American (n=4) patients or between older (over 50 years of age) and younger (less than 50 years of age) patients. Table 5. Normalized Array Data for Sixteen Normal Cervix Tissue Samples (NCXl -NCXl 6). TV, threshold value. miRNAs > TV, number of miRNAs ex ressed at greater than threshold value. %, ercenta e of miRNAs ex ressed at greater than threshold value.
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
[00196] To identify miRNAs enriched or uniquely expressed in the normal cervix, the inventors performed a global comparison of miRNA expression between the 16 normal cervix samples and a reference set of human tissues. miRNA expression in 33 different human tissues was determined using the same microarray platform described above. The human reference set consisted of FirstChoice® Total RNA samples (Ambion) isolated from 33 distinct tissues: adipose, adrenal, aorta, bladder, bone marrow, brain, breast, colon, duodenum, esophagus, fallopian tube, heart, ileum, jejunum, kidney, liver, lung, lymph node, muscle, ovary, pancreas, pituitary, placenta, prostate, small intestine, spleen, stomach, testis, thymus, thyroid, trachea, uterus, and vena cava.
[00197] The inventors observed that mean expression levels for 103 human miRNAs in the normal cervix samples were significantly different from the mean expression levels in the 33 reference human tissues (Table 6). These include 94 miRNAs previously identified in humans (hsa-miRNAs) and nine new human miRNAs (ambi-miRs). Of these, 41 miRNAs expressed at higher levels in the cervix samples showed an expression increase greater than 2- fold; whereas, 25 miRNAs expressed at lower levels in the cervix samples showed an expression decrease greater than 2-fold. Several human miRNAs (hsa-miR-205, -196b, -203, -503, -196a, -99a, -187 and Ambi-miR-7083 and -7101) were enriched by at least five-fold (ΔH (NCX-REF) >1.6) in normal cervix samples compared to the reference tissues; whereas, two human miRNAs (hsa-miR-7 and -215) were underexpressed by at least five- fold (ΔH (NCX-REF) <-1.6) in the normal cervix samples.
Table 6. miRNAs with Significantly Different Expression in 16 Normal Cervix Samples vs 33 Different Human Tissues. NCX, normal cervix. REF, 33 human tissues reference set.
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
EXAMPLE 3:
MICRORNA EXPRESSION PROFILING DISTINGUISHES NORMAL AND CANCEROUS CERVIX TISSUE SAMPLES
[00198] miRNAs potentially relevant to carcinogenesis frequently exhibit differential expression in cancer versus normal samples collected from the same tissue type. In addition, miRNAs with differential expression in normal and cancerous samples may be used in the diagnosis of pre-cancerous and cancerous lesions. To identify miRNAs that may be useful for diagnosis of the most common type of cervical cancer (cervical squamous cell carcinoma), the inventors used the microarray platform described in Example 2 to compare global miRNA expression in nine cervical squamous cell carcinomas and nine paired normal adjacent tissue samples from the same patients. The results are shown below in Table 7. On average, 244 miRNAs were detected above background (>2.32) in normal adjacent tissues from cancer patients and 208 miRNAs were detected above background (>2.64) in cervical tumors.
[00199] The miRNA expression data from the nine Ca and nine NAT samples (Table 7) were normalized together with the data from the 16 NCX samples (Table 5). Mean miRNA expression levels are altered in the nine squamous cell carcinomas (Ca) when compared to miRNA expression levels in the adjacent normal cervical samples (NAT) and in the 16 normal cervix samples (NCX) (Table 8).
Table 7. Normalized Array Data for miRNA Expression in Nine Cervical Tumor Tissue Samples (CaI -Ca9) and Nine Cervical Normal Adjacent Tissue Samples (NATl -NAT-9). TV, threshold value. miRNAs > TV, miRNAs greater than threshold value. %, Percentage of miRNAs reater than threshold value.
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
[00200] The inventors identified 107 human miRNAs (96 hsa-miRNAs and 11 ambi- miRNAs) whose average expression levels were significantly different between the nine squamous cell carcinomas (Ca) and the nine samples from adjacent normal regions of the cervixes (NAT) in the same patients (Table 8, Flag (NAT vs Ca)=I). Among these miRNAs, 53 were down-regulated (ΔH(NAT-Ca)>0.69), and 21 were up-regulated (ΔH(NAT-Ca)<- 0.69) by at least 2-fold in Ca samples, when compared to their expression in NAT samples (Fold Change NAT vs Ca ≡2.0). Eleven miRNAs (hsa-miR-1, -133a, -204, -218, -143, -368, - 99a, -100, -195, -376a, and ambi-miR-7029) were down-regulated by more than 5-fold in Ca samples versus NAT samples (ΔH(NAT-Ca)>1.6). Three miRNAs (hsa-miR-205, -141, and - 182) were up-regulated more than 5-fold in Ca samples versus NAT samples (ΔH(NAT-Ca)<- 1.6).
[00201] The inventors identified 133 human miRNAs (121 hsa-miRNAs and 12 ambi- miRNAs) whose average expression levels were significantly different between the nine cervical tumor samples (Ca) and the sixteen normal cervical samples (NCX) (Table 8, Flag (NCX vs Ca)=I). Among these miRNAs, 70 were down-regulated (ΔH(NCX-Ca)>0.69) and 27 were up-regulated (ΔH(NCX-Ca)<-0.69), by at least 2-fold in Ca samples, when compared to their expression in NCX samples (Fold Change NCX vs Ca _≤.0). Seventeen miRNAs (hsa-miR-1, -133a, -204, -368, -99a, -100, -376a, -195, -218, -424, -497, -143, -299-5p, -154, -134, and ambi-miR-7101 and -7029) were down-regulated by more than 5-fold in Ca samples versus NCX samples (ΔH(NCX-Ca)>1.6). Seven miRNAs (hsa-miR-205, -183, -31, -224, -182, -21, and -203) were up-regulated more than 5-fold in Ca samples versus NCX sample (ΔH(NCX-Ca)<-1.6).
[00202] Overall, 103 human miRNAs were significantly differentially expressed between the cancer samples (Ca) and both the normal cervix samples (NCX) and the normal adjacent tissue samples (NAT). Thirty human miRNAs were significantly differentially expressed between Ca and NCX samples that were not significantly differentially expressed between Ca and NAT samples. Four distinct human miRNAs were significantly differentially expressed between Ca and NAT samples that were not significantly differentially expressed between Ca and NCX samples.
[00203] Unsupervised hierarchical clustering analysis (as determined by ANOVA) on the global miRNA expression in the 34 samples (16 NCX, 9 Ca, and 9 NAT) showed a clear segregation of the cancer samples away from the two normal tissue sample groups. Additional clustering and principal component analyses on all expressed miRNAs (FIG. 7), also showed clear segregation of the cancer samples from the two normal tissue sample groups. Similar results were obtained for principal component analysis of the miRNAs that were differentially expressed among the three groups.
[00204] The data described in this example demonstrate that miRNA expression analysis can be used to distinguish a cancerous cervical tissue sample from a normal, non-cancerous cervical tissue sample. The inventors have shown that specific miRNAs are differentially expressed (up-regulated or down-regulated) in cancerous cervical tissue as opposed to normal cervical tissues. Comparing the expression levels of these specific miRNAs in a cervical tissue sample that is suspected of being cancerous with their expression levels in a reference non-cancerous cervical tissue sample can indicate whether or not the suspect tissue sample is cancerous.
EXAMPLE 4 23 MIRNA BIOMARKERS FOR CERVICAL SQUAMOUS CELL CARCINOMA
[00205] Microarray profiling of cervical squamous cell carcinomas and normal primary tissues, using the microarrays described in Example 2, identified 103 mis-regulated (differentially expressed) miRNAs between the cervical cancer samples and both normal sample types (Table 8). The inventors identified 23 miRNAs that were differentially expressed, with at least a 5-fold change, between the Ca and NCX or NAT samples and a p- value<0.0001. These 23 miRNAs whose expression is significantly affected in cervical cancer represent novel biomarkers and therapeutic targets for cervical squamous cell carcinoma. Their identity and associated array data are summarized in Table 9.
Table 9. Top 23 miRNAs Identified on mrWana™ miRNA Bioarrays Vl (Ambion) as Differentially Expressed in Cervical Squamous Cell Carcinoma.
Figure imgf000106_0001
Figure imgf000107_0001
EXAMPLE 5 MICROARRAY DATA VALIDATION BY QRT-PCR
[00206] To verify array data, the inventors performed real-time qRT-PCR reactions for 12 of the top 23 miRNAs in Table 9 with very distinct expression patterns between cervical squamous cell carcinomas (Ca) and normal adjacent tissues (NAT) or normal cervix (NCX) samples (hsa-miR-1, -15b, -133a, -143, -205, -21, -204, -195, -100, -99a, -368, and -183) and one miRNA with no significant change (hsa-miR-16), which was used for normalization. qRT-PCR reactions were performed using TaqMan® MicroRNA Assays (Applied Biosystems; Foster City, CA, USA) according to the manufacturer's instructions. Reactions included 15 ng of total RNA per reaction and were incubated in the 7900HT Fast Real-Time PCR System (Applied Biosystems). Initial data analysis was done using the 7500 Fast System SDS 2.3 software. The inventors analyzed the 34 samples previously profiled (16 NCX and 9 paired Ca and NAT samples).
[00207] As illustrated in FIGs. 2A-2B, the relative variations of miRNA expression levels were very similar for qRT-PCR and array data. All 34 samples showed the expected miRNA expression patterns characteristic of normal cervix, normal adjacent tissue and squamous cell carcinoma, thus validating the array data. This was further confirmed after performing pair wise comparisons between qRT-PCR and array data for each paired Ca-NAT sample. EXAMPLE 6:
MIRNA EXPRESSION IN PRECANCEROUS CERVICAL SQUAMOUS INTRAEPITHELIAL LESIONS
[00208] The inventors sought to determine if the top 23 misregulated miRNAs in cervical cancer (Table 9) could distinguish precancerous cervical squamous epithelial lesions from normal cervical samples. The inventors evaluated miRNA expression in three cervical squamous intraepithelial lesions (SILs) (also known as cervical intraepithelial neoplasias, CINs) (Table 10). Pathological analysis classified two lesions (SILl, SIL2) as low grade (LSIL) and one (SIL3) as high grade (HSIL).
Table 10. Histology and HPV (Human Papillomavirus) Status for Three Cervical Cancer Precursor Lesions. CIN, cervical intraepithelial neoplasia.
Figure imgf000108_0001
[00209] miRNA expression analysis was carried out as described in Example 2, except that 7 μg of total RNA from each SIL sample were hybridized to the arrays. To avoid introducing a bias due to the differences in the mass input amounts for array analysis of the various samples (SIL, NCX, Ca, NAT) the inventors used the non parametric "rank product" method (Breitling et al, 2004) for identifying differentially expressed genes. Rank Product (RP) uses fold-change to assign ranks for all features in a dataset, providing reliable and sensitive results. Importantly, RP does not depend on the variance calculation for every gene and therefore is powerful when a small number of replicates is available. The robustness of this method has been demonstrated in previous studies where the list of genes generated from RP and a Student's T-test at a false discovery rate-corrected p-value of 5% were found in agreement (Breitling et al, 2004).
[00210] Using the RP method, 29 human miRNAs were identified as the most differentially expressed miRNAs between the SILs and the sixteen normal cervix samples, based on the rank value and an estimated percentage of false positive (PFP) of 5%. (Table 11). Among those, 15 miRNAs were down-regulated (Table 11, FC (NCX/SIL)>1) and 14 miRNAs were up-regulated in SILs (Table 11, FC (SIL/NCX)>1) compared to NCX. Interestingly, 11 out of the 15 miRNAs down-regulated in SILs were among the miRNAs down-regulated by at least 2-fold in Ca versus NCX (miR-1, -133a, -187, -204, -145, -143, -125a, -376a, -505, -100, and -99a). It is also noteworthy that miR-1, which is the most significantly down-regulated miRNA in the cervical cancers of our series, was also the most down-regulated miRNA in SILs. Among the 14 miRNAs up-regulated in SILs compared to the normal cervix samples, eight miRNAs (miR-141, -205, -146a, -200b, -182, -203, -21, and -31) were also up-regulated by at least 2-fold in the Ca samples compared to NCX samples. The presence of numerous identical miRNAs differentially expressed between Ca and NCX samples and between SIL and NCX samples, suggests that misregulation of these miRNAs is likely an early event in cervical tumorigenesis.
Table 11. miRNAs Differentially Expressed Between Squamous Intraepithelial Lesions (SILs) and Normal Cervix (NCX) Samples. RP/Rsum, Computed Rank Product/Sum of miRNA Probes on Array (377); FC, Fold Change; FC (NCX/SIL), miRNA expression is higher in normal cervix samples; FC (SIL/NCX), miRNA expression is higher in squamous intraepithelial lesions; pfp percentage of false positive.
Figure imgf000109_0001
Figure imgf000110_0001
[00211] The data described in this example demonstrate that miRNA expression analysis can be used to distinguish a pre-cancerous cervical squamous intraepithelial lesion (SIL) or a cervical intraepithelial neoplasia (CIN) from normal non-cancerous cervical tissue. Specific miRNAs are differentially expressed (up-regulated or down-regulated) in cervical SILs (CINs). Comparing the expression levels of these specific miRNAs in a cervical tissue sample that is suspected of being pre-cancerous with their expression levels in a reference non-cancerous cervical tissue sample can indicate whether or not the suspect tissue is precancerous.
EXAMPLE 7; MIRNA EXPRESSION IN CERVICAL CARCINOMA-DERIVED CELL LINES
[00212] The inventors determined miRNA expression profiles of five cervical cancer-derived cell lines using the same microarray platform described in Example 2. To compare miRNA expression profiles, miRNA expression data from the five cervical cancer cell lines (CL) were normalized together with the data described in earlier examples from the nine pairs of cervical tumors (Ca) and normal adjacent tissue specimens (NAT) and from the sixteen normal cervix samples (NCX) (Table 12).
Table 12. miRNAs Differentially Expressed Among Five Cervical Cancer-Derived Cell Lines, Nine Cervical Cancer Samples (Ca), Nine Paired Normal Adacent Tissue Sam les NAT , and Sixteen Normal Cervix Tissue Sam les NCX). FC; Fold Change
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
[00213] On average, 186 miRNAs were detected above background in the cell lines, corresponding to 49.5% of the miRNAs present on the array. As more than 200 miRNAs were detected above background in the normal cervix, cervical cancer, and normal adjacent tissue specimens, this indicates that expression of some miRNAs is lost in the cell lines evaluated here. Thirty-three miRNAs expressed in normal cervix samples, 27 miRNAs expressed in cervical tumor samples, and 41 miRNAs expressed in normal adjacent tissue samples were not expressed above background levels in the cell lines.
[00214] One hundred and forty-five human miRNAs (130 hsa-miRNAs and 15 ambi-miRs) had significantly different expression levels between the cell lines and the normal cervix samples (Table 12; Flag (CL vs NCX)=I). Among these miRNAs, 83 were down-regulated and 39 were up-regulated by at least 2-fold in the CL samples (FC, CLvsNCX Ξ2.0). Of these, 48 miRNAs were down-regulated by more than 5-fold in the CL samples (ΔH(CL- NCX)<-1.6), 31 were down-regulated by more than 10-fold in the CL samples (ΔH(CL- NCX)<-2.3), and 10 (hsa-miR-145, -143, -199a, -199aAS, -368, -214, -126, -133a, 199b, and Ambi-miR 7029) were down-regulated by more than 100-fold in the CL samples (ΔH(CL- NCX)<-4.61) (Table 12). In contrast, 11 miRNAs were up-regulated by more than 5-fold in the cell lines compared to the normal cervix samples (ΔH(CL-NCX)>1.6), including 3 miRNAs (hsa-miR-183, -182, and -96) that were over-expressed by more than 10-fold (ΔH(CL-NCX)>2.3). Overall, 56 miRNAs were identified that had a differential expression of more than 2-fold between Ca and NCX samples or between CL and NCX samples.
[00215] Hierarchical clustering and principal component analysis on the global miRNA expression data showed a clear segregation between the cell line samples (CL), and the three tissue samples (Ca, NAT, NCX). Further, the normal cervix (NCX) and normal adjacent tissue samples (NAT) clustered away from the cervical tumor samples (Ca) and the cell lines (CL) (FIG. 3). This observation was confirmed by pair-wise comparison of Pearson correlation values, calculated using mean normalized data for the 377 individual miRNAs on the array (Table 13). Overall, the mean miRNA expression levels in cell lines correlated better with expression levels in cervical cancer samples than with expression levels in normal adjacent and normal tissue samples.
Table 13. Paired Pearson Correlation Values. Values were calculated using mean normalized miRNA expression levels.
Figure imgf000120_0001
EXAMPLE 8
MIRNA EXPRESSION DIFFERENCES IN CERVICAL CANCER SPECIMENS AND CERVICAL CANCER CELL LINES ASSOCIATED WITH HPV STATUS
[00216] Among the nine cervical cancer specimens (Ca) analyzed, 4 were identified as positive for HPVl 6, 3 were positive for HPVl 8, 1 was positive for both HPVl 6 and HPVl 8, and 1 was negative for both HPVl 6 and 18, as determined by RT-PCR, using specific primers for the E6 open reading frames of HPVl 6 and HPVl 8. To investigate whether miRNA expression profiles could help distinguish between HPV16-positive and HPV18-positive cervical cancers, a pair wise t-test comparison was performed between the four HPVl 6- positive cancers and the three HPV18-positive cancers. No statistically significant differences were observed between the microRNA expression profiles of HPV16-positive and HPVl 8- positive cervical cancers..
[00217] Similarly, a pair wise t-test comparison between the three cell lines that are HPVl 8- positive and the two cell lines that are HPV16-positive did not reveal any miRNAs with significantly different expression between the two groups.
EXAMPLE 9:
IDENTIFICATION OF A CLASSIFIER FOR CERVICAL CANCER
BY QRT-PCR
[00218] Analyses of global miRNA expression profiles (Table 5, Table 7, FIG. 12), differentially expressed miRNAs (Table 8, Table 12), and the top 23 differentially expressed miRNA markers (Table 9) indicated that miRNA expression can distinguish normal and cancerous cervical tissues. To take this classification one step further, the minimal set of miRNAs that could discriminate between cancerous and non-cancerous tissues were identified by applying a quantitative RT-PCR analysis of mature miRNAs. The inventors found that the difference between raw Ct values of two miRNAs (miR-1 and -21) fulfilled the criteria (FIG. 4). qRT-PCR experiments performed on the 34 samples (NCX, CA, NAT) showed a perfect segregation between normal cervix (NCX), cervical cancer (Ca) and normal adjacent tissue (NAT) with a/?-value of 1.36 x 10"16.
EXAMPLE 10:
HIGH DENISTY MICROARRAY ANALYSIS OF MIRNAS IN NORMAL CERVIX
AND CERVICAL CANCER SAMPLES
[00219] miRNA array expression analysis: microRNA-containing fractions were recovered from 10 μg of total RNA isolated from each RNA sample, using the FlashPage fractionation system (Ambion, Inc.). Purified small RNAs were enzymatically biotinylated at the 3 '-termini and hybridized to a custom-made Affymetrix based microRNA array, Asuragen's Disco v Arrays V.I containing probes for 14,215 verified and candidate miRNAs from a number of sources including Sanger miRBase v9.2 database (http:microrna.sanger.ac.uk/sequences/) and published reports. Hybridization, washing, staining, imaging, and signal extraction were performed as recommended by the manufacturer, except that the 2OX GeneChip® Eukaryotic Hybridization Control cocktail was omitted.
[00220] Array data processing: The signal processing implemented for the Ambion miRCHIP is a multi-step process involving probe specific signal detection calls, background estimate and correction, constant variance stabilization and either array scaling or global normalization. For an overview of miRNA processing and analysis, see Davison et al. (2006). For more details, see supplementary information.
[00221] Probe specific signal detection calls: Each probe on an array is assayed for detection based on a Wilcoxon rank-sum test of the miRNA probe signal compared to the distribution of signals from GC-content matched anti-genomic probes. If the resulting p- value for the probes is <0.06 then it is considered "Detected above background." Probes with p- values > 0.06 have insufficient signal to discriminate from the background and are thus considered not detected.
[00222] Background estimate and correction: The same set of anti-genomic probes used to determine detection calls are used to estimate GC-content matched background signals. Each miRNA probe signal has a GC-content matched background estimate subtracted from its value. This GC-specific background contribution is estimated by the median signal from the distribution of GC-matched anti-genomic probes. [00223] Constant variance stabilization: Probes with low signal often exhibit a high degree of variability once transformed into logarithmic scales. To stabilize this low signal variability we add a constant value of 16 to the background corrected signal. This technique is commonly performed on microarray data. It was the recommended data preprocessing method by Affymetrix and Illumina in the Micro Array Quality Control (MAQC) project.
[00224] Normalization: The data were normalized with the VSN method (Huber et al, 2002; Szafranska et al, 2007). Briefly, VSN is a global normalization process that stabilizes the variance evenly across the entire range of expression. Differences in VSN transformed expression are denoted by Log2 diff and were used for all subsequent data analyses. Differences in normalized expression values between samples (Log 2 diff) can be transformed to a generalized fold change via exponentiation base 2. These values will exhibit a compression for small differences in expression.
[00225] Nine cervical cancer specimens (CaI -Ca9) and four normal cervix samples (NCX3, NCXlO, NCXl 3, and a new normal cervix sample - NCXl 7) were analyzed by the DiscovArray™ miRNA Expression Profiling Service (Asuragen). The DiscovArray™ microarray contains probes for 994 miRNAs from Sanger miRBase V9.2 (Griffiths- Jones et al, 2006) (467 human miRNAs, 234 rat miRNAs, 293 mouse miRNAs) and 12,894 predicted human miRNAs (see world wide web page at asuragen.com/services/discovarray.htmt).
[00226] One hundred forty four miRNAs were significantly differentially expressed between NCX and Ca samples by at least 2-fold (Table 14). Of those, 136 were down-regulated (Log2 Diff Ca vs NCX <-1.0) and 8 were up-regulated (Log2 Diff Ca vs NCX ≥l.O) in Ca samples compared to NCX samples. Sixty-three (63) miRNAs were down-regulated in Ca samples by more than 5-fold (Log2 Diff Ca vs NCX <-2.3) with a p-value <0.0001. Seventeen of those miRNAs were down-regulated by over 10-fold in Ca samples (Log2 Diff Ca vs NCX <-3.3).
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
EXAMPLE 11
AGILENT HUMAN MICRORNA MICRO ARRAY ANALYSIS OF MIRNAS IN NORMAL CERVIX AND CERVICAL CANCER SAMPLES
[00227] miRNA array expression analysis: miRNA expression in six cervical cancer specimens (CaI -4, Ca6, and Ca8) and six normal cervix samples (NCXl -3, NCXlO, NCXl 3, and NCXl 6) were further evaluated using the Human miRNA Microarray (V2) (Agilent Technologies, Inc.; Santa Clara, CA, USA). The Human miRNA Microarray contains probes for 723 human and 76 human viral microRNAs from the Sanger database v.10.1. Samples for miRNA profiling studies were processed by Asuragen Services (Asuragen, Inc.; Austin, TX, USA). Total RNA from each sample (200 ng) was dephosphorylated and the pCp-Cy3 labeling molecule was ligated to the 3' end of the RNA molecules. The labeled RNA was purified using a Bio-Spin P-6 column (Bio-Rad Laboratories Inc.; Hercules, CA, USA). Array hybridization, washing, staining, imaging, and signal extraction were performed according to Agilent's recommended procedures.
[00228] miRNA array signal processing: The signal processing implemented for the Agilent miRNA array is a multi-step process involving probe specific signal detection calls, background correction, and global normalization. For each probe, the contribution of signal due to background was estimated and removed by the Agilent Feature Extraction software as part of the data file output. Similarly, detection calls were based on the Agilent Feature Extraction software. Arrays within a specific analysis experiment were normalized together according to the VSN method described by Huber et al, 2002.
[00229] Background estimate and correction and probe detection: Three types of data are provided to evaluate each hybridization. The "Total Gene Signal" is the total probe signal multiplied by the number of probes per gene and is calculated after the background effects have been accounted for. The "Total Gene Error" is the square root of the square of the total probe error multiplied by the number of probes per gene. The "Total Probe Error" is the robust average for each replicated probe multiplied by the total number of probe replicates. The "Detection Call" is a binary number that indicates if the gene was detected on the miRNA microarray. Probes detected at least once across all samples in the experiment were considered for statistical analysis.
[00230] Global normalization: The inventors have found that the Variance Stabilization Normalization (VSN) algorithm provides an ideal balance of accuracy and precision while optimizing sensitivity and specificity of signal. One advantage of VSN, is that it accommodates negative values by using the generalized 1Og2 transformation.
[00231] Generalized Iog2 transformed: The post-normalized data scale is reported as generalized log2 data. The distribution of microarray data is typically log normal (i.e, it tends to follow a normal distribution pattern after log transformation). A normal distributed data is amendable to classical statistical treatments, including t-tests and one-way or two-way ANOVA.
[00232] For statistical hypothesis testing, a two-sample t-Test, with assumption of equal variance, was applied. This test is used to define which probes are considered to be significantly differentially expressed, or "significant", based on false discovery rate set at 0.05.
[00233] A total of 382 human miRNAs were expressed above background level in the normal cervix samples, representing 53% of the miRNAs present on the arrays. A total of 337 miRNAs were expressed above background level in the tumor samples, representing 46.6% of the miRNAs present on the arrays.
[00234] A total of 202 human miRNAs were significantly differentially expressed between NCX and Ca samples (Table 15). Among these, 129 were down-regulated (Log2 diff (NCX vs Ca) >1) and 41 were up-regulated (Log2 diff (NCX vs Ca) <1) by more than 2-fold in Ca samples compared to NCX samples. Thirty-three miRNAs were down-regulated in Ca samples by more than 5-fold (Log2 diff (NCX vs Ca) >2.3) with a Student t-test p-value < 0.001. Of those, 7 miRNAs (hsa-miR-204, -133b, -1, -133a, -885-5p, -99a, and -143) were down-regulated in Ca samples by more than 10-fold (Log2 diff (NCX vs Ca) >3.3). Among the miRNAs up-regulated in Ca samples, 11 miRNAs (hsa-miR-205, -135b, -182, -31, -31*, - 96, -224, -141, -21*, -183, and -944) were up-regulated by more than 5-fold compared to NCX with a Student t-test p-value ≤0.005. Table 15. miRNAs Differentially Expressed Among Six Cervical Cancer Samples (Ca) and Six Normal Cervix Tissue Samples (NCX) Following Expression Analysis on Agilent Human miRNA Microarrays. Positive Log2Diff (NCX vs Ca) values represent miRNAs down- regulated in cancer samples
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
REFERENCES
The following references, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated herein by reference.
U.S. Patent 4,337,063
U.S. Patent 4,404,289
U.S. Patent 4,405,711
U.S. Patent 4,659,774 U.S. Patent 4,682,195
U.S. Patent 4,683,202
U.S. Patent 4,704,362
U.S. Patent 4,816,571
U.S. Patent 4,959,463 U.S. Patent 5,141,813
U.S. Patent 5,143,854
U.S. Patent 5,202,231
U.S. Patent 5,214,136
U.S. Patent 5,221,619 U.S. Patent 5,223,618
U.S. Patent 5,242,974
U.S. Patent 5,264,566
U.S. Patent 5,268,486
U.S. Patent 5,288,644 U.S. Patent 5,324,633
U.S. Patent 5,378,825
U.S. Patent 5,384,261
U.S. Patent 5,405,783
U.S. Patent 5,412,087 U.S. Patent 5,424,186
U.S. Patent 5,428,148
U.S. Patent 5,429,807
U.S. Patent 5,432,049 U.S. Patent 5,436,327
U.S. Patent 5,445,934
U.S. Patent 5,446,137
U.S. Patent 5,466,786 U.S. Patent 5,468,613
U.S. Patent 5,470,710
U.S. Patent 5,470,967
U.S. Patent 5,472,672
U.S. Patent 5,480,980 U.S. Patent 5,492,806
U.S. Patent 5,503,980
U.S. Patent 5,510,270
U.S. Patent 5,525,464
U.S. Patent 5,527,681 U.S. Patent 5,529,756
U.S. Patent 5,532,128
U.S. Patent 5,545,531
U.S. Patent 5,547,839
U.S. Patent 5,554,501 U.S. Patent 5,554,744
U.S. Patent 5,556,752
U.S. Patent 5,561,071
U.S. Patent 5,571,639
U.S. Patent 5,574,146 U.S. Patent 5,580,726
U.S. Patent 5,580,732
U.S. Patent 5,583,013
U.S. Patent 5,593,839
U.S. Patent 5,599,672 U.S. Patent 5,599,695
U.S. Patent 5,602,240
U.S. Patent 5,602,244
U.S. Patent 5,610,289
U.S. Patent 5,610,287 U.S. Patent 5,614,617
U.S. Patent 5,623,070
U.S. Patent 5,624,711
U.S. Patent 5,631,134 U.S. Patent 5,637,683
U.S. Patent 5,639,603
U.S. Patent 5,645,897
U.S. Patent 5,652,099
U.S. Patent 5,654,413 U.S. Patent 5,658,734
U.S. Patent 5,661,028
U.S. Patent 5,665,547
U.S. Patent 5,667,972
U.S. Patent 5,670,663 U.S. Patent 5,672,697
U.S. Patent 5,681,947
U.S. Patent 5,695,940
U.S. Patent 5,700,637
U.S. Patent 5,700,922 U.S. Patent 5,705,629
U.S. Patent 5,708,154
U.S. Patent 5,714,606
U.S. Patent 5,728,525
U.S. Patent 5,744,305 U.S. Patent 5,763,167
U.S. Patent 5,777,092
U.S. Patent 5,792,847
U.S. Patent 5,800,992
U.S. Patent 5,807,522 U.S. Patent 5,830,645
U.S. Patent 5,837,196
U.S. Patent 5,847,219
U.S. Patent 5,858,988
U.S. Patent 5,859,221 U.S. Patent 5,871,928
U.S. Patent 5,872,232
U.S. Patent 5,876,932
U.S. Patent 5,886,165 U.S. Patent 5,919,626
U.S. Patent 6,004,755
U.S. Patent 6,087,102
U.S. Patent 6,251,666
U.S. Patent 6,368,799 U.S. Patent 6,383,749
U.S. Patent 6,617,112
U.S. Patent 6,638,717
U.S. Patent 6,720,138
U.S. Patent 6,723,509 U.S. Patent 5,770,358
U.S. Patent 5,789,162
U.S. Patent 5,708,153
U.S. Patent 6,040,193
U.S. Patent 5,800,992 U.S. Patent Application serial number 09/545,207
U.S. Patent Application serial number 10/667,126
U.S. Patent Application serial number 11/273,640
ALTS Group (The Atypical Squamous Cells of Undetermined Significance/Low-Grade Squamous Intraepithelial Lesions Triage Study), J Natl. Cancer Inst., 92(5):397-402, 2000.
Bagga et al., Cell, 122(4):553-563, 2005.
Beaucage, and Lyer, Tetrahedron, 48:2223-2311, 1992.
Bentwich et al, Nat Genet., 37(7):766-770, 2005.
Berezikov et α/, Cell, 120(l):21-24, 2005. Bergeron et al, Obstet. Gynecol, 95:821-827, 2000.
Bosch and de Sanjose, Curr. Oncol. Rep., 4(2):175-183, 2002.
Breitling et al, FEBS Letters, 513:83-92, 2004.
Calin and Croce, Nat. Rev. Cancer, 6(l l):857-866, 2006.
Carrington and Ambros, Science, 301(5631):336-338, 2003. Carrington et al. Science, 301(5631):336-338, 2003.
Clifford et al, Br. J. Cancer, 88(l):63-73, 2003
Cummins et al, In: IRT: Nucleosides and nucleosides, La Jolla CA, 72, 1996.
Davison et al, Meth. Enzymol, 411 :14-34, 2006. de Cremoux et al, Amer. J. Clin. Pathol, 120:492-499, 2003.
Denli et al, Trends Biochem. ScL, 28:196, 2003.
EP 266,032
EP 373 203
EP 785 280 EP 799 897
Esquela-Kerscher and Slack, Nat. Rev. Cancer, 6(4):259-269, 2006.
Fodor et al., Science, 251 :767-777 (1991)
Froehler et al, Nucleic Acids Res., 14(13):5399-5407, 1986.
Gillam et al, J. Biol Chem., 253:2532, 1978. Gillam et al , Nucleic Acids Res., 6:2973, 1979.
Gillison et al, J. Natl. Cancer. Inst, 92(9):709-720, 2000.
Griffey et al, J Mass Spectrom, 32(3):305-13, 1997.
Griffiths- Jones et al, Nucleic Acids Res., 34:D140-D144, 2006.
Huber et al, Bioinformatics, 18:Suppl 1 :S96-104, 2002. Itakura and Riggs, Science, 209:1401-1405, 1980.
Itakura et al, J. Biol. Chem., 250:4592, 1975.
Jemal et al, CA Cancer J. Clin., 57:43-66, 2007.
Khorana, Science, 203, 614 1979.
Kornberg and Baker, In: DNA Replication, 2d Ed., Freeman, San Francisco, 1992. Lagos-Quintana et al, Science, 294(5543):853-858, 2001.
Lau et al, Science, 294(5543):858-862, 2001.
Lee and Ambros, Science, 294(5543):862-864, 2001.
Lee et al, EMBOJ. 21 :4663-70, 2002.
Lim et al, Nature, 433(7027):769-773, 2005. Logsdon et al, Cancer Res., 63(10):2649-2657. 2003.
Olsen et al, Dev. Biol, 216:671, 1999.
Parkin et al, CA Cancer J. Clin., 55(2):74-108, 2005.
Pisani et al, Int. J. Cancer, 83:18-29, 1999.
Pisani et al, Int. J. Cancer, 97(1):72-81, 2002. Richart et al, CHn. Obstet. Gynecol, 10:748-784, 1967.
Rose et al, Transplantation, 82(4):570-573, 2006.
Sambrook et al, In: DNA microaarays: a molecular cloning manual, Cold Spring Harbor
Laboratory Press, Cold Spring Harbor, NY, 2003. Sambrook et al, In: Molecular cloning: a laboratory manual, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989.
Sambrook et al., In: Molecular cloning: a laboratory manual, 3 Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 2001.
Seggerson et al, Dev. Biol, 243:215, 2002. Shingara et al, RNA, (9):1461-1470, 2005.
Szafeanska et al, Oncogene, 26(30):4442-4452, 2007.
U.K. Patent 1,529,202
UK 8 803 000
WO 0168255 WO 03020898
WO 03022421
WO 03023058
WO 03029485
WO 03040410 WO 03053586
WO 03066906
WO 03067217
WO 03076928
WO 03087297 WO 03091426
WO 03093810
WO 03100448
WO 04020085
WO 04027093 WO 09923256
WO 09936760
WO 93/17126
WO 95/11995
WO 95/21265 WO 96/31622 WO 97/10365 WO 97/27317 WO 9743450 WO 99/35505 WO 0138580 WO 03100012 Xie, et al, Nature, 434(7031):338-345, 2005. zur Hausen, Nat. Rev. Cancer, 2(5):342-350, 2002.

Claims

1. A method for diagnosing a condition in a patient comprising measuring an expression profile of one or more miRNAs in a sample from a patient suspected of having a precancerous or cancerous cervical condition, wherein a difference in the expression profile in the sample from the patient and an expression profile of a normal sample or a reference is indicative of a pre-cancerous or cancerous condition; wherein one or more miRNA is selected from hsa-let-7a, hsa-let-7b, hsa-let-7c, hsa-let-7d, hsa-let-7e, hsa-let-7f, hsa-let-7g, hsa-let-7i, hsa-miR-1, hsa-miR-lOa, hsa-miR-10b, hsa-miR-100, hsa-miR-101, hsa-miR-106a, hsa-miR- 106b, hsa-miR-124a, hsa-miR-125a, hsa-miR-125b, hsa-miR-126, hsa-miR-126-AS, hsa- miR-127, hsa-miR-130a, hsa-miR-130b, hsa-miR-133a, hsa-miR-134, hsa-miR-135b, hsa- miR-139, hsa-miR-142-5p, hsa-miR-140, hsa-miR-141, hsa-miR-143, hsa-miR-145, hsa-miR- 146a, hsa-miR-149, hsa-miR-150, hsa-miR-152, hsa-miR-153, hsa-miR-154, hsa-miR-155, hsa-miR-15b, hsa-miR-16, hsa-miR-17-5p, hsa-miR-18a, hsa-miR-181a, hsa-miR-181b, hsa- miR-182, hsa-miR-183, hsa-miR-185, hsa-miR-186, hsa-miR-187, hsa-miR-189, hsa-miR- 190, hsa-miR-195, hsa-miR-196a, hsa-miR-196b, hsa-miR-199a, hsa-miR-199a- AS, hsa-miR- 199b, hsa-miR-20a, hsa-miR-200a, hsa-miR-200b, hsa-miR-200c, hsa-miR-203, hsa-miR- 204, hsa-miR-205, hsa-miR-21, hsa-miR-210, hsa-miR-214, hsa-miR-215, hsa-miR-218, hsa- miR-223, hsa-miR-224, hsa-miR-23b, hsa-miR-24, hsa-miR-25, hsa-miR-26a, hsa-miR-26b, hsa-miR-27b, hsa-miR-28, hsa-miR-296, hsa-miR-299-5p, hsa-miR-29a, hsa-miR-29b, hsa- miR-29c, hsa-miR-302d, hsa-miR-3Oa-3p, hsa-miR-30a-5p, hsa-miR-30b, hsa-miR-30d, hsa- miR-31, hsa-miR-320, hsa-miR-324-3p, hsa-miR-325, hsa-miR-328, hsa-miR-330, hsa-miR- 335, hsa-miR-339, hsa-miR-361, hsa-miR-365, hsa-miR-368, hsa-miR-370, hsa-miR-373-AS, hsa-miR-376a, hsa-miR-377, hsa-miR-379, hsa-miR-381, hsa-miR-382, hsa-miR-423, hsa- miR-424, hsa-miR-429, hsa-miR-432, hsa-miR-450, hsa-miR-491, hsa-miR-492, hsa-miR- 495, hsa-miR-500, hsa-miR-501, hsa-miR-505, hsa-miR-513, hsa-miR-515-5p, hsa-miR- 518b, hsa-miR-520d, hsa-miR-92, hsa-miR-93, hsa-miR-95, hsa-miR-98, hsa-miR-99a, hsa- miR-99b, ambi-miR-7029, hsa-miR-498, ambi-miR-7062, ambi-miR-7066, ambi-miR-7068- 1, ambi-miR-7070, hsa-miR-497, ambi-miR-7074, ambi-miR-7075, ambi-miR-7079, ambi- miR-7083, ambi-miR-7085, ambi-miR-7100, ambi-miR-7101, hsa-miR-133b, hsa-miR-455, hsa-asg-5021_stl, hsa-asg-13254_stl, hsa-asg-14176_stl, hsa-miR-487b, hsa-miR-411, hsa- miR-574, hsa-miR-542-5p, hsa-asg-5617_stl, hsa-asg-14172_stl, hsa-asg-13304_st2, hsa- asg-13297_stl, hsa-miR157_st2, hsa-cand317_stl, hsa-miR-329, hsa-asg-10202_st2, hsa- miR-369-5p, hsa-asg-13284_stl, hsa-asg-9687_stl, hsa-miR-433, hsa-miR-565, hsa-asg- 562_stl, hsa-asg-279_st2, hsa-asg-8411_st2, hsa-asg-7472_st2, hsa-asg-13279_stl, hsa-miR- 487a, hsa-cand206_stl, hsa-cand345_stl, hsa-asg-9696_stl, hsa-miR-485-3p, hsa-asg- 13166_st2, hsa-miR-594_st2, hsa-miR-299-3p, hsa-asg-924_stl, hsa-miR-539, hsa-asg- 10883_stl, hsa-miR-585, hsa-miR-493-5p, hsa-asg-12964_st2, hsa-asg-4557_st2, hsa-asg- 10674_stl, hsa-asg-14230_stl, hsa-asg-9681_stl, hsa-miR-628, hsa-asg-13237_stl, hsa-asg- 13230_st2, hsa-miR-493-3p, hsa-miR-654, hsa-asg-2919_stl, hsa-asg-8067_st2, hsa-asg- 11199_st2, hsa-asg-12346_st2, hsa-asg-11883_stl, hsa-miR102_st2, hsa-asg-2027_stl, hsa- asg-3711_st2, hsa-asg-3145_stl, hsa-asg-7465_st2, hsa-asg-3376_stl, hsa-cand283_stl, hsa- asg-2301_st2, hsa-cand207_stl, hsa-miR-598, hsa-asg-10278_st2, hsa-miR-18b, hsa-asg- 11181_stl, hsa-asg-7023_st2, hsa-asg-3597_st2, hsa-asg-5304_stl, hsa-asg-11688_stl, hsa- cand720_stl, hsa-asg-13308_st2, hsa-asg-3038_st2, hsa-asg-13966_st2, hsa-asg-13189_stl, hsa-asg-11938_stl, hsa-asg-5740_st2, hsa-asg-8477_stl, hsa-asg-12325_st2, hsa-asg- 12356_stl, hsa-asg-6758_stl, hsa-asg-522_stl, hsa-asg-4564_st2, hsa-asg-6951_st2, hsa-asg- 9920_stl, hsa-asg-13613_st2, hsa-miR-184, hsa-miR-503, hsa-miR-485-5p, hsa-miR-494, hsa-miR-504, hsa-miR-211, hsa-miR-99b, hsa-miR-499, hsa-miR-422b, hsa-miR-338, hsa- miR-422a, hsa-miR-331, hsa-miR-489, hsa-miR-324-5p, hsa-miR-151, hsa-miR-17-3p, hsa- miR-340, , hsa-miR-194, hsa-let-7d*, hsa-let-7e*, hsa-miR-100*, hsa-miR-101*, hsa-miR- 106b*, hsa-miR-lOb*, hsa-miR-125a-5p, hsa-miR-125b-2*, hsa-miR-126*, hsa-miR-127-3p, hsa-miR-129-3p, hsa-miR-135a, hsa-miR-136, hsa-miR-136*, hsa-miR-139-5p, hsa-miR-140- 3p, hsa-miR-140-5p, hsa-miR-141*, hsa-miR-142-3p, hsa-miR-143*, hsa-miR-144, hsa-miR- 144*, hsa-miR-145*, hsa-miR-146b-5p, hsa-miR-151-3p, hsa-miR-154*, hsa-miR-15b*, hsa- miR-17, hsa-miR-181c, hsa-miR-181d, hsa-miR-183*, hsa-miR-193a-5ρ, hsa-miR-195*, hsa- miR-197, hsa-miR-199a-5p, hsa-miR-199b-3p, hsa-miR-199b-5p, hsa-miR-20a*, hsa-miR- 21*, hsa-raiR-212, hsa-miR-214*, hsa-miR-24-1*, hsa-miR-26b*, hsa-miR-28-3p, hsa-miR- 28-5p, hsa-miR-29a*, hsa-miR-29b-l*, hsa-miR-29b-2*, hsa-miR-29c*, hsa-miR-30a, hsa- miR-30a*, hsa-miR-30b*, hsa-miR-30c-2*, hsa-miR-31*, hsa-miR-32, hsa-miR-330-3p, hsa- miR-337-3ρ, hsa-miR-337-5p, hsa-miR-345, hsa-miR-361-5p, hsa-miR-374a, hsa-miR-374b, hsa-miR-374b*, hsa-miR-375, hsa-miR-376b, hsa-miR-376c, hsa-miR-377*, hsa-miR-410, hsa-miR-424*, hsa-miR-425, hsa-miR-431*, hsa-miR-450a, hsa-miR-451 , hsa-miR-455-3p, hsa-miR-455-5p, hsa-miR-483-3p, hsa-miR-486-5p, hsa-miR-488, hsa-miR-490-3p, hsa-miR- 499-5ρ, hsa-miR-501-5p, hsa-miR-505*, hsa-miR-512-3p, hsa-miR-513c, hsa-miR-517*, hsa- miR-542-3p, hsa-miR-543, hsa-miR-574-3ρ, hsa-miR-602, hsa-miR-628-5p, hsa-miR-629*, hsa-miR-630, hsa-miR-650, hsa-miR-654-3p, hsa-miR-654-5p, hsa-miR-655, hsa-miR-656, hsa-miR-744, hsa-miR-766, hsa-miR-768-3p, hsa-miR-873, hsa-miR-885-5ρ, hsa-miR-886- 3p, hsa-miR-886-5p, hsa-miR-889, hsa-miR-93*, hsa-miR-940, hsa-miR-944, hsa-miR-96, hsa-miR-99a*, or hsa-miR-99b*.
2. The method of claim 1, wherein the miRNA is one or more of hsa-miR-1, hsa-miR- 100, hsa-miR-133a, hsa-miR-134, hsa-miR-143, hsa-miR-154, hsa-miR-182, hsa-miR-183, hsa-miR-195, hsa-miR-204, hsa-miR-205, hsa-miR-21, hsa-miR-218, hsa-miR-224, hsa-miR- 299-5p, hsa-miR-31, hsa-miR-368, hsa-miR-376a, hsa-miR-424, hsa-miR-99a, ambi-miR- 7029, hsa-miR-497, ambi-miR-7101, hsa-miR-133b, hsa-miR-455, hsa-asg-5021_stl, hsa- asg-13254_stl, hsa-asg-14176_stl, hsa-miR-487b, hsa-miR-411, hsa-miR-574, hsa-miR-542- 5p, hsa-asg-5617_stl, hsa-asg-14172_stl, hsa-asg-13304_st2, hsa-asg-13297_stl, hsa- miR157_st2, hsa-cand317_stl, hsa-miR-329, hsa-asg-10202_st2, hsa-miR-369-5p, hsa-asg- 13284_stl, hsa-asg-9687_stl, hsa-miR-433, hsa-miR-565, hsa-asg-562_stl, hsa-asg-279_st2, hsa-asg-8411_st2, hsa-asg-7472_st2, hsa-asg-13279_stl, hsa-miR-487a, hsa-cand206_stl, hsa-cand345_stl, hsa-asg-9696_stl, hsa-miR-485-3p, hsa-asg-13166_st2, hsa-miR-594_st2, hsa-miR-299-3p, hsa-asg-924_stl, hsa-miR-539, hsa-asg-10883_stl, hsa-miR-184, hsa-miR- 503, hsa-miR-485-5ρ, hsa-miR-494, hsa-miR-504, hsa-miR-211, hsa-miR-99b, hsa-miR-495, hsa-miR-381, hsa-miR-127, hsa-miR-124a, hsa-miR-432, hsa-mir-214, hsa-miR-379, hsa- miR-189, hsa-miR-130a, hsa-miR-382, hsa-miR-885-5p, hsa-miR-145*, hsa-miR-517*, hsa- miR-139-5ρ, hsa-miR-145, hsa-miR-376c, hsa-miR-135b, hsa-miR-136*, hsa-miR-99a*, hsa- miR-125b-2*, hsa-miR-31*, hsa-miR-125b, hsa-miR-96, hsa-miR-451, hsa-miR-143*, hsa- miR-135a, hsa-miR-141, hsa-miR-375, hsa-miR-21*, hsa-miR-654-3p, hsa-miR-455-5p, hsa- miR-337-3ρ, hsa-miR-886-5p, hsa-miR-410, hsa-miR-944, or hsa-miR-337-5ρ.
3. The method of claim 2, wherein the miRNA is hsa-miR-1, hsa-miR-21 or both hsa- miR-1 and hsa-miR-21.
4. The method of claim 1, wherein differential expression of one or more miRNA is indicative of cervical carcinoma, wherein the miRNA is one or more of hsa-let-7a, hsa-let-7b, hsa-let-7c, hsa-let-7d, hsa-let-7e, hsa-let-7f, hsa-let-7g, hsa-let-7i, hsa-miR-1, hsa-miR-1 Oa, hsa-miR-10b, hsa-miR-100, hsa-miR-101, hsa-miR-106a, hsa-miR-106b, hsa-miR-124a, hsa- miR-125a, hsa-miR-125b, hsa-miR-126, hsa-miR-126-AS, hsa-miR-127, hsa-miR-130a, hsa- miR-130b, hsa-miR-133a, hsa-miR-134, hsa-miR-135b, hsa-miR-139, hsa-miR-140, hsa- miR-141, hsa-miR-143, hsa-miR-145, hsa-miR-146a, hsa-miR-149, hsa-miR-150, hsa-miR- 152, hsa-miR-153, hsa-miR-154, hsa-miR-155, hsa-miR-15b, hsa-miR-16, hsa-miR-17-5p, hsa-miR-18a, hsa-miR-181a, hsa-miR-181b, hsa-miR-182, hsa-miR-183, hsa-miR-185, hsa- miR-186, hsa-miR-187, hsa-miR-189, hsa-miR-190, hsa-miR-195, hsa-miR-196b, hsa-miR- 199a, hsa-miR-199a-AS, hsa-miR-199b, hsa-miR-20a, hsa-miR-200a, hsa-miR-200b, hsa- miR-200c, hsa-miR-203, hsa-miR-204, hsa-miR-205, hsa-miR-21, hsa-miR-210, hsa-miR- 214, hsa-miR-215, hsa-miR-218, hsa-miR-223, hsa-miR-224, hsa-miR-23b, hsa-miR-24, hsa- miR-25, hsa-miR-26a, hsa-miR-26b, hsa-miR-27b, hsa-miR-28, hsa-miR-296, hsa-miR-299- 5p, hsa-miR-29a, hsa-miR-29b, hsa-miR-29c, hsa-miR-302d, hsa-miR-30a-3p, hsa-miR-30a- 5p, hsa-miR-30b, hsa-miR-30d, hsa-miR-31, hsa-miR-320, hsa-miR-324-3p, hsa-miR-328, hsa-miR-330, hsa-miR-335, hsa-miR-339, hsa-miR-361, hsa-miR-365, hsa-miR-368, hsa- miR-370, hsa-miR-373-AS, hsa-miR-376a, hsa-miR-377, hsa-miR-379, hsa-miR-381, hsa- miR-382, hsa-miR-423, hsa-miR-424, hsa-miR-429, hsa-miR-432, hsa-miR-450, hsa-miR- 491, hsa-miR-492, hsa-miR-495, hsa-miR-501, hsa-miR-505, hsa-miR-515-5p, hsa-miR- 518b, hsa-miR-520d, hsa-miR-92, hsa-miR-93, hsa-miR-95, hsa-miR-98, hsa-miR-99a, hsa- miR-99b, ambi-miR-7029, hsa-miR-498, ambi-miR-7062, ambi-miR-7066, ambi-miR-7068- 1, ambi-miR-7070, hsa-miR-497, ambi-miR-7074, ambi-miR-7075, ambi-miR-7079, ambi- miR-7083, ambi-miR-7085, ambi-miR-7100, ambi-miR-7101, hsa-miR-133b, hsa-miR-455, hsa-asg-5021_stl, hsa-asg-13254_stl, hsa-asg-14176_stl, hsa-miR-487b, hsa-miR-411, hsa- miR-574, hsa-miR-542-5p, hsa-asg-5617_stl, hsa-asg-14172_stl, hsa-asg-13304_st2, hsa- asg-13297_stl, hsa-miR157_st2, hsa-cand317_stl, hsa-miR-329, hsa-asg-10202_st2, hsa- miR-369-5p, hsa-asg-13284_stl, hsa-asg-9687_stl, hsa-miR-433, hsa-miR-565, hsa-asg- 562_stl, hsa-asg-279_st2, hsa-asg-8411_st2, hsa-asg-7472_st2, hsa-asg-13279_stl, hsa-miR- 487a, hsa-cand206_stl, hsa-cand345_stl, hsa-asg-9696_stl, hsa-miR-485-3p, hsa-asg- 13166_st2, hsa-miR-594_st2, hsa-miR-299-3p, hsa-asg-924_stl, hsa-miR-539, hsa-asg- 10883_stl, hsa-miR-585, hsa-miR-493-5p, hsa-asg-12964_st2, hsa-asg-4557_st2, hsa-asg- 10674_stl, hsa-asg-14230_stl, hsa-asg-9681_stl, hsa-miR-628, hsa-asg-13237_stl, hsa-asg- 13230_st2, hsa-miR-493-3p, hsa-miR-654, hsa-asg-2919_stl, hsa-asg-8067_st2, hsa-asg- 11199_st2, hsa-asg-12346_st2, hsa-asg-11883_stl, hsa-miR102_st2, hsa-asg-2027_stl, hsa- asg-3711_st2, hsa-asg-3145_stl, hsa-asg-7465_st2, hsa-asg-3376_stl, hsa-cand283_stl, hsa- asg-2301_st2, hsa-cand207_stl, hsa-miR-598, hsa-asg-10278_st2, hsa-miR-18b, hsa-asg- 11181_stl, hsa-asg-7023_st2, hsa-asg-3597_st2, hsa-asg-5304_stl, hsa-asg-11688_stl, hsa- cand720_stl, hsa-asg-13308_st2, hsa-asg-3038_st2, hsa-asg-13966_st2, hsa-asg-13189_stl, hsa-asg-11938_stl, hsa-asg-5740_st2, hsa-asg-8477_stl, hsa-asg-12325_st2, hsa-asg- 12356_stl, hsa-asg-6758_stl, hsa-asg-522_stl, hsa-asg-4564_st2, hsa-asg-6951_st2, hsa-asg- 9920_stl, hsa-asg-13613_st2, hsa-miR-184, hsa-miR-503, hsa-miR-485-5p, hsa-miR-494, hsa-miR-504, hsa-miR-211, hsa-miR-99b, hsa-miR-499, hsa-miR-422b, hsa-miR-338, hsa- miR-422a, hsa-miR-331, hsa-miR-489, hsa-miR-324-5p, hsa-miR-151, hsa-miR-17-3p, hsa- miR-340, hsa-miR-194, hsa-let-7d*, hsa-let-7e*, hsa-miR-100*, hsa-miR-101*, hsa-miR- 106b*, hsa-miR-10b*, hsa-miR-125a-5p, hsa-miR-125b-2*, hsa-miR-126*, hsa-miR-127-3p, hsa-miR-129-3p, hsa-miR-135a, hsa-miR-136, hsa-miR-136*, hsa-miR-139-5p, hsa-miR-140- 3p, hsa-miR-140-5p, hsa-miR-141*, hsa-miR-142-3p, hsa-miR-142-5p, hsa-miR-143*, hsa- miR-144, hsa-miR-144*, hsa-miR-145*, hsa-miR-146b-5p, hsa-miR-151-3ρ, hsa-miR-154*, hsa-miR-15b*, hsa-miR-17, hsa-miR-181c, hsa-miR-181d, hsa-miR-183*, hsa-miR-193a-5p, hsa-miR-195*, hsa-miR-197, hsa-miR-199a-5p, hsa-miR-199b-3ρ, hsa-miR-199b-5p, hsa- miR-20a*, hsa-miR-21*, hsa-miR-212, hsa-miR-214*, hsa-miR-24-1*, hsa-miR-26b*, hsa- miR-28-3p, hsa-miR-28-5p, hsa-miR-29a*, hsa-miR-29b-l*, hsa-miR-29b-2*, hsa-miR-29c*, hsa-miR-30a, hsa-miR-30a*, hsa-miR-30b*, hsa-miR-3 Oc-2*, hsa-miR-31 *, hsa-miR-32, hsa- miR-330-3p, hsa-miR-337-3p, hsa-miR-337-5p, hsa-miR-345, hsa-miR-36 l-5p, hsa-miR- 374a, hsa-miR-374b, hsa-miR-374b*, hsa-miR-375, hsa-miR-376b, hsa-miR-376c, hsa-miR- 377*, hsa-miR-410, hsa-miR-424*, hsa-miR-425, hsa-miR-431*, hsa-miR-450a, hsa-miR- 451, hsa-miR-455-3ρ, hsa-miR-455-5p, hsa-miR-483-3ρ, hsa-miR-486-5p, hsa-miR-488, hsa- miR-490-3p, hsa-miR-499-5p, hsa-miR-501-5ρ, hsa-miR-505*, hsa-miR-512-3ρ, hsa-miR- 513c, hsa-miR-517*, hsa-miR-542-3p, hsa-miR-543, hsa-miR-574-3p, hsa-miR-602, hsa- miR-628-5p, hsa-miR-629*, hsa-miR-630, hsa-miR-650, hsa-miR-654-3p, hsa-miR-654-5p, hsa-miR-655, hsa-miR-656, hsa-miR-744, hsa-miR-766, hsa-miR-768-3p, hsa-miR-873, hsa- miR-885-5p, hsa-miR-886-3ρ, hsa-miR-886-5p, hsa-miR-889, hsa-miR-93*, hsa-miR-940, hsa-miR-944, hsa-miR-96, hsa-miR-99a*, or hsa-miR-99b*.
5. The method of claim 1, wherein differential expression of one or more miRNA is indicative of cervical intraepithelial neoplasia or cervical squamous intraepithelial lesions, wherein the miRNA is one or more of hsa-miR-1, hsa-miR-133a, hsa-miR-124a, hsa-miR- 187, hsa-miR-204, hsa-miR-145, hsa-miR-143, hsa-miR-325, hsa-miR-500, hsa-miR-196a, hsa-miR-125a, hsa-miR-376a, hsa-miR-505, hsa-miR-100, hsa-miR-99a, hsa-miR-141, hsa- miR-200a, ambi-miR-7029, hsa-miR-223, hsa-miR-205, hsa-miR-146a, hsa-miR-429, hsa- miR-200b, hsa-miR-182, hsa-miR-142-5p, hsa-miR-203, hsa-miR-21, hsa-miR-31, or hsa- miR-513.
6. The method of claim 1 , wherein reduced expression of an miRNA in a sample relative to normal or normal adjacent tissue is indicative of a cancerous condition, wherein the miRNA is one or more of hsa-let-7a, hsa-let-7b, hsa-let-7c, hsa-let-7d, hsa-let-7e, hsa-let-7f, hsa-let-7g, hsa-let-7i, hsa-miR-1, hsa-miR-10a, hsa-miR-10b, hsa-miR-100, hsa-miR-101, hsa-miR-124a, hsa-miR-125a, hsa-miR-125b, hsa-miR-126, hsa-miR-126-AS, hsa-miR-127, hsa-miR-130a, hsa-miR-133a, hsa-miR-134, hsa-miR-139, hsa-miR-142-5p, hsa-miR-140, hsa-miR-143, hsa-miR-145, hsa-miR-149, hsa-miR-152, hsa-miR-154, hsa-miR-186, hsa- miR-189, hsa-miR-190, hsa-miR-195, hsa-miR-196a, hsa-miR-196b, hsa-miR-199a, hsa-miR- 199a-AS, hsa-miR-199b, hsa-miR-204, hsa-miR-214, hsa-miR-215, hsa-miR-218, hsa-miR- 23b, hsa-miR-24, hsa-miR-26a, hsa-miR-26b, hsa-miR-27b, hsa-miR-28, hsa-miR-299-5p, hsa-miR-29a, hsa-miR-29b, hsa-miR-29c, hsa-miR-3Oa-3p, hsa-miR-30a-5p, hsa-miR-30b, hsa-miR-30d, hsa-miR-320, hsa-miR-324-3p, hsa-miR-325, hsa-miR-328, hsa-miR-335, hsa- miR-361, hsa-miR-365, hsa-miR-368, hsa-miR-376a, hsa-miR-377, hsa-miR-379, hsa-miR- 381, hsa-miR-382, hsa-miR-424, hsa-miR-432, hsa-miR-450, hsa-miR-495, hsa-miR-500, hsa-miR-505, hsa-miR-513, hsa-miR-98, hsa-miR-99a, hsa-miR-99b, ambi-miR-7029, ambi- miR-7066, ambi-miR-7068-1, ambi-miR-7070, hsa-miR-497, ambi-miR-7075, ambi-miR- 7083, ambi-miR-7085, ambi-miR-7100, ambi-miR-7101, hsa-miR-133b, hsa-miR-455, hsa- asg-5021_stl, hsa-asg-13254_stl, hsa-asg-14176_stl, hsa-miR-487b, hsa-miR-411, hsa-miR- 574, hsa-miR-542-5p, hsa-asg-5617_stl, hsa-asg-14172_stl, hsa-asg-13304_st2, hsa-asg- 13297_stl, hsa-miR157_st2, hsa-miR-329, hsa-asg-10202_st2, hsa-miR-369-5p, hsa-asg- 13284_stl, hsa-asg-9687_stl, hsa-miR-433, hsa-miR-565, hsa-asg-562_stl, hsa-asg-279_st2, hsa-asg-8411_st2, hsa-asg-7472_st2, hsa-asg-13279_stl, hsa-miR-487a, hsa-cand206_stl, hsa-cand345_stl, hsa-asg-9696_stl, hsa-miR-485-3p, hsa-asg-13166_st2, hsa-miR-594_st2, hsa-miR-299-3p, hsa-miR-539, hsa-asg-10883_stl, hsa-miR-585, hsa-miR-493-5ρ, hsa-asg- 12964_st2, hsa-asg-4557_st2, hsa-asg-10674_stl, hsa-asg-14230_stl, hsa-asg-9681_stl, hsa- miR-628, hsa-asg-13237_stl, hsa-asg-13230_st2, hsa-miR-493-3p, hsa-miR-654, hsa-asg- 33_stl, hsa-asg-8067_st2, hsa-asg-11199_st2, hsa-asg-12346_st2, hsa-asg-11883_stl, hsa- miR102_st2, hsa-asg-2027_stl, hsa-asg-3145_stl, hsa-asg-7465_st2, hsa-asg-3376_stl, hsa- cand283_stl, hsa-asg-2301_st2, hsa-cand207_stl, hsa-miR-598, hsa-asg-10278_st2, hsa-asg- 11181_stl, hsa-asg-7023_st2, hsa-asg-3597_st2, hsa-asg-5304_stl, hsa-cand720_stl, hsa-asg- 13308_st2, hsa-asg-3038_st2, hsa-asg-13966_st2, hsa-asg-13189_stl, hsa-asg-11938_stl, hsa-asg-5740_st2, hsa-asg-8477_stl, hsa-asg-12325_st2, hsa-asg-12356_stl, hsa-asg- 6758_stl, hsa-asg-522_stl, hsa-asg-4564_st2, hsa-asg-6951_st2, hsa-asg-9920_stl, hsa-asg- 13613_st2, hsa-miR-184, hsa-miR-503, hsa-miR-485-5p, hsa-miR-494, hsa-miR-504, hsa- miR-211, hsa-miR-99b, hsa-miR-499, hsa-miR-422b, hsa-miR-338, hsa-miR-422a, hsa-miR- 331, hsa-miR-489, hsa-miR-324-5p, hsa-miR-151, hsa-miR-17-3p, hsa-miR-340, hsa-miR- 194, hsa-miR-885-5p, hsa-miR-145*, hsa-miR-517*, hsa-miR-139-5ρ, hsa-miR-376c, hsa- miR-136*, hsa-miR-99a*, hsa-miR-125b-2*, hsa-miR-451, hsa-miR-143*, hsa-miR-135a, hsa-miR-375, hsa-miR-654-3p, hsa-miR-455-5p, hsa-miR-337-3p, hsa-miR-886-5p, hsa-miR- 410, hsa-miR-337-5p, or a combination thereof.
7. The method of claim 1, wherein increased expression of an miRNA in a sample relative to normal or normal adjacent tissue is indicative of a cancerous condition, wherein the miRNA is one or more of hsa-miR-106a, hsa-miR-106b, hsa-miR-130b, hsa-miR-135b, hsa- miR-205, hsa-miR-141, hsa-miR-182, hsa-miR-146a, hsa-miR-150, hsa-miR-155, hsa-miR- 15b, hsa-miR-16, hsa-miR-17-5p, hsa-miR-18a, hsa-miR-181a, hsa-miR-181b, hsa-miR-183, hsa-miR-185, hsa-miR-20a, hsa-miR-200a, hsa-miR-200b, hsa-miR-200c, hsa-miR-203, hsa- miR-21, hsa-miR-210, hsa-miR-223, hsa-miR-224, hsa-miR-25, hsa-miR-31, hsa-miR-330, hsa-miR-339, hsa-miR-423, hsa-miR-429, hsa-miR-492, hsa-miR-92, hsa-miR-93, hsa-miR- 518b, hsa-miR-520d, hsa-miR-491, hsa-miR-501, hsa-miR-515-5ρ, ambi-miR-7062, ambi- miR-7074, ambi-miR-7079, hsa-asg-11688_stl, hsa-miR-18b, hsa-miR-18a, hsa-asg- 3711_st2, hsa-asg-2919_stl, hsa-asg-924_stl, hsa-cand317_stl, hsa-miR-31*, hsa-miR-96, hsa-miR-21*, hsa-miR-944, or a combination thereof.
8. The method of claim 1 , wherein reduced expression of an miRNA in a sample relative to normal or normal adjacent tissue is indicative of a pre-cancerous condition, wherein the miRNA is one or more of hsa-miR-1, hsa-miR-133a, hsa-miR-124a, hsa-miR-187, hsa-miR- 204, hsa-miR-143, hsa-miR-145, hsa-miR-325, hsa-miR-99a, hsa-miR-100, hsa-miR-376a, hsa-miR-505, hsa-miR-125a, hsa-miR-196a, hsa-miR-500 or a combination thereof.
9. The method of claim 1, wherein increased expression of an miRNA in a sample relative to normal or normal adjacent tissue is indicative of a pre-cancerous condition, wherein the miRNA is one or more of hsa-miR-205, hsa-miR-182, hsa-miR-141, hsa-miR- 142-5p, hsa-miR200a, hsa-miR-200b, hsa-miR-223, hsa-miR-21, hsa-miR-203, hsa-miR-31, ambi-miR-7029, hsa-miR-146a, hsa-miR-429, hsa-miR-513 or a combination thereof.
10. The method of claim 1, wherein the sample is isolated RNA, fresh tissue or cells, frozen tissue or cells, fixed tissue or cells, or embedded tissue or cells.
11. The method of claim 1 , wherein the sample from the patient and the normal sample are cervix samples.
12. The method of claim 1, 8, or 9 wherein the pre-cancerous condition is cervical squamous intraepithelial lesion or cervical intraepithelial neoplasia.
13. The method of claim 1, 6, or 7 wherein the cancerous condition is cervical squamous cell carcinoma.
14. The method of claim 1, further comprising obtaining a sample from the patient.
15. The method of claim 14, further comprising labeling miRNA from the sample.
16. The method of claim 15, further comprising hybridizing the labeled miRNA to one or more miRNA probes.
17. The method of claim 16, wherein the miRNA probes are coupled to a support.
18. The method of claim 17, wherein the support is glass, plastic, metal, or latex.
19. The method of claim 18, wherein the support is planar.
20. The method of claim 18, wherein the support is a bead.
21. The method of claim 1, wherein expression of the miRNA is determined by an amplification assay or a hybridization assay.
22. The method of claim 21, wherein the miRNA amplified is all or part of one or more of hsa-miR-1, hsa-miR-15b, hsa-miR-133a, hsa-miR-143, hsa-miR-205, hsa-miR-21, hsa-miR- 204, hsa-miR-195, hsa-miR-100, hsa-miR-99a, hsa-miR-368, hsa-miR-183 or combination thereof.
23. The method of claim 21, wherein the miRNA amplified is all or part of hsa-miR-1, hsa-miR-21, or hsa-miR-1 and hsa-miR-21,
24. The method of claim 21, wherein amplification assay is a quantitative amplification assay.
25. The method of claim 24, wherein the quantitative amplification assay is quantitative RT-PCR.
26. The method of claim 21, wherein the hybridization assay is an array hybridization assay or a solution hybridization assay.
27. The method of claim 1 , wherein diagnosing is screening for a pathological condition, assessing prognosis of a pathological condition, staging a pathological condition, or assessing response of a pathological condition to therapy.
28. A method of detecting a cervical cell comprising evaluating the expression of an miRNA selected from one or more of hsa-let-7a, hsa-let-7b, hsa-let-7c, hsa-let-7i, hsa-miR- 100, hsa-miR-101, hsa-miR-106a, hsa-miR-125a, hsa-miR-125b, hsa-miR-126-AS, hsa-miR- 127, hsa-miR-130a, hsa-miR-134, hsa-miR-142-3p, hsa-miR-143, hsa-miR-145, hsa-miR- 148b, hsa-miR-149, hsa-miR-151, hsa-miR-152, hsa-miR-154, hsa-miR-15a, hsa-miR-15b, hsa-miR-16, hsa-miR-17-5p, hsa-miR-181a, hsa-miR-185, hsa-miR-186, hsa-miR-187, hsa- miR-192, hsa-miR-193a, hsa-miR-195, hsa-miR-196a, hsa-miR-196b, hsa-miR-199a, hsa- miR-199a-AS, hsa-miR-199b, hsa-miR-19a, hsa-miR-20a, hsa-miR-203, hsa-miR-205, hsa- miR-21, hsa-miR-214, hsa-miR-215, hsa-miR-22, hsa-miR-221, hsa-miR-223, hsa-miR-23b, hsa-miR-24, hsa-miR-25, hsa-miR-26a, hsa-miR-26b, hsa-miR-299-5p, hsa-miR-29b, hsa- miR-29c, hsa-miR-302c-AS, hsa-miR-30a-3p, hsa-miR-30a-5p, hsa-miR-30b, hsa-miR-30c, hsa-miR-30e-3p, hsa-miR-30e-5p, hsa-miR-31, hsa-miR-320, hsa-miR-324-3p, hsa-miR-335, hsa-miR-338, hsa-miR-342, hsa-miR-34a, hsa-miR-361, hsa-miR-365, hsa-miR-367, hsa- miR-368, hsa-miR-370, hsa-miR-374, hsa-miR-376a, hsa-miR-379, hsa-miR-381, hsa-miR- 423, hsa-miR-424, hsa-miR-450, hsa-miR-7, hsa-miR-93, hsa-miR-95, hsa-miR-96, hsa-miR- 99a, hsa-miR-99b, ambi-miR-7027, ambi-miR-7029, hsa-miR-509, hsa-miR-193b, ambi-miR- 7039, hsa-miR-526b, hsa-miR-498, hsa-miR-452, ambi-miR-7062, ambi-miR-7070, hsa-miR- 497, ambi-miR-7079, ambi-miR-7083, ambi-miR-7085, hsa-miR-503, or ambi-miR-7101 or complements thereof.
29. A kit for analysis of a pathological sample by assessing miRNA profile for a sample comprising, in suitable container means, two or more miRNA hybridization or amplification reagents comprising one or more of the miRNA listed in Table 1.
30. The kit of claim 29, further comprising reagents for labeling miRNA in the sample.
31. The kit of claim 30, wherein the labeling reagents include at least one amine-modified nucleotide, poly(A) polymerase, and poly(A) polymerase buffer.
32. The kit of claim 31 , wherein the labeling reagents include an amine-reactive dye.
33. The kit of claim 29, wherein miRNA hybridization reagent comprises hybridization probes.
34. The kit of claim 29, wherein miRNA amplification reagent comprises amplification primers.
35. A method of treating a cancerous condition by contacting a cervical cell with one or more nucleic acid comprising a miRNA sequence, wherein expression of an endogenous miRNA is modulated in the cervical cell; where the miRNA sequence is at least 85% identical to one or more of miRNA listed in claim 1.
36. The method of claim 35, wherein the cervical cell is a cervical cancer cell.
37. The method of claim 35, further comprising administering a second therapy.
38. The method of claim 37, wherein the second therapy is chemotherapy, radiotherapy, surgery, or immunotherapy.
PCT/US2008/076246 2007-09-14 2008-09-12 Micrornas differentially expressed in cervical cancer and uses thereof WO2009036332A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP08831073A EP2198050A1 (en) 2007-09-14 2008-09-12 Micrornas differentially expressed in cervical cancer and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US97264607P 2007-09-14 2007-09-14
US60/972,646 2007-09-14

Publications (1)

Publication Number Publication Date
WO2009036332A1 true WO2009036332A1 (en) 2009-03-19

Family

ID=40040003

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/076246 WO2009036332A1 (en) 2007-09-14 2008-09-12 Micrornas differentially expressed in cervical cancer and uses thereof

Country Status (3)

Country Link
US (2) US8361714B2 (en)
EP (1) EP2198050A1 (en)
WO (1) WO2009036332A1 (en)

Cited By (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101851682A (en) * 2010-06-25 2010-10-06 中国人民解放军南京军区南京总医院 MiRNA combination used for detecting esophageal squamous cell carcinoma and application thereof
WO2010139810A1 (en) * 2009-06-05 2010-12-09 Febit Holding Gmbh Mirna fingerprint in the diagnosis of lung cancer
CN101921759A (en) * 2010-09-08 2010-12-22 南京医科大学 Serum/plasma miRNA serum marker related to cervical carcinoma and precancerous lesions thereof and application thereof
WO2011014980A1 (en) * 2009-08-07 2011-02-10 Capitalbio Corporation Methods and compositions diagnosing cervical cancer and cervical dysplasia, guidding subsequent treatment, determining prognosis, and improving patient survival
EP2283846A1 (en) * 2009-08-12 2011-02-16 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. miRNA compounds for treatment of prostate carcinoma
WO2011026909A1 (en) * 2009-09-02 2011-03-10 L'oreal Epidermal differentiation microrna signature and uses thereof
WO2010083464A3 (en) * 2009-01-16 2011-05-12 Cepheid Methods of detecting cervical cancer
WO2011149354A1 (en) * 2010-05-28 2011-12-01 Vereniging Voor Christelijk Hoger Onderwijs, Wetenschappelijk Onderzoek En Patiëntenzorg Mirnas involved in the blood brain barrier function
WO2012025709A1 (en) * 2010-08-23 2012-03-01 Sistemic Uk Cell characterisation
CN102443650A (en) * 2011-12-20 2012-05-09 苏州福英基因科技有限公司 In-situ hybridization detection kit for MICRORNA (MICRO Ribonucleic Acid)-233 level at pathologic evolution early stage of colon cancer as well as microRNA-233 in-situ hybridization detection method and application of microRNA-233 to preparation of in-situ hybridization detection kit for colon cancer
CN102510905A (en) * 2009-08-07 2012-06-20 博奥生物有限公司 Methods and compositions for the diagnosis and prognosis of cervical intraepithelial neoplasia and cervical cancer
CN102533987A (en) * 2011-12-20 2012-07-04 苏州福英基因科技有限公司 Horizontal in-situ hybridization detection kit and detection method as well as application for MICRORNA-143 in earlier stage of cancer pathologic evolution
CN102533977A (en) * 2011-12-15 2012-07-04 苏州福英基因科技有限公司 Kit and method for detecting horizontal in situ hybridization of MICRORNA-372 at the early pathological evolution stage of a variety of cancers and application thereof
CN102533978A (en) * 2011-12-15 2012-07-04 苏州福英基因科技有限公司 Kit and method for detecting horizontal in situ hybridization of MICRORNA-373 at the early pathological evolution stage of a variety of cancers and application thereof
EP2519648A1 (en) * 2009-12-30 2012-11-07 Febit Holding GmbH miRNA FINGERPRINT IN THE DIAGNOSIS OF PROSTATE CANCER
JP2012532149A (en) * 2009-07-09 2012-12-13 中国医学科学院▲腫▼瘤研究所 Use of two microRNAs in lung cancer prognosis and drug preparation
CN102864224A (en) * 2012-09-11 2013-01-09 南京大学 Serum microRNAs (ribonucleic acid) kit and application thereof
US20130053275A1 (en) * 2010-04-29 2013-02-28 Medical Prognosis Institute A/S Methods and devices for predicting treatment efficacy
WO2013039394A1 (en) * 2011-09-15 2013-03-21 Self-Screen B.V. Methylation analysis on self-samples as triage tool for hpv-positive women
US8895509B2 (en) 2010-11-23 2014-11-25 Georgia Tech Research Corporation MIR-200 family induces mesenchymal-to-epithelial transition (MET) in ovarian cancer cells
US9089589B2 (en) 2007-05-23 2015-07-28 University Of South Florida Micro-RNAs modulating immunity and inflammation
CN105486866A (en) * 2014-09-19 2016-04-13 杭州德同生物技术有限公司 Application of microRNA in preparation of kit for diagnosis of cervical carcinoma or precancerous lesions
US9334498B2 (en) 2012-05-10 2016-05-10 Uab Research Foundation Methods and compositions for modulating MIR-204 activity
JP6011945B2 (en) * 2011-05-20 2016-10-25 公一 中城 Composition comprising microRNA or expression system thereof
EP2963125A4 (en) * 2013-02-15 2016-11-02 Nat Univ Corp Tokyo Med & Dent Method for assaying microrna, cancer therapeutic agent, and medicinal composition containing same for cancer therapy
CN106661632A (en) * 2014-08-19 2017-05-10 生物辐射实验室股份有限公司 RNA amplification methods
CN106636307A (en) * 2015-11-02 2017-05-10 广东医学院 Application of miRNA-146a mutant in preparing test strip for early screening and rapid diagnosis of cervical cancer
US10000808B2 (en) 2009-01-16 2018-06-19 Cepheid Methods of detecting cervical cancer
CN108721317A (en) * 2018-05-15 2018-11-02 唐山市人民医院 Detect esophageal squamous cell carcinoma peripheral blood marker microRNA-602 and the application in drug and kit
WO2019022605A1 (en) * 2017-07-25 2019-01-31 Stichting Vumc Micro rna-based methods and compositions for detection of hpv-induced invasive cancers, and their high-grade precursor lesions
US20190062754A1 (en) * 2016-11-01 2019-02-28 The Research Foundation For The State University Of New York 5-halouracil-modified micrornas and their use in the treatment of cancer
US10570457B2 (en) 2014-09-26 2020-02-25 Medical Prognosis Institute A/S Methods for predicting drug responsiveness
US10907214B2 (en) 2016-12-30 2021-02-02 Oncology Venture ApS Methods for predicting drug responsiveness in cancer patients
KR20210071377A (en) * 2019-12-06 2021-06-16 아주대학교산학협력단 Biomarker composition for predicting prognosis of combined chemo radio therapy in a patient with cervical cancer
US11207269B2 (en) 2009-09-17 2021-12-28 Bio-Bedst Aps Medical use of sPLA2 hydrolysable liposomes
US11236337B2 (en) 2016-11-01 2022-02-01 The Research Foundation For The State University Of New York 5-halouracil-modified microRNAs and their use in the treatment of cancer
KR20220064042A (en) * 2020-11-11 2022-05-18 아주대학교산학협력단 Biomarkers for diagnosing early progression of cervical cancer, and uses thereof
US11421284B2 (en) 2016-10-07 2022-08-23 Allarity Therapeutics Europe ApS Methods for predicting drug responsiveness in cancer patients
WO2023128399A1 (en) * 2021-12-30 2023-07-06 주식회사 이노제닉스 Nucleic acid fragment specifically binding to 3' untranslated region of phosphatidylinositol 3-kinase catalytic subunit alpha gene, and applications thereof

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2471924A1 (en) 2004-05-28 2012-07-04 Asuragen, INC. Methods and compositions involving microRNA
CA2850323A1 (en) 2004-11-12 2006-12-28 Asuragen, Inc. Methods and compositions involving mirna and mirna inhibitor molecules
CN101424640B (en) * 2007-11-02 2012-07-25 江苏命码生物科技有限公司 Method for detecting miRNA in blood serum, detection kit, biochip, making method thereof and application method
US20100234445A1 (en) * 2008-07-01 2010-09-16 Weng Onn Lui Patterns of known and novel small RNAS in human cervical cancer
WO2012027462A2 (en) * 2010-08-24 2012-03-01 Steward St. Elizabeth's Medical Center Of Boston, Inc. Compositions and methods for treating neoplasia
JP5946466B2 (en) * 2010-12-01 2016-07-06 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル Method for predicting colorectal cancer outcome by analyzing miRNA expression
KR101232119B1 (en) * 2010-12-30 2013-02-12 연세대학교 산학협력단 Compositions for Preventing or Treating Neoplastic Disorders Comprising miRNA as an Active Ingredient
US20120225925A1 (en) * 2011-02-07 2012-09-06 Biomirna Holdings Ltd. Micro-RNA Biomarkers and Methods of Using Same
WO2012145743A1 (en) * 2011-04-22 2012-10-26 University Of Houston Microrna-140-5p as a tumor suppressor and sensitizing agent for chemotherapy
CN103931936B (en) * 2014-04-09 2015-08-19 长沙学院 A kind of fresh-water fishes parvalbumin anaphylactogen inhibitor
EP2940151A1 (en) * 2014-05-02 2015-11-04 Ruprecht-Karls-Universität Heidelberg Circulating miRNAs as early detection marker and prognostic marker
WO2017079571A1 (en) * 2015-11-05 2017-05-11 Arphion Diagnostics Process for the indentication of patients at risk for oscc
JP7218887B2 (en) * 2017-06-29 2023-02-07 学校法人藤田学園 Specimens for cervical cancer screening
CN109182512A (en) * 2018-11-27 2019-01-11 东北大学 A kind of biomarker and kit for screening congenital heart disease
KR102534200B1 (en) * 2020-11-11 2023-05-19 아주대학교산학협력단 Biomarkers for diagnosing metastasis of cervical cancer, and uses thereof
KR102534201B1 (en) * 2020-11-11 2023-05-19 아주대학교산학협력단 Biomarkers for diagnosing early progression of cervical cancer, and uses thereof
CN113265468B (en) * 2021-07-21 2021-10-12 北京大学第三医院(北京大学第三临床医学院) MiRNA related to cervical cancer diagnosis, treatment and prognosis
WO2023205586A1 (en) * 2022-04-21 2023-10-26 Inovio Pharmaceuticals, Inc. Methods of treatment of high-grade squamous intraepithelial lesion (hsil)
WO2024091990A1 (en) * 2022-10-25 2024-05-02 Inovio Pharmaceuticals, Inc. Methods of treatment of high-grade squamous intraepithelial lesion (hsil)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005118806A2 (en) * 2004-05-28 2005-12-15 Ambion, Inc. METHODS AND COMPOSITIONS INVOLVING MicroRNA
US20080076674A1 (en) * 2006-07-06 2008-03-27 Thomas Litman Novel oligonucleotide compositions and probe sequences useful for detection and analysis of non coding RNAs associated with cancer
WO2008095096A2 (en) * 2007-01-31 2008-08-07 Immune Disease Institute Let-7 microrna and mimetics thereof as therapeutics for cancer

Family Cites Families (284)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US203544A (en) * 1878-05-14 Improvement in treatment of flour
US3867155A (en) * 1973-10-31 1975-02-18 Cons Ceramic Products Smokeless exothermic hot topping compositions
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US5011769A (en) 1985-12-05 1991-04-30 Meiogenics U.S. Limited Partnership Methods for detecting nucleic acid sequences
US4876187A (en) 1985-12-05 1989-10-24 Meiogenics, Inc. Nucleic acid compositions with scissile linkage useful for detecting nucleic acid sequences
US5824311A (en) 1987-11-30 1998-10-20 Trustees Of The University Of Pennsylvania Treatment of tumors with monoclonal antibodies against oncogene antigens
US4999290A (en) 1988-03-31 1991-03-12 The Board Of Regents, The University Of Texas System Detection of genomic abnormalities with unique aberrant gene transcripts
US6040138A (en) 1995-09-15 2000-03-21 Affymetrix, Inc. Expression monitoring by hybridization to high density oligonucleotide arrays
GB8920097D0 (en) 1989-09-06 1989-10-18 Ici Plc Amplification processes
US5545522A (en) 1989-09-22 1996-08-13 Van Gelder; Russell N. Process for amplifying a target polynucleotide sequence using a single primer-promoter complex
US5366860A (en) 1989-09-29 1994-11-22 Applied Biosystems, Inc. Spectrally resolvable rhodamine dyes for nucleic acid sequence determination
DE69034150T2 (en) 1989-10-24 2005-08-25 Isis Pharmaceuticals, Inc., Carlsbad 2'-modified oligonucleotides
US5188934A (en) 1989-11-14 1993-02-23 Applied Biosystems, Inc. 4,7-dichlorofluorescein dyes as molecular probes
US5486603A (en) 1990-01-08 1996-01-23 Gilead Sciences, Inc. Oligonucleotide having enhanced binding affinity
US5859221A (en) 1990-01-11 1999-01-12 Isis Pharmaceuticals, Inc. 2'-modified oligonucleotides
US6153737A (en) 1990-01-11 2000-11-28 Isis Pharmaceuticals, Inc. Derivatized oligonucleotides having improved uptake and other properties
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
US5965364A (en) 1990-10-09 1999-10-12 Benner; Steven Albert Method for selecting functional deoxyribonucleotide derivatives
WO1992007095A1 (en) 1990-10-15 1992-04-30 Stratagene Arbitrarily primed polymerase chain reaction method for fingerprinting genomes
US6287792B1 (en) 1991-06-17 2001-09-11 The Regents Of The University Of California Drug delivery of antisense oligonucleotides and peptides to tissues in vivo and to cells using avidin-biotin technology
AU662906B2 (en) 1991-06-26 1995-09-21 F. Hoffmann-La Roche Ag Methods for detection of carcinoma metastases by nucleic acid amplification
US5256555A (en) 1991-12-20 1993-10-26 Ambion, Inc. Compositions and methods for increasing the yields of in vitro RNA transcription and other polynucleotide synthetic reactions
US5260191A (en) 1992-01-30 1993-11-09 Agracetus, Inc. Method for diagnosing tumors
US5262311A (en) 1992-03-11 1993-11-16 Dana-Farber Cancer Institute, Inc. Methods to clone polyA mRNA
GB9208545D0 (en) 1992-04-21 1992-06-03 Gatsby Charitable Foundation T Virus resistant plants
US5801005A (en) 1993-03-17 1998-09-01 University Of Washington Immune reactivity to HER-2/neu protein for diagnosis of malignancies in which the HER-2/neu oncogene is associated
US5767259A (en) 1994-12-27 1998-06-16 Naxcor Oligonucleotides containing base-free linking groups with photoactivatable side chains
US6045996A (en) 1993-10-26 2000-04-04 Affymetrix, Inc. Hybridization assays on oligonucleotide arrays
US5925517A (en) 1993-11-12 1999-07-20 The Public Health Research Institute Of The City Of New York, Inc. Detectably labeled dual conformation oligonucleotide probes, assays and kits
US5538848A (en) 1994-11-16 1996-07-23 Applied Biosystems Division, Perkin-Elmer Corp. Method for detecting nucleic acid amplification using self-quenching fluorescence probe
WO1995014106A2 (en) 1993-11-17 1995-05-26 Id Biomedical Corporation Cycling probe cleavage detection of nucleic acid sequences
GB9506466D0 (en) 1994-08-26 1995-05-17 Prolifix Ltd Cell cycle regulated repressor and dna element
US6096314A (en) 1994-10-07 2000-08-01 Yeda Research And Development Co. Ltd. Peptides and pharmaceutical compositions comprising them
EP1505159B1 (en) 1995-03-17 2008-05-28 John Wayne Cancer Institute Detection of breast metastases with a multiple marker assay
US5801155A (en) 1995-04-03 1998-09-01 Epoch Pharmaceuticals, Inc. Covalently linked oligonucleotide minor grove binder conjugates
IL122290A0 (en) 1995-06-07 1998-04-05 Inex Pharmaceuticals Corp Lipid-nucleic acid complex its preparation and use
US6111095A (en) 1995-06-07 2000-08-29 Merck & Co., Inc. Capped synthetic RNA, analogs, and aptamers
US5981501A (en) 1995-06-07 1999-11-09 Inex Pharmaceuticals Corp. Methods for encapsulating plasmids in lipid bilayers
US6998268B2 (en) 1995-07-03 2006-02-14 Dainippon Sumitomo Pharma Co. Ltd. Gene preparations
US5871697A (en) 1995-10-24 1999-02-16 Curagen Corporation Method and apparatus for identifying, classifying, or quantifying DNA sequences in a sample without sequencing
US6418382B2 (en) 1995-10-24 2002-07-09 Curagen Corporation Method and apparatus for identifying, classifying, or quantifying DNA sequences in a sample without sequencing
US5998203A (en) 1996-04-16 1999-12-07 Ribozyme Pharmaceuticals, Inc. Enzymatic nucleic acids containing 5'-and/or 3'-cap structures
EP0880598A4 (en) 1996-01-23 2005-02-23 Affymetrix Inc Nucleic acid analysis techniques
JP2002515738A (en) 1996-01-23 2002-05-28 アフィメトリックス,インコーポレイティド Nucleic acid analysis
US6020481A (en) 1996-04-01 2000-02-01 The Perkin-Elmer Corporation Asymmetric benzoxanthene dyes
DE69738687D1 (en) 1996-04-12 2008-06-26 Phri Properties Inc PROBES, KITS AND ASSAYS
US5847162A (en) 1996-06-27 1998-12-08 The Perkin Elmer Corporation 4, 7-Dichlororhodamine dyes
US5800996A (en) 1996-05-03 1998-09-01 The Perkin Elmer Corporation Energy transfer dyes with enchanced fluorescence
US5863727A (en) 1996-05-03 1999-01-26 The Perkin-Elmer Corporation Energy transfer dyes with enhanced fluorescence
US5945526A (en) 1996-05-03 1999-08-31 Perkin-Elmer Corporation Energy transfer dyes with enhanced fluorescence
US6184037B1 (en) 1996-05-17 2001-02-06 Genemedicine, Inc. Chitosan related compositions and methods for delivery of nucleic acids and oligonucleotides into a cell
SE506700C2 (en) 1996-05-31 1998-02-02 Mikael Kubista Probe and Methods for Analysis of Nucleic Acid
US5739169A (en) 1996-05-31 1998-04-14 Procept, Incorporated Aromatic compounds for inhibiting immune response
ATE295427T1 (en) 1996-06-04 2005-05-15 Univ Utah Res Found MONITORING HYBRIDIZATION DURING PCR
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
GB9618050D0 (en) 1996-08-29 1996-10-09 Cancer Res Campaign Tech Global amplification of nucleic acids
US6048326A (en) * 1996-12-31 2000-04-11 The Procter & Gamble Company Disposable elastic thermal knee wrap
DE69841038D1 (en) 1997-03-07 2009-09-17 Siemens Healthcare Diagnostics
WO1998040489A1 (en) 1997-03-10 1998-09-17 Japan Tobacco Inc. Antisense base sequences
US6090556A (en) 1997-04-07 2000-07-18 Japan Science & Technology Corporation Method for quantitatively determining the expression of a gene
NO972006D0 (en) 1997-04-30 1997-04-30 Forskningsparken I Aas As New method for diagnosis of diseases
EP1027033B1 (en) 1997-05-14 2009-07-22 The University Of British Columbia High efficiency encapsulation of nucleic acids in lipid vesicles
US6008379A (en) 1997-10-01 1999-12-28 The Perkin-Elmer Corporation Aromatic-substituted xanthene dyes
WO1999022018A2 (en) 1997-10-27 1999-05-06 Boston Probes, Inc. Methods, kits and compositions pertaining to pna molecular beacons
US6485901B1 (en) 1997-10-27 2002-11-26 Boston Probes, Inc. Methods, kits and compositions pertaining to linear beacons
ATE537270T1 (en) 1997-10-30 2011-12-15 Cold Spring Harbor Lab PROBE ARRANGEMENTS AND THEIR USES FOR DNA DISTINCTION PROCEDURES
US5936087A (en) 1997-11-25 1999-08-10 The Perkin-Elmer Corporation Dibenzorhodamine dyes
US6458533B1 (en) 1997-12-19 2002-10-01 High Throughput Genomics, Inc. High throughput assay system for monitoring ESTs
US6238869B1 (en) 1997-12-19 2001-05-29 High Throughput Genomics, Inc. High throughput assay system
US6232066B1 (en) 1997-12-19 2001-05-15 Neogen, Inc. High throughput assay system
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US6269846B1 (en) 1998-01-13 2001-08-07 Genetic Microsystems, Inc. Depositing fluid specimens on substrates, resulting ordered arrays, techniques for deposition of arrays
US5942398A (en) 1998-02-26 1999-08-24 Millennium Pharmaceuticals, Inc. Nucleic acid molecules encoding glutx and uses thereof
AUPP249298A0 (en) 1998-03-20 1998-04-23 Ag-Gene Australia Limited Synthetic genes and genetic constructs comprising same I
US6004755A (en) 1998-04-07 1999-12-21 Incyte Pharmaceuticals, Inc. Quantitative microarray hybridizaton assays
JP2003525017A (en) 1998-04-20 2003-08-26 リボザイム・ファーマシューティカルズ・インコーポレーテッド Nucleic acid molecules with novel chemical composition that can regulate gene expression
US6037129A (en) 1998-05-28 2000-03-14 Medical University Of South Carolina Multi-marker RT-PCR panel for detecting metastatic breast cancer
US6576420B1 (en) 1998-06-23 2003-06-10 Regents Of The University Of California Method for early diagnosis of, and determination of prognosis in, cancer
GB9815799D0 (en) 1998-07-21 1998-09-16 Pharmagene Lab Limited Quantitative analysis of gene expression
US6730477B1 (en) 1998-08-04 2004-05-04 Diadexus, Inc. Method of diagnosing, monitoring and staging breast cancer
US6140054A (en) 1998-09-30 2000-10-31 University Of Utah Research Foundation Multiplex genotyping using fluorescent hybridization probes
US6361947B1 (en) 1998-10-27 2002-03-26 Affymetrix, Inc. Complexity management and analysis of genomic DNA
DE19956568A1 (en) 1999-01-30 2000-08-17 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
GB9904991D0 (en) 1999-03-05 1999-04-28 Univ Nottingham Genetic screening
ATE465168T1 (en) 1999-03-18 2010-05-15 Exiqon As XYLO-LNA ANALOGUE
US6383752B1 (en) 1999-03-31 2002-05-07 Hybridon, Inc. Pseudo-cyclic oligonucleobases
AU776362B2 (en) 1999-05-04 2004-09-09 Roche Innovation Center Copenhagen A/S L-ribo-LNA analogues
US6132997A (en) 1999-05-28 2000-10-17 Agilent Technologies Method for linear mRNA amplification
EP1206234A4 (en) 1999-06-03 2005-06-01 Jessie L S Au Methods and compositions for modulating cell proliferation and cell death
WO2000075356A1 (en) 1999-06-04 2000-12-14 Lin Shi Lung Rna polymerase chain reaction
US6890553B1 (en) * 1999-07-08 2005-05-10 Johnson & Johnson Consumer Companies, Inc. Exothermic topical delivery device
US6964847B1 (en) 1999-07-14 2005-11-15 Packard Biosciences Company Derivative nucleic acids and uses thereof
US6201112B1 (en) 1999-07-22 2001-03-13 Agilent Technologies Inc. Method for 3′ end-labeling ribonucleic acids
US7005261B1 (en) 1999-07-29 2006-02-28 British Biocell International Limited Method for detecting nucleic acid target sequences involving in vitro transcription from an RNA polymerase promoter
US6140500A (en) 1999-09-03 2000-10-31 Pe Corporation Red-emitting [8,9]benzophenoxazine nucleic acid dyes and methods for their use
US6511832B1 (en) 1999-10-06 2003-01-28 Texas A&M University System In vitro synthesis of capped and polyadenylated mRNAs using baculovirus RNA polymerase
US20020094546A1 (en) 1999-10-07 2002-07-18 Shimkets Richard A. Growth factor polypeptides and nucleic acids encoding same
AU1086501A (en) 1999-10-15 2001-04-30 Carnegie Institution Of Washington Rna interference pathway genes as tools for targeted genetic interference
US6528254B1 (en) 1999-10-29 2003-03-04 Stratagene Methods for detection of a target nucleic acid sequence
US6191278B1 (en) 1999-11-03 2001-02-20 Pe Corporation Water-soluble rhodamine dyes and conjugates thereof
US6458382B1 (en) 1999-11-12 2002-10-01 Mirus Corporation Nucleic acid transfer complexes
US6435245B1 (en) 1999-11-18 2002-08-20 Pitney Bowes Inc. System for folding and tabbing sheets
GB9927444D0 (en) 1999-11-19 2000-01-19 Cancer Res Campaign Tech Inhibiting gene expression
DE10160151A1 (en) 2001-01-09 2003-06-26 Ribopharma Ag Inhibiting expression of target gene, useful e.g. for inhibiting oncogenes, by administering double-stranded RNA complementary to the target and having an overhang
DE10100586C1 (en) 2001-01-09 2002-04-11 Ribopharma Ag Inhibiting gene expression in cells, useful for e.g. treating tumors, by introducing double-stranded complementary oligoRNA having unpaired terminal bases
US7205105B2 (en) 1999-12-08 2007-04-17 Epoch Biosciences, Inc. Real-time linear detection probes: sensitive 5′-minor groove binder-containing probes for PCR analysis
AU2001234608A1 (en) 2000-01-28 2001-08-07 Genetrace Systems, Inc. Methods for analysis of gene expression
US20020065406A1 (en) 2000-03-24 2002-05-30 Meyers Rachel A. 18221, a novel dual specificity phosphatase and uses thereof
US6447723B1 (en) 2000-03-13 2002-09-10 Packard Instrument Company, Inc. Microarray spotting instruments incorporating sensors and methods of using sensors for improving performance of microarray spotting instruments
US20030084471A1 (en) 2000-03-16 2003-05-01 David Beach Methods and compositions for RNA interference
WO2001070095A2 (en) 2000-03-23 2001-09-27 Diadexus, Inc. Compositions and methods of diagnosing, monitoring, staging, imaging and treating prostate cancer
WO2001072780A2 (en) 2000-03-27 2001-10-04 Diadexus, Inc. Polynucleotides for diagnosing mammary gland cancer
AU2001251013A1 (en) 2000-03-28 2001-10-08 Diadexus, Inc. Compositions and methods of diagnosing, monitoring, staging, imaging and treating colon cancer
US20020119156A1 (en) 2000-03-29 2002-08-29 Sei-Yu Chen Compositions and methods of diagnosing, monitoring, staging, imaging and treating lung cancer
KR101215789B1 (en) 2000-03-30 2012-12-26 화이트헤드 인스티튜트 포 바이오메디칼 리서치 Rna sequence-specific mediators of rna interference
US20020068307A1 (en) 2000-03-30 2002-06-06 Jason Pluta Compositions and methods for diagnosing, monitoring, staging, imaging and treating stomach cancer
US6573048B1 (en) 2000-04-18 2003-06-03 Naxcor Degradable nucleic acid probes and nucleic acid detection methods
AU2001269690A1 (en) 2000-05-10 2001-11-20 David A. Sirbasku Compositions and methods for demonstrating secretory immune system regulation of steroid hormone responsive cancer cell growth
EP2279757A3 (en) 2000-06-02 2011-08-03 Bracco Suisse SA Compounds for targeting endothelial cells
CA2414396A1 (en) 2000-06-26 2002-01-03 The United States Of America, As Represented By The Secretary Of Agricul Ture Production of vaccines using transgenic plants
JP2004501666A (en) 2000-06-30 2004-01-22 エピゲノミクス アーゲー Methods and nucleic acids for methylation status analysis of pharmacogenomics
US6596490B2 (en) 2000-07-14 2003-07-22 Applied Gene Technologies, Inc. Nucleic acid hairpin probes and uses thereof
US6797471B2 (en) 2000-08-04 2004-09-28 Board Of Regents, The University Of Texas System Detection and diagnosis of smoking related cancers
AU2001292842A1 (en) 2000-09-19 2002-04-02 Diadexus, Inc. Compositions and methods relating to prostate specific genes and proteins
US20030082604A1 (en) 2000-09-27 2003-05-01 Swanson Melvin J. High density arrays
US7162371B1 (en) 2000-10-11 2007-01-09 Georgia Tech Research Corporation Differentially-expressed conifer cDNAs, and their use in improving somatic embryogenesis
US6350580B1 (en) 2000-10-11 2002-02-26 Stratagene Methods for detection of a target nucleic acid using a probe comprising secondary structure
US7001724B1 (en) 2000-11-28 2006-02-21 Applera Corporation Compositions, methods, and kits for isolating nucleic acids using surfactants and proteases
CZ302719B6 (en) 2000-12-01 2011-09-21 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Isolated double-stranded RNA molecule, process for its preparation and use
GB0029360D0 (en) 2000-12-01 2001-01-17 Univ Nottingham Humanised antibodies and uses thereof
DK1229130T3 (en) 2000-12-04 2014-02-24 Primagen B V TEST BASED ON nucleic acids OF ENDOSYMBIONTISKE, cellular organelles
US20030099976A1 (en) 2001-01-17 2003-05-29 Tai-Jay Chang Androgen receptor complex-associated protein
WO2002057496A2 (en) 2001-01-18 2002-07-25 Socratech L.L.C. Gene expression profiling of endothelium in alzheimer's disease
US7015047B2 (en) 2001-01-26 2006-03-21 Aviva Biosciences Corporation Microdevices having a preferential axis of magnetization and uses thereof
US20040058373A1 (en) 2001-01-31 2004-03-25 Winkler Matthew M. Competitive amplification of fractionated targets from multiple nucleic acid samples
WO2002064835A2 (en) 2001-01-31 2002-08-22 Ambion, Inc. Methods for nucleic acid fingerprint analysis
US20040110191A1 (en) 2001-01-31 2004-06-10 Winkler Matthew M. Comparative analysis of nucleic acids using population tagging
WO2002073504A1 (en) 2001-03-14 2002-09-19 Gene Logic, Inc. A system and method for retrieving and using gene expression data from multiple sources
US20030170623A1 (en) 2001-04-13 2003-09-11 Jingwen Chen Multiplexed gene analysis on a mobile solid support
WO2002086071A2 (en) 2001-04-20 2002-10-31 Ludwig Institute For Cancer Research Cancer-testis antigens
AU2002311869A1 (en) 2001-04-27 2002-11-11 Sunnybrook And Women's College Health Sciences Centre Breast cancer-associated genes and uses thereof
AUPR480901A0 (en) 2001-05-04 2001-05-31 Genomics Research Partners Pty Ltd Diagnostic method for assessing a condition of a performance animal
CA2448915A1 (en) 2001-06-01 2002-12-12 Prosanos Corporation Information processing method for disease stratification and assessment of disease progressing
US7171311B2 (en) 2001-06-18 2007-01-30 Rosetta Inpharmatics Llc Methods of assigning treatment to breast cancer patients
US20040086504A1 (en) 2001-06-21 2004-05-06 Deepak Sampath Cyr61 as a target for treatment and diagnosis of breast cancer
AU2002315413A1 (en) 2001-06-22 2003-01-08 Gene Logic, Inc. Platform for management and mining of genomic data
WO2003001985A2 (en) 2001-06-28 2003-01-09 Dermtech International Method for detection of melanoma
US7553939B2 (en) 2001-06-29 2009-06-30 The Board Of Trustees Of The Leland Stanford Junior University T cell regulatory genes and methods of use thereof
US20030143571A1 (en) 2001-08-08 2003-07-31 North Carolina State University Infectious disease microarray
AU2002331777A1 (en) 2001-08-30 2003-03-18 Spectral Genomics, Inc. Arrays comprising pre-labeled biological molecules and methods for making and using these arrays
US20030198627A1 (en) 2001-09-01 2003-10-23 Gert-Jan Arts siRNA knockout assay method and constructs
US7078180B2 (en) 2001-09-05 2006-07-18 The Children's Hospital Of Philadelphia Methods and compositions useful for diagnosis, staging, and treatment of cancers and tumors
AU2002333801B2 (en) 2001-09-06 2008-07-24 Merck Patent Gmbh Genetic analysis of biological samples in arrayed expanded representations of their nucleic acids
EP1425090A2 (en) 2001-09-07 2004-06-09 Corning Incorporated MICROCOLUMN&minus;PLATFORM BASED ARRAY FOR HIGH&minus;THROUGHPUT ANALYSIS
DE60234369D1 (en) 2001-09-19 2009-12-24 Alexion Pharma Inc MANIPULATED MATRICES AND THEIR USE IN SINGLE PRIMER AMPLIFICATION
WO2003024388A2 (en) 2001-09-20 2003-03-27 Cornell Research Foundation, Inc. Methods and compositions for treating and preventing skin disorders using binding agents specific for psma
US6593091B2 (en) 2001-09-24 2003-07-15 Beckman Coulter, Inc. Oligonucleotide probes for detecting nucleic acids through changes in flourescence resonance energy transfer
EP2428568B1 (en) 2001-09-28 2018-04-25 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Microrna molecules
EP1434883A2 (en) 2001-10-02 2004-07-07 Azign Bioscience A/S Specific differential display arrays
WO2003091426A1 (en) 2001-10-12 2003-11-06 Spectral Genomics, Inc. Compilations of nucleic acids and arrays and methods of using them
US20030087298A1 (en) 2001-11-02 2003-05-08 Roland Green Detection of hybridization on oligonucleotide microarray through covalently labeling microarray probe
US20040063654A1 (en) 2001-11-02 2004-04-01 Davis Mark E. Methods and compositions for therapeutic use of RNA interference
IL161733A0 (en) 2001-11-02 2005-11-20 Insert Therapeutics Inc Methods and compositions for therapeutic use of rna interference
CA2467456A1 (en) 2001-11-19 2003-05-30 Protometrix, Inc. Method of using a non-antibody protein to detect and measure an analyte
JP2005513457A (en) 2001-12-19 2005-05-12 アフィメトリックス インコーポレイテッド Array plate and method for constructing array plate
WO2003057148A2 (en) 2001-12-27 2003-07-17 Agy Therapeutics, Inc. Use of biomolecular targets in the treatment and visualization of tumors
US7141372B2 (en) 2002-01-18 2006-11-28 Health Research Incorporated Universal RT-coupled PCR method for the specific amplification of mRNA
US20030215842A1 (en) 2002-01-30 2003-11-20 Epigenomics Ag Method for the analysis of cytosine methylation patterns
CA2474530A1 (en) 2002-02-07 2003-08-14 Eastern Virginia Medical School Of The Medical College Of Hampton Roads Diagnostic microarray and method of use thereof
US20030232356A1 (en) 2002-02-08 2003-12-18 Dooley Thomas P. Skin cell biomarkers and methods for identifying biomarkers using nucleic acid microarrays
EP1488228A4 (en) 2002-03-07 2008-09-17 Univ Utah Res Found Methods for identifying large subsets of differentially expressed genes based on multivariate microarray data analysis
US20070025997A1 (en) 2002-04-03 2007-02-01 Usha Nagavarapu Use of biomolecular targets in the treatment and visualization of brain tumors
WO2003085125A1 (en) 2002-04-03 2003-10-16 Agy Therapeutics, Inc. Use of biomolecular targets in the treatment and visualization of brain tumors
US7006680B2 (en) 2002-05-03 2006-02-28 Vialogy Corp. System and method for characterizing microarray output data
EP1504126B1 (en) 2002-05-03 2014-02-26 Duke University A method of regulating gene expression
WO2003100382A1 (en) 2002-05-20 2003-12-04 Northrop Grumman Corporation Automatic point source biological agent detection system
WO2003100012A2 (en) 2002-05-24 2003-12-04 Nimblegen Systems, Inc. Microarrays and method for running hybridization reaction for multiple samples on a single microarray
AUPS261402A0 (en) 2002-05-28 2002-06-20 Compusign Pty Ltd Array monitoring
US20030228579A1 (en) 2002-06-05 2003-12-11 Uri Alon Ordering genes by analysis of expression kinetics
EP1527176B2 (en) 2002-08-05 2017-03-22 Silence Therapeutics GmbH Further novel forms of interfering rna molecules
US8729036B2 (en) 2002-08-07 2014-05-20 University Of Massachusetts Compositions for RNA interference and methods of use thereof
US20040029121A1 (en) 2002-08-08 2004-02-12 Susan Cottrell Methods and nucleic acids for the analysis of CpG dinucleotide methylation status associated with the calcitonin gene
US20040029128A1 (en) 2002-08-08 2004-02-12 Epigenomics, Inc. Methods and nucleic acids for the analysis of CpG dinucleotide methylation status associated with the calcitonin gene
EP1560924A4 (en) 2002-08-16 2006-05-31 Wayne John Cancer Inst Molecular lymphatic mapping of sentinel lymph nodes
AU2003270654A1 (en) 2002-09-12 2004-04-30 Baylor College Of Medecine System and method for image segmentation
EP1556506A1 (en) 2002-09-19 2005-07-27 The Chancellor, Masters And Scholars Of The University Of Oxford Molecular arrays and single molecule detection
CA2500224C (en) 2002-09-25 2015-04-28 University Of Massachusetts In vivo gene silencing by chemically modified and stable sirna
AU2003291287A1 (en) 2002-11-01 2004-06-07 John Fonda Crary Apkc isoforms in nervous system disorders and cancer
EP1418241A1 (en) 2002-11-08 2004-05-12 PrimaGen Holding B.V. Method for quantifying a ratio between at least two nucleic acid sequences
AU2003291433B2 (en) 2002-11-13 2008-05-22 Thomas Jefferson University Compositions and methods for cancer diagnosis and therapy
US7655785B1 (en) 2002-11-14 2010-02-02 Rosetta Genomics Ltd. Bioinformatically detectable group of novel regulatory oligonucleotides and uses thereof
WO2004046324A2 (en) 2002-11-15 2004-06-03 University Of Massachusetts Allele-targeted rna interference
US20040175732A1 (en) 2002-11-15 2004-09-09 Rana Tariq M. Identification of micrornas and their targets
JPWO2004050125A1 (en) 2002-12-03 2006-03-30 株式会社高研 Inhibitor of tumor cell growth and / or invasion
JP4230457B2 (en) 2002-12-18 2009-02-25 サード・ウェーブ・テクノロジーズ・インク Detection method of low molecular nucleic acid
US7851150B2 (en) 2002-12-18 2010-12-14 Third Wave Technologies, Inc. Detection of small nucleic acids
CN101061236A (en) 2003-01-13 2007-10-24 艾默瑞大学 Methods of detecting gene expression in normal and cancerous cells
ATE389728T1 (en) 2003-01-16 2008-04-15 Univ North Carolina State DEPUPPERATION OF ENDOGENE PRIMORDIAL GERM CELLS IN BIRD SPECIES
WO2004066183A2 (en) 2003-01-22 2004-08-05 European Molecular Biology Laboratory Microrna
US20040147027A1 (en) 2003-01-28 2004-07-29 Troy Carol M. Complex for facilitating delivery of dsRNA into a cell and uses thereof
EP1592791A2 (en) 2003-02-10 2005-11-09 National Institute of Advanced Industrial Science and Technology Regulation of gene expression by dna interference
US7816337B2 (en) 2003-02-18 2010-10-19 Roche Madison Inc. Reversible attachment of a membrane active polymer to a polynucleotide
US20040224337A1 (en) 2003-03-04 2004-11-11 Erik Foehr Use of biomolecular targets in the treatment and visualization of tumors
US20050112604A1 (en) 2003-03-14 2005-05-26 Akihide Fujimoto Loss of heterozygosity of the DNA markers in the 12q22-23 region
WO2004086949A2 (en) 2003-03-25 2004-10-14 John Wayne Cancer Institute Dna markers for management of cancer
CA2488224A1 (en) 2003-04-03 2004-10-21 Alnylam Pharmaceuticals, Inc. Irna conjugates
US20040229211A1 (en) 2003-05-13 2004-11-18 Yeung Wah Hin Alex Sensitive diagnostic testing methodology using multiplex real time PCR with one dye (MOD) and its use as in severe acute respiratory syndrome (SARS)
US7640114B2 (en) 2003-05-21 2009-12-29 The Wistar Institute Of Anatomy & Biology Method of diagnosis of cancer based on gene expression profiles in cells
ES2864206T3 (en) 2003-06-02 2021-10-13 Univ Massachusetts Methods and compositions to improve the efficacy and specificity of RNAi
US7750144B2 (en) 2003-06-02 2010-07-06 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNA silencing
PL1633767T3 (en) 2003-06-02 2019-07-31 University Of Massachusetts Methods and compositions for controlling efficacy of rna silencing
US20050059024A1 (en) 2003-07-25 2005-03-17 Ambion, Inc. Methods and compositions for isolating small RNA molecules
EP1648914A4 (en) 2003-07-31 2009-12-16 Regulus Therapeutics Inc Oligomeric compounds and compositions for use in modulation of small non-coding rnas
US8106180B2 (en) 2003-08-07 2012-01-31 Whitehead Institute For Biomedical Research Methods and products for expression of micro RNAs
US20050037362A1 (en) 2003-08-11 2005-02-17 Eppendorf Array Technologies, S.A. Detection and quantification of siRNA on microarrays
US20060183128A1 (en) 2003-08-12 2006-08-17 Epigenomics Ag Methods and compositions for differentiating tissues for cell types using epigenetic markers
US6920873B2 (en) * 2003-09-11 2005-07-26 Energy Conversion Devices, Inc. Portable heating pack
US20070031840A1 (en) 2003-11-10 2007-02-08 Noxxon Pharma Ag Nucleic acids specifically binding bioactive ghrelin
US20050130170A1 (en) 2003-12-16 2005-06-16 Jeanne Harvey Identification and verification of methylation marker sequences
US20050130172A1 (en) 2003-12-16 2005-06-16 Bayer Corporation Identification and verification of methylation marker sequences
WO2005078139A2 (en) 2004-02-09 2005-08-25 Thomas Jefferson University DIAGNOSIS AND TREATMENT OF CANCERS WITH MicroRNA LOCATED IN OR NEAR CANCER-ASSOCIATED CHROMOSOMAL FEATURES
US20050182005A1 (en) 2004-02-13 2005-08-18 Tuschl Thomas H. Anti-microRNA oligonucleotide molecules
AU2005214904B2 (en) 2004-02-13 2011-07-21 Rockefeller University Anti-microRNA oligonucleotide molecules
KR100522307B1 (en) 2004-02-19 2005-10-19 변영철 Cosmetics brush
US7402389B2 (en) 2004-02-24 2008-07-22 The Translational Genomics Research Institute (Tgen) Compositions and methods for prognosis of cancers
US20060195266A1 (en) 2005-02-25 2006-08-31 Yeatman Timothy J Methods for predicting cancer outcome and gene signatures for use therein
WO2005102456A1 (en) 2004-03-27 2005-11-03 The Arizona Board Of Regents On Behalf Of The University Of Arizona Composition and method for cancer treatment
US20060134639A1 (en) 2004-04-06 2006-06-22 Huffel Christophe V Method for the determination of cellular transcriptional regulation
US7365058B2 (en) 2004-04-13 2008-04-29 The Rockefeller University MicroRNA and methods for inhibiting same
EP2338996A3 (en) 2004-05-04 2013-12-18 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for reducing viral genome amounts in a target cell
CA2566519C (en) 2004-05-14 2020-04-21 Rosetta Genomics Ltd. Micrornas and uses thereof
WO2005116261A2 (en) 2004-05-24 2005-12-08 Albert Einstein College Of Medecine Of Yeshiva University Rna expression microarrays
US7575863B2 (en) 2004-05-28 2009-08-18 Applied Biosystems, Llc Methods, compositions, and kits comprising linker probes for quantifying polynucleotides
US20050287539A1 (en) 2004-06-29 2005-12-29 Emmanuel Labourier Methods and compositions for preparing capped RNA
JP5192234B2 (en) 2004-08-10 2013-05-08 アルナイラム ファーマシューティカルズ, インコーポレイテッド Chemically modified oligonucleotide
EP2298897B1 (en) 2004-09-02 2013-08-14 Yale University Regulation of oncogenes by microRNAs
US20060057595A1 (en) 2004-09-16 2006-03-16 Applera Corporation Compositions, methods, and kits for identifying and quantitating small RNA molecules
US7592441B2 (en) 2004-10-04 2009-09-22 Rosetta Genomics Ltd Liver cancer-related nucleic acids
US7825229B2 (en) 2005-03-25 2010-11-02 Rosetta Genomics Ltd. Lung cancer-related nucleic acids
US20090186353A1 (en) 2004-10-04 2009-07-23 Rosetta Genomics Ltd. Cancer-related nucleic acids
US7642348B2 (en) 2004-10-04 2010-01-05 Rosetta Genomics Ltd Prostate cancer-related nucleic acids
US20060078894A1 (en) 2004-10-12 2006-04-13 Winkler Matthew M Methods and compositions for analyzing nucleic acids
WO2006042328A2 (en) 2004-10-12 2006-04-20 John Wayne Cancer Institute Treatment of cancer and compositions
US7575380B2 (en) * 2004-10-15 2009-08-18 Emcore Corporation Integrated optical fiber and electro-optical converter
FR2877350B1 (en) 2004-11-03 2010-08-27 Centre Nat Rech Scient IDENTIFICATION AND USE OF miRNAs INVOLVED IN THE DIFFERENTIATION OF CELLS FROM MYELOID LEUKEMIA
CA2850323A1 (en) 2004-11-12 2006-12-28 Asuragen, Inc. Methods and compositions involving mirna and mirna inhibitor molecules
US7074622B2 (en) 2004-11-15 2006-07-11 Eastman Kodak Company Method and system for sorting and separating particles
US20060252057A1 (en) 2004-11-30 2006-11-09 Mitch Raponi Lung cancer prognostics
US20060154275A1 (en) 2004-12-02 2006-07-13 The Board Of Trustees Of The Leland Stanford Junior University Regulated genes in cervical cancer
US20060185027A1 (en) 2004-12-23 2006-08-17 David Bartel Systems and methods for identifying miRNA targets and for altering miRNA and target expression
WO2006069584A2 (en) 2004-12-29 2006-07-06 Exiqon A/S NOVEL OLIGONUCLEOTIDE COMPOSITIONS AND PROBE SEQUENCES USEFUL FOR DETECTION AND ANALYSIS OF microRNAs AND THEIR TARGET mRNAs
WO2006113679A2 (en) 2005-04-15 2006-10-26 Board Of Regents, The University Of Texas System Delivery of sirna by neutral lipid compositions
AU2006211960A1 (en) 2005-02-08 2006-08-17 Board Of Regents, The University Of Texas System Compositions and methods involving MDA-7 for the treatment of cancer
US7495073B2 (en) 2005-03-24 2009-02-24 Asia Hepato Gene Company Short isoform of Annexin A10 at chromosome 4q, termed Annexin 10s (ANXA10s) and methods of use
US20090062184A1 (en) 2005-03-24 2009-03-05 Dainippon Sumitomo Pharma Co., Ltd. Fine particulate preparation comprising complex of nucleic acid molecule and collagen
WO2006119365A2 (en) 2005-05-02 2006-11-09 Cold Spring Harbor Laboratory Composition and methods for cancer diagnosis utilizing the mir 17-92 cluster
GB0601102D0 (en) 2006-01-19 2006-03-01 Nuclea Biomarkers Llc Kinase Peptides And Antibodies
US20070054287A1 (en) 2005-05-31 2007-03-08 Applera Corporation Method for identifying medically important cell populations using micro rna as tissue specific biomarkers
WO2006128245A1 (en) 2005-06-03 2006-12-07 Southern Adelaide Health Service-Flinders Medical Centre Targeting cells with altered microrna expression
US20070065844A1 (en) 2005-06-08 2007-03-22 Massachusetts Institute Of Technology Solution-based methods for RNA expression profiling
US20070048758A1 (en) 2005-06-09 2007-03-01 Epoch Biosciences, Inc. Improved primer-based amplification methods
US20060292616A1 (en) 2005-06-23 2006-12-28 U.S. Genomics, Inc. Single molecule miRNA-based disease diagnostic methods
CN105902559A (en) 2005-08-01 2016-08-31 俄亥俄州立大学研究基金会 Diagnosis and prognosis for breast cancer, micro RNA-based method and micro RNA-based composition for treating breast cancer
IL177006A0 (en) 2005-08-02 2006-12-10 Veridex Llc Predicting bone relapse of breast cancer
US20070213292A1 (en) 2005-08-10 2007-09-13 The Rockefeller University Chemically modified oligonucleotides for use in modulating micro RNA and uses thereof
US20070041934A1 (en) 2005-08-12 2007-02-22 Regents Of The University Of Michigan Dendrimer based compositions and methods of using the same
CN101296702B (en) 2005-09-12 2012-11-28 俄亥俄州立大学研究基金会 Compositions and methods for the diagnosis and therapy of BCL2-associated cancers
US7390792B2 (en) 2005-12-15 2008-06-24 Board Of Regents, The University Of Texas System MicroRNA1 therapies
EP1976567B1 (en) 2005-12-28 2020-05-13 The Scripps Research Institute Natural antisense and non-coding rna transcripts as drug targets
EP1966390A1 (en) 2005-12-29 2008-09-10 Exiqon A/S Detection of tissue origin of cancer
CN103993082B (en) 2006-01-05 2017-01-11 俄亥俄州立大学研究基金会 Microrna-based methods and compositions for the diagnosis, prognosis and treatment of lung cancer
EP2586455B1 (en) 2006-01-05 2014-06-25 The Ohio State University Research Foundation MicroRNA expressions abnormalities in pancreatic endocrine and acinar tumors
EP2468890B1 (en) 2006-01-05 2014-03-19 The Ohio State University Research Foundation MicroRNA-based methods and compositions for the diagnosis and treatment of solid cancers of the breast or lung
US20070259827A1 (en) 2006-01-25 2007-11-08 University Of Massachusetts Compositions and methods for enhancing discriminatory RNA interference
EP2522750A1 (en) 2006-03-02 2012-11-14 The Ohio State University MicroRNA expression profile associated with pancreatic cancer
US7955848B2 (en) 2006-04-03 2011-06-07 Trustees Of Dartmouth College MicroRNA biomarkers for human breast and lung cancer
JP5131927B2 (en) 2006-06-16 2013-01-30 大正製薬株式会社 Use of RPN2 gene expression inhibitor
EP2455492B1 (en) 2006-07-13 2013-11-20 The Ohio State University Research Foundation Micro-RNA-based methods and compositions for the diagnosis and treatment of colon related diseases
WO2008014008A2 (en) 2006-07-28 2008-01-31 The Johns Hopkins University Compositions and methods for modulating angiogenesis
US20080131878A1 (en) 2006-12-05 2008-06-05 Asuragen, Inc. Compositions and Methods for the Detection of Small RNA
CN101622348A (en) 2006-12-08 2010-01-06 奥斯瑞根公司 Gene and the approach regulated as the miR-20 of targets for therapeutic intervention
EP2610347B1 (en) 2007-04-30 2015-04-15 The Ohio State University Research Foundation Methods of determining the prognosis of a subject with pancreatic cancer
AU2008247427A1 (en) 2007-05-03 2008-11-13 Rosetta Inpharmatics Llc Compositions comprising miR34 therapeutic agents for treating cancer

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005118806A2 (en) * 2004-05-28 2005-12-15 Ambion, Inc. METHODS AND COMPOSITIONS INVOLVING MicroRNA
US20080076674A1 (en) * 2006-07-06 2008-03-27 Thomas Litman Novel oligonucleotide compositions and probe sequences useful for detection and analysis of non coding RNAs associated with cancer
WO2008095096A2 (en) * 2007-01-31 2008-08-07 Immune Disease Institute Let-7 microrna and mimetics thereof as therapeutics for cancer

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ESQUELA-KERSCHER AURORA ET AL: "Oncomirs - microRNAs with a role in cancer", NATURE REVIEWS CANCER, vol. 6, no. 4, April 2006 (2006-04-01), pages 259 - 269, XP002506706, ISSN: 1474-175X *
MURALIDHAR B ET AL: "Global microRNA profiles in cervical squamous cell carcinoma depend on Drosha expression levels", JOURNAL OF PATHOLOGY, CHICHESTER, SUSSEX, GB, vol. 212, no. 4, 1 August 2007 (2007-08-01), pages 368 - 377, XP002491292, ISSN: 0022-3417 *

Cited By (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9089589B2 (en) 2007-05-23 2015-07-28 University Of South Florida Micro-RNAs modulating immunity and inflammation
EP2641976A3 (en) * 2009-01-16 2014-01-08 Cepheid Methods of detecting cervical cancer
WO2010083464A3 (en) * 2009-01-16 2011-05-12 Cepheid Methods of detecting cervical cancer
US10000808B2 (en) 2009-01-16 2018-06-19 Cepheid Methods of detecting cervical cancer
WO2010139810A1 (en) * 2009-06-05 2010-12-09 Febit Holding Gmbh Mirna fingerprint in the diagnosis of lung cancer
US9758827B2 (en) 2009-06-05 2017-09-12 Comprehensive Biomarker Center Gmbh miRNA fingerprint in the diagnosis of lung cancer
US9702008B2 (en) 2009-06-05 2017-07-11 Hummingbird Diagnostics Gmbh miRNA fingerprint in the diagnosis of diseases
US9187785B2 (en) 2009-06-05 2015-11-17 Comprehensive Biomarker Center Gmbh miRNA fingerprint in the diagnosis of multiple sclerosis
US9194002B2 (en) 2009-06-05 2015-11-24 Comprehensive Biomarker Center Gmbh miRNA fingerprint in the diagnosis of diseases
JP2012532149A (en) * 2009-07-09 2012-12-13 中国医学科学院▲腫▼瘤研究所 Use of two microRNAs in lung cancer prognosis and drug preparation
WO2011015040A1 (en) * 2009-08-07 2011-02-10 博奥生物有限公司 Methods and compositions for the diagnosis and prognosis of cervical intraepithelial neoplasia and cervical cancer
US8957039B2 (en) 2009-08-07 2015-02-17 Capitalbio Corporation Methods and compositions for the diagnosis and prognosis of cervical intraepithelial neoplasia and cervical cancer
CN102510905A (en) * 2009-08-07 2012-06-20 博奥生物有限公司 Methods and compositions for the diagnosis and prognosis of cervical intraepithelial neoplasia and cervical cancer
WO2011014980A1 (en) * 2009-08-07 2011-02-10 Capitalbio Corporation Methods and compositions diagnosing cervical cancer and cervical dysplasia, guidding subsequent treatment, determining prognosis, and improving patient survival
US20120202872A1 (en) * 2009-08-07 2012-08-09 Wenyan Qin Methods and compositions for the diagnosis and prognosis of cervical intraepithelial neoplasia and cervical cancer
WO2011018506A1 (en) * 2009-08-12 2011-02-17 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. miRNA COMPOUNDS FOR TREATMENT OF PROSTATE CARCINOMA
EP2283846A1 (en) * 2009-08-12 2011-02-16 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. miRNA compounds for treatment of prostate carcinoma
US8980854B2 (en) 2009-08-12 2015-03-17 Fraunhofer-Gesellschaft Zur Forderung Der Angewandten Forschung E.V. miRNA compounds for treatment of prostate carcinoma
KR101873440B1 (en) 2009-09-02 2018-07-02 로레알 Epidermal differentiation microrna signature and uses thereof
CN102625853A (en) * 2009-09-02 2012-08-01 欧莱雅 Epidermal differentiation microRNA signature and uses thereof
US9090937B2 (en) 2009-09-02 2015-07-28 L'oreal Epidermal differentiation microrna signature and uses thereof
WO2011026909A1 (en) * 2009-09-02 2011-03-10 L'oreal Epidermal differentiation microrna signature and uses thereof
US11207269B2 (en) 2009-09-17 2021-12-28 Bio-Bedst Aps Medical use of sPLA2 hydrolysable liposomes
EP2772551A1 (en) * 2009-12-30 2014-09-03 Comprehensive Biomarker Center GmbH miRNA fingerprint from patient's blood samples in the diagnosis of Wilms' tumour
EP2519648A1 (en) * 2009-12-30 2012-11-07 Febit Holding GmbH miRNA FINGERPRINT IN THE DIAGNOSIS OF PROSTATE CANCER
EP2519648B1 (en) * 2009-12-30 2016-09-28 Hummingbird Diagnostics GmbH miRNA fingerprint in the diagnosis of prostate cancer
US9598734B2 (en) * 2010-04-29 2017-03-21 Medical Prognosis Institute A/S Methods and devices for predicting treatment efficacy
US20130053275A1 (en) * 2010-04-29 2013-02-28 Medical Prognosis Institute A/S Methods and devices for predicting treatment efficacy
US9297007B2 (en) 2010-05-28 2016-03-29 Vereniging Voor Christelijk Hoger Onderwijs, Wetenschappelijk Onderzoek En Patientenzorg miRNAs involved in the blood brain barrier function
WO2011149354A1 (en) * 2010-05-28 2011-12-01 Vereniging Voor Christelijk Hoger Onderwijs, Wetenschappelijk Onderzoek En Patiëntenzorg Mirnas involved in the blood brain barrier function
CN101851682A (en) * 2010-06-25 2010-10-06 中国人民解放军南京军区南京总医院 MiRNA combination used for detecting esophageal squamous cell carcinoma and application thereof
WO2012025709A1 (en) * 2010-08-23 2012-03-01 Sistemic Uk Cell characterisation
CN103403180A (en) * 2010-08-23 2013-11-20 西斯特米克苏格兰有限公司 Cell characterisation
CN101921759A (en) * 2010-09-08 2010-12-22 南京医科大学 Serum/plasma miRNA serum marker related to cervical carcinoma and precancerous lesions thereof and application thereof
US8895509B2 (en) 2010-11-23 2014-11-25 Georgia Tech Research Corporation MIR-200 family induces mesenchymal-to-epithelial transition (MET) in ovarian cancer cells
JP6011945B2 (en) * 2011-05-20 2016-10-25 公一 中城 Composition comprising microRNA or expression system thereof
WO2013039394A1 (en) * 2011-09-15 2013-03-21 Self-Screen B.V. Methylation analysis on self-samples as triage tool for hpv-positive women
CN102533977A (en) * 2011-12-15 2012-07-04 苏州福英基因科技有限公司 Kit and method for detecting horizontal in situ hybridization of MICRORNA-372 at the early pathological evolution stage of a variety of cancers and application thereof
CN102533978A (en) * 2011-12-15 2012-07-04 苏州福英基因科技有限公司 Kit and method for detecting horizontal in situ hybridization of MICRORNA-373 at the early pathological evolution stage of a variety of cancers and application thereof
CN102533987A (en) * 2011-12-20 2012-07-04 苏州福英基因科技有限公司 Horizontal in-situ hybridization detection kit and detection method as well as application for MICRORNA-143 in earlier stage of cancer pathologic evolution
CN102443650A (en) * 2011-12-20 2012-05-09 苏州福英基因科技有限公司 In-situ hybridization detection kit for MICRORNA (MICRO Ribonucleic Acid)-233 level at pathologic evolution early stage of colon cancer as well as microRNA-233 in-situ hybridization detection method and application of microRNA-233 to preparation of in-situ hybridization detection kit for colon cancer
US9334498B2 (en) 2012-05-10 2016-05-10 Uab Research Foundation Methods and compositions for modulating MIR-204 activity
CN102864224A (en) * 2012-09-11 2013-01-09 南京大学 Serum microRNAs (ribonucleic acid) kit and application thereof
US10876115B2 (en) 2013-02-15 2020-12-29 National University Corporation Tokyo Medical And Dental University Method for assaying MicroRNA, cancer therapeutic agent, and medical composition containing same for cancer therapy
EP2963125A4 (en) * 2013-02-15 2016-11-02 Nat Univ Corp Tokyo Med & Dent Method for assaying microrna, cancer therapeutic agent, and medicinal composition containing same for cancer therapy
US9994843B2 (en) 2013-02-15 2018-06-12 National University Corporation Tokyo Medical And Dental University Method for assaying microRNA, cancer therapeutic agent, and medicinal composition containing same for cancer therapy
CN106661632A (en) * 2014-08-19 2017-05-10 生物辐射实验室股份有限公司 RNA amplification methods
CN105486866A (en) * 2014-09-19 2016-04-13 杭州德同生物技术有限公司 Application of microRNA in preparation of kit for diagnosis of cervical carcinoma or precancerous lesions
US10570457B2 (en) 2014-09-26 2020-02-25 Medical Prognosis Institute A/S Methods for predicting drug responsiveness
CN106636307A (en) * 2015-11-02 2017-05-10 广东医学院 Application of miRNA-146a mutant in preparing test strip for early screening and rapid diagnosis of cervical cancer
US11421284B2 (en) 2016-10-07 2022-08-23 Allarity Therapeutics Europe ApS Methods for predicting drug responsiveness in cancer patients
US11584932B2 (en) * 2016-11-01 2023-02-21 The Research Foundation For The State University Of New York 5-halouracil-modified microRNAs and their use in the treatment of cancer
US20190062754A1 (en) * 2016-11-01 2019-02-28 The Research Foundation For The State University Of New York 5-halouracil-modified micrornas and their use in the treatment of cancer
US11236337B2 (en) 2016-11-01 2022-02-01 The Research Foundation For The State University Of New York 5-halouracil-modified microRNAs and their use in the treatment of cancer
US10907214B2 (en) 2016-12-30 2021-02-02 Oncology Venture ApS Methods for predicting drug responsiveness in cancer patients
WO2019022605A1 (en) * 2017-07-25 2019-01-31 Stichting Vumc Micro rna-based methods and compositions for detection of hpv-induced invasive cancers, and their high-grade precursor lesions
CN108721317A (en) * 2018-05-15 2018-11-02 唐山市人民医院 Detect esophageal squamous cell carcinoma peripheral blood marker microRNA-602 and the application in drug and kit
KR102280516B1 (en) * 2019-12-06 2021-07-22 아주대학교산학협력단 Biomarker composition for predicting prognosis of combined chemo radio therapy in a patient with cervical cancer
KR20210071377A (en) * 2019-12-06 2021-06-16 아주대학교산학협력단 Biomarker composition for predicting prognosis of combined chemo radio therapy in a patient with cervical cancer
KR20220064042A (en) * 2020-11-11 2022-05-18 아주대학교산학협력단 Biomarkers for diagnosing early progression of cervical cancer, and uses thereof
KR20230056644A (en) * 2020-11-11 2023-04-27 아주대학교산학협력단 Composition for diagnosing early progression of cervical cancer
KR20230057325A (en) * 2020-11-11 2023-04-28 아주대학교산학협력단 Method of providing information for diagnosing early progression of cervical cancer
KR20230060492A (en) * 2020-11-11 2023-05-04 아주대학교산학협력단 Kit for diagnosing early progression of cervical cancer
KR102534199B1 (en) * 2020-11-11 2023-05-19 아주대학교산학협력단 Biomarkers for diagnosing early progression of cervical cancer, and uses thereof
KR102555308B1 (en) 2020-11-11 2023-07-14 아주대학교산학협력단 Composition for diagnosing early progression of cervical cancer
KR102555309B1 (en) 2020-11-11 2023-07-14 아주대학교산학협력단 Method of providing information for diagnosing early progression of cervical cancer
KR102601997B1 (en) 2020-11-11 2023-11-14 아주대학교산학협력단 Kit for diagnosing early progression of cervical cancer
WO2023128399A1 (en) * 2021-12-30 2023-07-06 주식회사 이노제닉스 Nucleic acid fragment specifically binding to 3' untranslated region of phosphatidylinositol 3-kinase catalytic subunit alpha gene, and applications thereof

Also Published As

Publication number Publication date
US20130184175A1 (en) 2013-07-18
EP2198050A1 (en) 2010-06-23
US20090186348A1 (en) 2009-07-23
US8361714B2 (en) 2013-01-29
US9080215B2 (en) 2015-07-14

Similar Documents

Publication Publication Date Title
US9080215B2 (en) MicroRNAs differentially expressed in cervical cancer and uses thereof
EP2260110B1 (en) miRNAs DIFFERENTIALLY EXPRESSED IN LYMPH NODES FROM CANCER PATIENTS
US20220213551A1 (en) Mirnas as biomakers for distinguishing benign from malignant thyroid neoplasms
US20190330623A1 (en) Micrornas differentially expressed in pancreatic diseases and uses thereof
US20190017122A1 (en) Mirnas as diagnostic biomarkers to distinguish benign from malignant thyroid tumors
US20130029874A1 (en) Microrna markers for recurrence of colorectal cancer
US20090092974A1 (en) Micrornas differentially expressed in leukemia and uses thereof
US20180327856A1 (en) Diagnostic mirnas for differential diagnosis of incidental pancreatic cystic lesions
Class et al. Patent application title: miRNAs Differentially Expressed in Lymph Nodes from Cancer Patients Inventors: Sylvie Beaudenon (Austin, TX, US) Laura Elizondo (Austin, TX, US) Martina Doleshal (Austin, TX, US) David Brown (Ausitn, TX, US) Emmanuel Labourier (Austin, TX, US) Assignees: Asuragen, Inc.

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: 08831073

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2008831073

Country of ref document: EP