EP2387616A2 - Verfahren zum nachweis von gebärmutterhalskrebs - Google Patents

Verfahren zum nachweis von gebärmutterhalskrebs

Info

Publication number
EP2387616A2
EP2387616A2 EP10703542A EP10703542A EP2387616A2 EP 2387616 A2 EP2387616 A2 EP 2387616A2 EP 10703542 A EP10703542 A EP 10703542A EP 10703542 A EP10703542 A EP 10703542A EP 2387616 A2 EP2387616 A2 EP 2387616A2
Authority
EP
European Patent Office
Prior art keywords
target rna
sequence
seq
complementary
nos
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP10703542A
Other languages
English (en)
French (fr)
Inventor
Cecilia Svanholm Barrie
Olivier Delfour
David H. Persing
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cepheid
Original Assignee
Cepheid
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 Cepheid filed Critical Cepheid
Priority to EP13150278.3A priority Critical patent/EP2641976A3/de
Publication of EP2387616A2 publication Critical patent/EP2387616A2/de
Withdrawn legal-status Critical Current

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
    • C12Q2600/158Expression markers
    • 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
    • C12Q2600/16Primer sets for multiplex assays
    • 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

  • Cervical cancer is the second most common cause of cancer-related mortality in women worldwide. Epidemiological and laboratory studies suggest a key role for human papillomavirus (HPV) in cervical carcinogenesis (Walboomers, J.M. et al.
  • HPV infection alone is not sufficient for cervical carcinogenesis, and additional steps occur over years or decades following initial infection. Most HPV infections resolve spontaneously, but if an oncogenic (high risk) HPV infection persists, there may be progression to a high grade cervical dysplasia or cervical cancer. (Nobbenhuis, M.A. et al. (2001) Lancet 358: 1782-1783). High risk HPVs include HPV- 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, 66, and 68, with HPV-16 and 18 accounting for up to 70% of cervical cancers worldwide.
  • HPV DNA testing can be more sensitive than cytologic testing in detecting high-grade cervical dysplasia.
  • HPV testing often has lower specificity than cytologic testing since most HPV infections are transient in nature.
  • CIN Cervical Intra-epithelial Neoplasia
  • the disclosure relates to a method for detecting the presence of cervical dysplasia in a human cervical sample.
  • the method comprises hybridizing nucleic acids of the sample with a polynucleotide that is complementary to a first target RNA in the sample or to a complement thereof and detecting a complex comprising polynucleotide hybridized to at least one nucleic acid selected from the first target RNA, a DNA amplicon of the first target RNA, and a complement of the first target RNA.
  • the first target RNA is capable of specifically hybridizing to a sequence selected from SEQ ID NO.:1 to 41 and 133 to 211.
  • the first target RNA comprises a sequence that is complementary to at least 15 contiguous nucleotides of a sequence selected from SEQ ID NO.:1 to 41 and 133 to 211. In some embodiments, the first target RNA comprises at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388. Detection of a level of the first complex that is greater than a normal level of the first complex indicates the presence of cervical dysplasia in the sample.
  • the sample comprises RNA that has been separated from DNA.
  • the first target RNA in its mature form, comprises fewer than 30 nucleotides.
  • the first target RNA is a microRNA.
  • the method further comprises hybridizing nucleic acids of the sample with a second polynucleotide that is complementary to a second target RNA in the sample or to a complement thereof, wherein the second RNA is different from the first target RNA, and detecting a second complex comprising the second polynucleotide hybridized to at least one nucleic acid selected from the second target RNA, a DNA amplicon of the second target RNA, and a complement of the second target RNA.
  • the second target RNA in its mature form, comprises fewer than 30 nucleotides.
  • the second target RNA is a microRNA. In other embodiments, the second target RNA is an mRNA.
  • a polynucleotide is complementary to at least 10 contiguous nucleotides of the target RNA or its complement. In some embodiments, a polynucleotide is complementary to at least 15 contiguous nucleotides of the target RNA or its complement. In some embodiments, a polynucleotide comprises a region that is not complementary to the target RNA or its complement.
  • a polynucleotide comprises a first region, wherein the first region comprises a sequence of at least 8 contiguous nucleotides, at least 9 contiguous nucleotides, at least 10 contiguous nucleotides, at least 11 contiguous nucleotides, or at least 12 contiguous nucleotides that are identical to a sequence of at least 8 contiguous nucleotides, at least 9 contiguous nucleotides, at least 10 contiguous nucleotides, at least 11 contiguous nucleotides, or at least 12 contiguous nucleotides of one or more of SEQ ID NOs: 1 to 7, 9 to 37, and 133 to 201.
  • a polynucleotide comprises a first region, wherein the first region comprises a sequence of at least 8 contiguous nucleotides, at least 9 contiguous nucleotides, at least 10 contiguous nucleotides, at least 11 contiguous nucleotides, or at least 12 contiguous nucleotides that are complementary to a sequence of at least 8 contiguous nucleotides, at least 9 contiguous nucleotides, at least 10 contiguous nucleotides, at least 11 contiguous nucleotides, or at least 12 contiguous nucleotides of one or more of SEQ ID NOs: 1 to 7, 9 to 37, and 133 to 201.
  • a polynucleotide comprises a first region, wherein the first region comprises a sequence of at least 8 contiguous nucleotides, at least 9 contiguous nucleotides, at least 10 contiguous nucleotides, at least 11 contiguous nucleotides, or at least 12 contiguous nucleotides that are identical to a sequence of at least 8 contiguous nucleotides, at least 9 contiguous nucleotides, at least 10 contiguous nucleotides, at least 11 contiguous nucleotides, or at least 12 contiguous nucleotides of one or more of SEQ ID NOs: 345 to 388.
  • a polynucleotide comprises a first region, wherein the first region comprises a sequence of at least 8 contiguous nucleotides, at least 9 contiguous nucleotides, at least 10 contiguous nucleotides, at least 11 contiguous nucleotides, or at least 12 contiguous nucleotides that are complementary to a sequence of at least 8 contiguous nucleotides, at least 9 contiguous nucleotides, at least 10 contiguous nucleotides, at least 11 contiguous nucleotides, or at least 12 contiguous nucleotides of one or more of SEQ ID NOs ⁇ 45-to-388.-
  • a polynucleotide comprises a detectable label. In some embodiments, a polynucleotide comprises a FRET label. In some embodiments, at least 8, at least 9, at least 10, at least 11, or at least 12 contiguous nucleotides of the polynucleotide are complementary to a target RNA. In some embodiments, a polynucleotide further comprises a second region that is not complementary to the target RNA.
  • a composition comprises a plurality of polynucleotides, wherein at least one, at least two, at least three, or at least five of the polynucleotides comprise a first region comprising a sequence of at least 8 contiguous nucleotides that is identical or complementary to a sequence of at least 8 contiguous nucleotides of one or more of SEQ ID NOs: 1 to 7, 9 to 37, 133 to 201, and 345 to 388.
  • kits are provided.
  • a kit comprises a composition described above.
  • a kit further comprises at least one polymerase.
  • a kit further comprises dNTPs.
  • FIG. 1 shows an exemplary electropherogram obtained on an Agilent Bioanalyser 2100 to assess the quality of total RNA purified as described in Example 1. Total RNA from cell line CaSki is shown.
  • FIG. 2 provides analysis by agarose gel electrophoresis under denaturing conditions of the quality of total RNA purified as described in Example 1 from cell lines CaSki, SW756, MEl 80, SiHA, C-4I, and C-4II.
  • FIG. 3 A and 3B show the Iog2 fold-changes ⁇ SD of certain mRNAs in tumor and normal samples relative to the Ambion normal sample, as discussed in Example 4. For each pair of bars, the left bar represents tumor samples and the right bar represents normal samples. [0021] FIG. 4 shows relative Iog2 fold changes of certain mRNAs in liquid PAP samples, as discussed in Example 5.
  • FIG. 5 shows relative Iog2 fold changes ⁇ SD of miR-205 in cervical tumor samples and normal tissue, as discussed in Example 6.
  • the left bar represents SCC samples
  • the middle bar represents ADC samples
  • the right bar represents normal samples.
  • FIG. 6 shows relative Iog2 fold changes ⁇ SD of miR-1290 in cervical tumor samples and normal tissue, as discussed in Example 6.
  • the left bar represents tumor samples and the right bar represents normal samples.
  • Methods of measuring levels of microRNA species disclosed herein are provided, wherein elevated levels of the microRNA species is indicative of cervical dysplasia.
  • methods are presented for detecting human cervical dysplasia, such as cervical dysplasia likely to progress to carcinoma.
  • the method comprises detecting an above-normal level of at least one target RNA that is capable of specifically hybridizing to a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211.
  • the method comprises detecting an above-normal level of at least one target RNA, wherein at least one target RNA comprises at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, or at least 24 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388. In some embodiments, the method comprises detecting an above-normal level of at least one target RNA that comprises a sequence that is complementary to at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, or at least 24 contiguous nucleotides of a sequence selected from SEQ ID NO.:1 to 41 and 133 to 211. In some embodiments, the target RNA, in its mature form, comprises fewer than 30 nucleotides. The target RNA, in some embodiments, is a microRNA.
  • a sequence selected from encompasses both “one sequence selected from” and “one or more sequences selected from.” Thus, when “a sequence selected from” is used, it is to be understood that one, or more than one, of the listed sequences may be chosen.
  • Detection of a level of target RNA that is greater than a normal level of target RNA indicates the presence of cervical dysplasia in the sample. In some embodiments, the detecting is done quantitatively. In other embodiments, the detecting is done qualitatively.
  • detecting a target RNA comprises forming a complex comprising a polynucleotide and a nucleic acid selected from a target RNA, a DNA amplicon of a target RNA, and a complement of a target RNA.
  • the level of the complex is then detected and compared to a normal level of the same complex.
  • the level of the complex correlates with the level of the target RNA in the sample.
  • Cervical dysplasia which is also known as cervical intraepithelial neoplasia (“CIN”), corresponds to precancerous changes of the cervix that are evidenced by an abnormal growth on the surface of the cervix. Cervical dysplasia is divided into three categories: CIN 1, which is mild dysplasia in which only a few cells are abnormal; CIN 2, which is moderate to marked dysplasia in which the abnormal cells involve about one-half of the thickness of the surface lining of the cervix; and CIN 3, which includes severe dysplasia to carcinoma-in-situ (i.e., precancerous cells limited to the top epithelial layer of the cervix). CIN 3 is unlikely to regress spontaneously, and if left untreated, can penetrate the basement membrane and become an invasive carcinoma.
  • Table 1 lists 41 hybridization probes that have been found to be complementary to, and hybridize with, target RNAs in cancer cells. These target RNAs were detected at elevated levels in certain human cervical cell lines that were assayed using microarrays (Example 1). Thirty-six of the probes are believed to be complementary to, and hybridize with, target RNA species that are expressed in human cells. The other five probes are complementary to, and hybridize with, publicly known microRNAs that have been deposited by others into miRBase (http://microrna.sanger.ac.uk/; see Griffiths-Jones S. et al. (2007) Nucl. Acids Res.
  • Table 11 lists hybridization probes that have been found to be complementary to, and hybridize with, target RNAs in cancer cells. These target RNAs were detected at elevated levels in certain human clinical cervix samples that were assayed using microarrays (Example 3). Seventy-three of the probes are believed to be complementary to, and hybridize with, target RNA species that are expressed in human cells. Four of those 73 probes were also detected at elevated levels in certain human cervical cell lines that were assayed using microarrays (Example 1), and are also in Table 1 (836-R4-1, 3371-L4-1, 9053-R3-1, and 9691-L4-1).
  • the remaining 19 probes are complementary to, and hybridize with, publicly known microRNAs that have been deposited by others into miRBase (http://microrna.sanger.ac.uk/; see Griffiths- Jones S. et al. (2007) Nucl. Acids Res. 36:154-158).
  • One of those 19 probes was also detected at elevated levels in certain human cervical cell lines that were assayed using microarrays (Example 1), and is also in Table 1 (miR-765).
  • microRNAs in Table 28 are isomirs of one another.
  • two isomirs have a common core sequence with one or both ends varying by one to three nucleotides.
  • AGCCGCTCTTCTCCCTGCCCACA SEQ ID NO: 355
  • AGCCGCTCTTCTCCCTGCCCACA SEQ ID NO: 356
  • CCCGGAGAGCGGAGCACAACACA SEQ ID NO: 346
  • CCGGAGAGCGGAGCACAAC (SEQ ID NO: 347) are isomirs.
  • a method comprises detecting multiple isomirs with a single probe. Detection of an elevated level of one or multiple isomirs is considered to be indicative of cervical dysplasia.
  • target RNA species are denominated “microRNAs” in the tables set forth herein and Example 1.
  • the target RNA is a single mature microRNA capable of specifically hybridizing to a hybridization probe set forth in Table 1 or Table 1 1.
  • a target RNA is a single mature microRNA that comprises a sequence that is complementary to at least 15 contiguous nucleotides of a sequence selected from SEQ ID NO.:1 to 41 and 133 to 211.
  • a target RNA is a single mature microRNA that comprises at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388.
  • target RNA may include a plurality of target RNAs, all of which are capable of specifically hybridizing to a single complementary probe sequence (for example, when two or more target microRNAs are isomirs).
  • the so-denominated "microRNA" is one or more RNA species capable of specifically hybridizing to the respective hybridization probe, such that one or more target RNAs do not meet canonical definitions for mature microRNAs.
  • a target RNA is an mRNA.
  • Mature human microRNAs are typically composed of 17-27 contiguous ribonucleotides, and often are 21 or 22 nucleotides in length.
  • the sequences of some target microRNAs that can be detected in accordance with the present disclosure can be found within the pre-microRNA sequences shown in Table 2 (SEQ ID NOs: 42 to 82) and Table 12 (SEQ ID NOs: 226 to 314).
  • the sequences of some microRNAs are shown in Table 28.
  • a microRNA comprises at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, or at least 26 contiguous nucleotides of a sequence in Table 28 (SEQ ID NOs: 345 to 388).
  • the sequences of the 23 publicly known mature microRNAs, obtained by query of miRBase, are also shown below in Table 3, along with the sequences of other previously known microRNAs that, in some embodiments, can be detected in the methods described herein.
  • mammalian microRNAs mature as described herein.
  • a gene coding for a microRNA is transcribed, leading to production of a microRNA precursor known as the "pri -microRNA" or "pri-miRNA.”
  • the pri-miRNA can be part of a polycistronic RNA comprising multiple pri-miRNAs.
  • the pri-miRNA forms a hairpin with a stem and loop, which may comprise mismatched bases.
  • the hairpin structure of the pri-miRNA is recognized by Drosha, which is an RNase III endonuclease protein.
  • Drosha can recognize terminal loops in the pri-miRNA and cleave approximately two helical turns into the stem to produce a 60-70 nucleotide precursor known as the "pre-microRNA” or "pre-miRNA.” Drosha can cleave the pri-miRNA with a staggered cut typical of RNase III endonucleases yielding a pre-miRNA stem loop with a 5' phosphate and an approximately 2-nucleotide 3' overhang. Approximately one helical turn of the stem (about 10 nucleotides) extending beyond the Drosha cleavage site can be essential for efficient processing.
  • the pre-miRNA is subsequently actively transported from the nucleus to the cytoplasm by Ran-GTP and the export receptor Exportin-5.
  • the pre-miRNA can be recognized by Dicer, another RNase III endonuclease. In some circumstances, Dicer recognizes the double-stranded stem of the pre-miRNA. Dicer may also recognize the 5' phosphate and 3' overhang at the base of the stem loop. Dicer may cleave off the terminal loop two helical turns away from the base of the stem loop leaving an additional 5' phosphate and an approximately 2-nucleotide 3' overhang.
  • the resulting siRNA-like duplex which may comprise mismatches, comprises the mature microRNA and a similar-sized fragment known as the microRNA*.
  • the microRNA and microRNA* may be derived from opposing arms of the pri-miRNA and pre-miRNA.
  • the mature microRNA is then loaded into the RNA-induced silencing complex ("RISC"), a ribonucleoprotein complex.
  • RISC RNA-induced silencing complex
  • the microRNA* also has gene silencing or other activity.
  • Table 1 the expression levels of target RNAs measured for each of the identified sample cell lines are expressed as fold-changes in expression relative to expression levels measured in normal human cervix total RNA (see Example 1).
  • target RNAs can be measured in samples collected at one or more times from a patient to monitor the status or progress of cervical dysplasia in the patient.
  • a sample to be tested is obtained using one or more techniques commonly used for preparing Pap smears, e.g., (i) endocervical swab, using a cotton applicator stick (or wire brush for endocervical specimens) advanced into the os of the cervix, with the stick gently rolled between the thumb and index finger; (ii) cervical scrape, in which the longer end of a cervical spatula is inserted into the os of the cervix and pressed gently, with turning and scraping.
  • the sample to be tested is a cervical biopsy, such as a punch biopsy or cone biopsy.
  • the sample to be tested is from a loop excision, or LEEP, procedure.
  • the clinical sample to be tested is, in some embodiments, freshly obtained. In other embodiments, the sample is a fresh frozen specimen. In some embodiments, the sample is a tissue sample, such as a formalin-fixed paraffin embedded sample. In some embodiments, the sample is a liquid cytology sample.
  • Exemplary liquid cytology preservative solutions include, but are not limited to, ThinPrepTM PreservCytTM solution (Hologic, Bedford, MA) and SurePathTM preservative solution (BD Diagnostics, NJ). Additional exemplary preservative solutions include, but are not limited to, RNAlater® (Ambion), formalin (e.g., 10% aqueous formalin), Universal Viral Transport Media (BD Diagnostics, NJ), M4, M4RT, PVA (polyvinyl-alcohol), PolyCyte (American Mastertech Scientific), Spray-Cyte cytological fixative (Becton-Dickinson), formaldehyde (e.g., 10% in phosphate buffer), NuFix Complete Collection Solution (QC Sciences), CarboFix (StatLab Medical Products), Cyto Jar (Surgipath Medical Industries), SED Fix (Surgipath Medical Industries), SprayFix (Surgipath Medical Industries), cytology fixative 50% alcohol solution (U.S.
  • RNAlater® Ambion
  • the clinical sample to be tested is obtained in conjunction with routine cytologic screening (e.g., by Pap smear), currently recommended for all women between the ages of 21 and 65, and women who are under 21 years old who have been sexually active for three years or more.
  • the sample to be tested is obtained from a woman who has a predisposition to develop cervical cancer, e.g., a woman who has tested positive for HPV infection, and especially positive for a high risk HPV type.
  • the clinical sample to be tested is obtained from women who have one or more of the following risk factors: multiparous, many sexual partners, first sexual intercourse at a young age, smoke cigarettes, use of oral contraceptives, and a weakened immune system.
  • the clinical sample is obtained from women who have diagnostic signs or clinical symptoms that may be associated with cervical cancer, such as abnormal Pap tests, abnormal bleeding or visible cervical lesions.
  • the methods described herein are used for early detection of cervical dysplasia in a sample of cervical cells, such as those obtained by routine Pap smear. In some embodiments, methods described herein can be used for early detection of cervical dysplasia in a sample of cervical cells, and to determine a likelihood that the detected cervical dysplasia will progress to cervical cancer.
  • methods of the present disclosure can be used for routine screening of healthy women with no risk factors.
  • methods herein are used to (1) screen women who have a history of abnormal Pap smears and/or of assays showing infection by one or more HPV strains associated with the development of cervical cancer, (2) screen women with one or more of the above- described risk factors, (3) confirm a diagnosis made by cytology, histology or HPV assay, and/or further characterize a diagnosis made by cytology or histology.
  • the methods described herein can be used to assess the effectiveness of a treatment for cervical cancer in a patient.
  • the target RNA expression levels are determined at various times during the treatment, and are compared to target RNA expression levels from an archival sample taken from the patient, e.g., by Pap smear, before the manifestation of any signs of cervical dysplasia or cervical cancer or before beginning treatment.
  • target RNA expression levels in the normal Pap smear sample evidence no aberrant changes in target RNA expression levels.
  • the progress of treatment of an individual with cervical dysplasia or cervical cancer can be assessed by comparison to a sample of cervical cells from the same individual when she was healthy or prior to beginning treatment.
  • a target RNA is capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7 and 8. In some embodiments, a target RNA is capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 and 12.
  • a target RNA is selected from miR-1246, miR-1308, miR- 491-3p, miR-1826, and miR-1290 (SEQ ID NOs: 208, 210, 205, 211, and 209), and target RNAs that are capable of specifically hybridizing to probes 13254-R5-1, 13252-L5-3, 13532-L5-2, 4440-L3-2, 6216-L1-1, and 6235-R5-2 (SEQ ID NOs: 194, 193, 172, 142, 151, and 153).
  • a target RNA is capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1, 5, 7, and 32.
  • a target RNA is capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1 to 41 set forth in Table 1 and SEQ ID NOs: 133 to 211 in Table 1 1.
  • a target RNA comprises at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388.
  • a target RNA comprises a sequence that is complementary to at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211.
  • a target RNA, in its mature form comprises fewer than 30 nucleotides.
  • a target RNA is a microRNA.
  • the expression levels of the plurality of target RNAs may be detected concurrently or simultaneously in the same assay reaction. In some embodiments, expression levels are detected concurrently or simultaneously in separate assay reactions. In some embodiments, expression levels are detected at different times, e.g., in serial assay reactions.
  • Table 2 identifies the chromosomal location of each of the 41 target RNAs capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1 to 41 in Table 1.
  • Table 12 identifies the chromosomal location of each of the target RNAs capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 133 to 211 in Table 11.
  • the level of expression of one or more target RNAs located within about 1 kilobase (kb), within about 2 kb, within about 5 kb, within about 10 kb, within about 20 kb, within about 30 kb, within about 40 kb, and even within about 50 kb of the chromosomal locations in Table 2 and Table 12 is detected in lieu of, or in addition to, measurement of expression of the respective tabulated target RNA in the methods described herein. See Baskerville, S. and Bartel D.P. (2005) RNA 11 :241-247.
  • methods herein further comprise detecting the level(s) of expression of at least one microRNA selected from miR-21, miR-31, miR-182, miR-183, miR-155, miR-9, miR-199a*, miR-199a, miR-199b, miR-205, miR-145, miR-133a, miR- 133b, miR-214, miR-127, miR-
  • methods herein further comprise detecting in a sample of human cervical cells the expression of at least one microRNA selected from miR-9, miR- 199a*, miR-199a, miR-199b, miR-145, miR-133a, miR-133b, miR-214 and miR-127 where invasive squamous cell cervical carcinoma is implicated.
  • an increase in expression of one or more microRNAs selected from miR-9, miR-199a*, miR-199a, miR-199b, miR-145, miR-133a, miR-133b, miR-214 and miR-127 in combination with an elevated level of one or more target RNAs capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211 and/or an elevated level of one or more target RNAs comprising at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388 and/or an elevated level of one or more target RNAs that comprise a sequence that is complementary to at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211, is indicative of the presence of cervical carcinoma in a sample of human cervical cells.
  • methods herein further comprise detecting in a sample of human cervical cells the expression of at least one microRNA selected from miR-210, miR-182 and miR-183 where human papilloma virus 16 ("HPV- 16") is implicated.
  • methods herein further comprise detecting in a sample of human cervical cells the expression of miR-146a in order to distinguish cervical cancer from pre-neoplastic lesions, e.g., HPV-infected cervical cells.
  • a target RNA in its mature form, comprises fewer than 30 nucleotides.
  • a target RNA is a microRNA.
  • the methods further comprise detecting in a sample of human cervical cells the expression of at least one target RNA gene located in close proximity to chromosomal features, such as cancer-associated genomic regions, fragile sites, and human papilloma virus integration sites.
  • methods herein in combination with detecting one or more target RNAs capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211 and/or detecting one or more target RNAs comprising at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388 and/or detecting one or more target RNAs that comprise a sequence that is complementary to at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211, methods herein further comprise detecting in a sample of human cervical cells the expression of at least one mRNA species.
  • the at least one mRNA is selected from the mRNAs for the genes set forth in Table 4, below. In some embodiments, at least one mRNA is selected from mRNAs for CDKN2A, MKI67, TOP2A, and MCM5. In some embodiments, at least one mRNA is selected from mRNAs for CDKN2A, MKI67, TOP2A, MCM5, BIRC5, MMP9, and MCM2.
  • an increase in expression of one or more mRNAs listed in the table above is indicative of the presence of cervical dysplasia or cervical cancer in a sample of human cervical cells.
  • more than one target RNA is detected simultaneously in a single reaction. In some embodiments, at least 2, at least 3, at least 5, or at least 10 target RNAs are detected simultaneously in a single reaction. In some embodiments, all target RNAs are detected simultaneously in a single reaction.
  • a normal level (a "control") for each target RNA can be determined as an average level or range that is characteristic of normal cervical cells or other reference material, against which the level measured in the sample can be compared.
  • the determined average or range of target RNA in normal subjects can be used as a benchmark for detecting above-normal levels of target RNA indicative of cervical dysplasia.
  • normal levels of target RNA can be determined using individual or pooled RNA-containing samples from one or more individuals, such as from patients undergoing hysterectomy for benign gynecologic disease.
  • determining a normal level of expression of a target RNA comprises detecting a complex comprising a probe hybridized to a nucleic acid selected from a target RNA, a DNA amplicon of the target RNA, and a complement of the target RNA. That is, in some embodiments, a normal level of expression can be determined by detecting a DNA amplicon of the target RNA, or a complement of the target RNA rather than the target RNA itself. In some embodiments, a normal level of such a complex is determined and used as a control. The normal level of the complex, in some embodiments, correlates to the normal level of the target RNA. Thus, when a normal level of a target is discussed herein, that level can, in some embodiments, be determined by detecting such a complex.
  • a control comprises RNA from cells of a single individual, e.g., a patient undergoing hysterectomy for benign gynecologic disease.
  • a control comprises RNA from a pool of cells from multiple individuals.
  • a control is drawn from anatomically and/or cytologically normal areas of the cervix of the individual from whom the test sample was obtained.
  • a control comprises commercially-available human RNA, such as, for example, human cervix total RNA (Ambion; AM6992).
  • a normal level or normal range has already been predetermined prior to testing a sample for an elevated level.
  • the normal level of target RNA can be determined from one or more continuous cell lines, typically cell lines previously shown to have expression levels of the at least one target RNA that approximate the level of expression in normal cervical cells.
  • a method comprises detecting the level of expression of at least one target RNA. In some embodiments, a method further comprises comparing the level of expression of at least one target RNA to a normal level of expression of the at least one target RNA. In some embodiments, a method further comprises comparing the level of expression of at least one target RNA to a control level of expression of the at least one target RNA.
  • a control level of expression of the at least one target RNA is, in some embodiments, the level of expression of the at least one target RNA in a normal cell. In some such embodiments, a control level may be referred to as a normal level. In some embodiments, a greater level of expression of the at least one target RNA relative to the level of expression of the at least one target RNA in a normal cell indicates cervical dysplasia.
  • the level of expression of the at least one target RNA is compared to a reference level of expression, e.g., from a confirmed cervical dysplasia. In some such embodiments, a similar level of expression of the at least one target RNA relative to the reference sample indicates cervical dysplasia.
  • a level of expression of at least one target RNA that is at least about two-fold greater than a normal level of expression of the respective at least one target RNA indicates the presence of cervical dysplasia.
  • a level of expression of at least one target RNA that is at least about two-fold greater than the level of the respective at least one target RNA in a control sample comprised of normal cells indicates the presence of a cervical dysplasia.
  • a level of expression of at least one target RNA that is at least about 3-fold, at least about 4- fold, at least about 5-fold, at least about 6-fold, at least about 7-fold, at least about 8-fold, at least about 9-fold, or at least about 10-fold greater than the level of expression of the respective at least one target RNA in a control sample comprised of normal cells indicates the presence of cervical dysplasia.
  • a level of expression of at least one target RNA that is at least about 3-fold, at least about 4-fold, at least about 5- fold, at least about 6-fold, at least about 7-fold, at least about 8-fold, at least about 9-fold, or at least about 10-fold greater than a normal level of expression of the at least one target RNA indicates the presence of cervical dysplasia.
  • an increase in expression of one or more target RNAs capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1 to 41 in Table 1 and SEQ ID NOs: 133 to 211 in Table 11 is indicative of the presence of cervical dysplasia or cervical cancer in a sample of human cervical cells.
  • an increase in expression of one or more target RNAs comprising at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388 is indicative of the presence of cervical dysplasia or cervical cancer in a sample of human cervical cells.
  • an increase in expression of one or more target RNAs comprising a sequence that is complementary to at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211 is indicative of the presence of cervical dysplasia or cervical cancer in a sample of human cervical cells.
  • an increase in expression of one or more target RNAs capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1 to 41 in Table 1 and SEQ ID NOs: 133 to 211 in Table 11 is indicative of the presence of cervical dysplasia in a sample of human cervical cells that is likely to proceed to cervical cancer.
  • an increase in expression of one or more target RNAs comprising at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388 is indicative of the presence of cervical dysplasia in a sample of human cervical cells that is likely to proceed to cervical cancer.
  • an increase in expression of one or more target RNAs comprising a sequence that is complementary to at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211 is indicative of the presence of cervical dysplasia in a sample of human cervical cells that is likely to proceed to cervical cancer.
  • a control level of expression of a target RNA is determined contemporaneously, such as in the same assay or batch of assays, as the level of expression of the target RNA in a sample. In some embodiments, a control level of expression of a target RNA is not determined contemporaneously as the level of expression of the target RNA in a sample. In some such embodiments, the control level of expression has been determined previously.
  • the level of expression of a target RNA is not compared to a control level of expression, for example, when it is known that the target RNA is expressed at very low levels, or not at all, in normal cells. In such embodiments, detection of a high level of the target RNA in a sample is indicative of cervical dysplasia.
  • Target RNA can be prepared by any appropriate method.
  • Total RNA can be isolated by any method, including, but not limited to, the protocols set forth in Wilkinson, M. (1988) Nucl. Acids Res. 16(22): 10,933; and Wilkinson, M. (1988) Nucl. Acids Res. 16(22): 10934, or by using commercially-available kits or reagents, such as the TRIzol® reagent (InvitrogenTM), Total RNA Extraction Kit (iNtRON Biotechnology), Total RNA Purification Kit (Norgen Biotek Corp.), RNAqueousTM (Ambion), MagMAXTM (Ambion), RecoverAllTM (Ambion), RNeasy (Qiagen), etc.
  • small RNAs are isolated or enriched.
  • small RNA refers to RNA molecules smaller than about 200 nucleotides (nt) in length.
  • small RNA refers to RNA molecules smaller than about 100 nt, smaller than about 90 nt, smaller than about 80 nt, smaller than about 70 nt, smaller than about 60 nt, smaller than about 50 nt, or smaller than about 40 nt.
  • Enrichment of small RNAs can be accomplished by method. Such methods include, but are not limited to, methods involving organic extraction followed by adsorption of nucleic acid molecules on a glass fiber filter using specialized binding and wash solutions, and methods using spin column purification. Enrichment of small RNAs may be accomplished using commercially-available kits, such as mirVanaTM Isolation Kit (Applied Biosystems), mirPremierTM microRNA Isolation Kit (Sigma- Aldrich), PureLinkTM miRNA Isolation Kit (Invitrogen), miRCURYTM RNA isolation kit (Exiqon), microRNA Purification Kit (Norgen Biotek Corp.), miRNeasy kit (Qiagen), etc.
  • mirVanaTM Isolation Kit Applied Biosystems
  • mirPremierTM microRNA Isolation Kit Sigma- Aldrich
  • PureLinkTM miRNA Isolation Kit Invitrogen
  • miRCURYTM RNA isolation kit Exiqon
  • microRNA Purification Kit Norgen Biotek Corp.
  • purification can be accomplished by the TRIzol® (Invitrogen) method, which employs a phenol/isothiocyanate solution to which chloroform is added to separate the RNA-containing aqueous phase. Small RNAs are subsequently recovered from the aqueous by precipitation with isopropyl alcohol. In some embodiments, small RNAs can be purified using chromatographic methods, such as gel electrophoresis using the flashPAGETM Fractionator available from Applied Biosystems.
  • small RNA is isolated from other RNA molecules to enrich for target RNAs, such that the small RNA fraction (e.g., containing RNA molecules that are 200 nucleotides or less in length, such as less than 100 nucleotides in length, such as less than 50 nucleotides in length, such as from about 10 to about 40 nucleotides in length) is substantially pure, meaning it is at least about 80%, 85%, 90%, 95% pure or more, but less than 100% pure, with respect to larger RNA molecules.
  • enrichment of small RNA can be expressed in terms of fold-enrichment.
  • small RNA is enriched by about, at least about, or at most about 5X, 10X, 2OX, 30X, 4OX, 50X, 6OX, 7OX, 80X, 9OX, 10OX, HOX, 120X, 130X, 140X, 150X, 160X, 170X, 180X, 190X, 200X, 210X, 220X, 230X, 240X, 250X, 260X, 270X, 280X, 290X, 300X, 310X, 320X, 330X, 340X, 350X, 360X, 370X, 380X, 390X, 400X, 410X, 420X, 430X, 440X, 450X, 460X, 470X, 480X, 490X, 500X, 600X, 700X, 800X, 900X, 100OX, HOOX, 1200X, 130OX, 1400X, 150OX, 1600X, 1700X, 1800X,
  • expression is measured in a sample in which RNA has not first been purified from the cells.
  • RNA is modified before target RNAs are detected.
  • the modified RNA is total RNA.
  • the modified RNA is small RNA that has been purified from total RNA or from cell lysates, such as RNA less than 200 nucleotides in length, such as less than 100 nucleotides in length, such as less than 50 nucleotides in length, such as from about 10 to about 40 nucleotides in length.
  • RNA modifications that can be utilized in the methods described herein include, but are not limited to, the addition of a poly-dA or a poly-dT tail, which can be accomplished chemically or enzymatically, and/or the addition of a small molecule, such as biotin.
  • one or more target RNAs are reverse transcribed.
  • RNA is modified when it is reverse transcribed, such as when a poly-dA or a poly-dT tail is added to the cDNA during reverse transcription.
  • RNA is modified before it is reverse transcribed.
  • total RNA is reverse transcribed.
  • small RNAs are isolated or enriched before the RNA is reverse transcribed.
  • a complement of the target RNA is formed.
  • the complement of the target RNA is detected rather than the target RNA itself (or a DNA copy thereof).
  • detection or determination may be carried out on a complement of the target RNA instead of, or in addition to, the target RNA itself.
  • a probe is used that is complementary to the complement of the target RNA.
  • the probe comprises at least a portion that is identical in sequence to the target RNA, although it may contain thymidine in place of uridine, and/or comprise other modified nucleotides.
  • the method of detecting one or more target RNAs comprises amplifying cDNA complementary to said target RNA.
  • amplification can be accomplished by any method. Exemplary methods include, but are not limited to, real time PCR, endpoint PCR, and amplification using T7 polymerase from a T7 promoter annealed to a cDNA, such as provided by the SenseAmp PlusTM Kit available at Implen, Germany.
  • a DNA amplicon of a target RNA is formed.
  • a DNA amplicon may be single stranded or double-stranded.
  • the sequence of the DNA amplicon is related to the target RNA in either the sense or antisense orientation.
  • the DNA amplicon of the target RNA is detected rather than the target RNA itself.
  • a target RNA when the methods discussed herein indicate that a target RNA is detected, or the level of a target RNA is determined, such detection or determination may be carried out on a DNA amplicon of the target RNA instead of, or in addition to, the target RNA itself.
  • a probe when the DNA amplicon of the target RNA is detected rather than the target RNA, a probe is used that is complementary to the complement of the target RNA.
  • a probe is used that is complementary to the target RNA.
  • multiple probes may be used, and some probes may be complementary to the target RNA and some probes may be complementary to the complement of the target RNA.
  • the method of detecting one or more target RNAs comprises RT-PCR, as described below.
  • detecting one or more target RNAs comprises real-time monitoring of an RT-PCR reaction, which can be accomplished by any method.
  • methods include, but are not limited to, the use of TaqMan®, Molecular beacon, or Scorpion probes (i.e., FRET probes) and the use of intercalating dyes, such as SYBR green, EvaGreen, thiazole orange, YO-PRO, TO-PRO, etc.
  • the method comprises detecting a level of expression of at least one target RNA capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1 to 41 set forth in Table 1 and SEQ ID NOs: 133 to 211 set forth in Table 11 that is greater in the sample than a normal level of expression of the at least one target RNA in a control sample, such as a sample derived from normal cervical cells.
  • a method comprises detecting a level of one or more target RNAs comprising at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388 that is greater in the sample than a normal level of expression of the at least one target RNA in a control sample. In some embodiments, a method comprises detecting a level of one or more target RNAs that comprise a sequence that is complementary to at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211 that is greater in the sample than a normal level of expression of the at least one target RNA in a control sample. In some embodiments, a target RNA, in its mature form, comprises fewer than 30 nucleotides. In some embodiments, a target RNA is a microRNA.
  • the method further comprises detecting a level of expression of at least one target RNA of the human miRNome that does not specifically hybridize to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1 to 41 set forth in Table 1 and SEQ ID NOs: 133 to 211 set forth in Table 11 that is greater in the sample than a normal level of expression of the at least one target RNA in a control sample.
  • a nucleic acid comprising a sequence selected from SEQ ID NOs: 1 to 41 set forth in Table 1 and SEQ ID NOs: 133 to 211 set forth in Table 11 that is greater in the sample than a normal level of expression of the at least one target RNA in a control sample.
  • the term "human miRNome” refers to all microRNA genes in a human cell and the mature microRNAs produced therefrom.
  • Any analytical procedure capable of permitting specific and quantifiable (or semi-quantifiable) detection of the desired at least one target RNA may be used in the methods herein presented.
  • Such analytical procedures include, but are not limited to, the microarray methods set forth in Example 1 and the RT-PCR methods set forth in Example 6, and methods known to those skilled in the art.
  • detection of a target RNA comprises forming a complex comprising a polynucleotide that is complementary to a target RNA or to a complement thereof, and a nucleic acid selected from the target RNA, a DNA amplicon of the target RNA, and a complement of the target RNA.
  • the polynucleotide forms a complex with a target RNA.
  • the polynucleotide forms a complex with a complement of the target RNA, such as a cDNA that has been reverse transcribed from the target RNA.
  • the polynucleotide forms a complex with a DNA amplicon of the target RNA.
  • the complex may comprise one or both strands of the DNA amplicon.
  • a complex comprises only one strand of the DNA amplicon.
  • a complex is a triplex and comprises the polynucleotide and both strands of the DNA amplicon.
  • the complex is formed by hybridization between the polynucleotide and the target RNA, complement of the target RNA, or DNA amplicon of the target RNA.
  • the polynucleotide in some embodiments, is a primer or probe.
  • a method comprises detecting the complex.
  • the complex does not have to be associated at the time of detection. That is, in some embodiments, a complex is formed, the complex is then dissociated or destroyed in some manner, and components from the complex are detected.
  • An example of such a system is a TaqMan® assay.
  • detection of the complex may comprise amplification of the target RNA, a complement of the target RNA, or a DNA amplicon of a target RNA.
  • the analytical method used for detecting at least one target RNA in the methods set forth herein includes real-time quantitative RT-PCR. See Chen, C. et al. (2005) Nucl. Acids Res. 33:el79 and PCT Publication No. WO 2007/117256, which are incorporated herein by reference in its entirety.
  • the analytical method used for detecting at least one target RNA includes the method described in U.S. Publication No. US2009/0123912 Al, which is incorporated herein by reference in its entirety.
  • an extension primer comprising a first portion and second portion, wherein the first portion selectively hybridizes to the 3' end of a particular microRNA and the second portion comprises a sequence for universal primer, is used to reverse transcribe the microRNA to make a cDNA.
  • a reverse primer that selectively hybridizes to the 5' end of the microRNA and a universal primer are then used to amplify the cDNA in a quantitative PCR reaction.
  • the analytical method used for detecting at least one target RNA includes the use of a TaqMan® probe.
  • the analytical method used for detecting at least one target RNA includes a TaqMan® assay, such as the TaqMan® MicroRNA Assays sold by Applied Biosystems, Inc.
  • a TaqMan® assay such as the TaqMan® MicroRNA Assays sold by Applied Biosystems, Inc.
  • total RNA is isolated from the sample.
  • the assay can be used to analyze about 10 ng of total RNA input sample, such as about 9 ng of input sample, such as about 8 ng of input sample, such as about 7 ng of input sample, such as about 6 ng of input sample, such as about 5 ng of input sample, such as about 4 ng of input sample, such as about 3 ng of input sample, such as about 2 ng of input sample, and even as little as about 1 ng of input sample containing microRNAs.
  • the TaqMan® assay utilizes a stem-loop primer that is specifically complementary to the 3 '-end of a target RNA.
  • hybridizing the stem-loop primer to the target RNA is followed by reverse transcription of the target RNA template, resulting in extension of the 3' end of the primer.
  • the result of the reverse transcription is a chimeric (DNA) amplicon with the step-loop primer sequence at the 5' end of the amplicon and the cDNA of the target RNA at the 3' end.
  • Quantitation of the target RNA is achieved by real time RT-PCR using a universal reverse primer having a sequence that is complementary to a sequence at the 5 ' end of all stem- loop target RNA primers, a target RNA-specific forward primer, and a target RNA sequence-specific TaqMan® probe.
  • the assay uses fluorescence resonance energy transfer ("FRET") to detect and quantitate the synthesized PCR product.
  • the TaqMan® probe comprises a fluorescent dye molecule coupled to the 5 '-end and a quencher molecule coupled to the 3 '-end, such that the dye and the quencher are in close proximity, allowing the quencher to suppress the fluorescence signal of the dye via FRET.
  • FRET fluorescence resonance energy transfer
  • the TaqMan® probe comprises a fluorescent dye molecule coupled to the 5 '-end and a quencher molecule coupled to the 3 '-end, such that the dye and the quencher are in close proximity, allowing the quencher to suppress the fluorescence signal of the dye via FRET.
  • the polymerase replicates the chimeric amplicon template to which the TaqMan® probe is bound
  • the 5 '-nuclease of the polymerase cleaves the probe, decoupling the dye and the quencher so that FRET is abolished and
  • RNA detection and/or quantification are described, e.g., in U.S. Publication No. US 2007/0077570 (Lao et al.), PCT Publication No. WO 2007/025281 (Tan et al.), U.S. Publication No. US2007/0054287 (Bloch), PCT Publication No. WO2006/0130761 (Bloch), and PCT Publication No. WO 2007/011903 (Lao et al.), which are incorporated by reference herein in their entireties for any purpose.
  • quantitation of the results of real-time RT-PCR assays is done by constructing a standard curve from a nucleic acid of known concentration and then extrapolating quantitative information for target RNAs of unknown concentration.
  • the nucleic acid used for generating a standard curve is an RNA (e.g., microRNA) of known concentration.
  • the nucleic acid used for generating a standard curve is a purified double- stranded plasmid DNA or a single-stranded DNA generated in vitro.
  • Ct values are inversely proportional to the amount of nucleic acid target in a sample.
  • Ct values of the target RNA of interest can be compared with a control or calibrator, such as RNA (e.g., microRNA) from normal tissue.
  • the Ct values of the calibrator and the target RNA samples of interest are normalized to an appropriate endogenous housekeeping gene.
  • real-time RT-PCR chemistries useful for detecting and quantitating PCR products in the methods presented herein include, but are not limited to, Molecular Beacons, Scorpion probes and intercalating dyes, such as SYBR Green, EvaGreen, thiazole orange, YO-PRO, TO-PRO, etc., which are discussed below.
  • real-time RT-PCR detection is performed specifically to detect and quantify the expression of a single target RNA.
  • the target RNA in some embodiments, is selected from a target RNA capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1 to 41 set forth in Table 1 and SEQ ID NOs: 133 to 211 set forth in Table 11.
  • the target RNA specifically hybridizes to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, or 8.
  • the target RNA specifically hybridizes to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12.
  • the target RNA specifically hybridizes to a nucleic acid comprising a sequence selected from SEQ ID NOs: 142, 151, 153, 193, 194, 205, 172, 208, 210, and 211. In some embodiments, the target RNA specifically hybridizes to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1, 5, 7, and 32. In some embodiments, the target RNA comprises at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388. In some embodiments, the target RNA comprises a sequence that is complementary to at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211. In some embodiments, a target RNA, in its mature form, comprises fewer than 30 nucleotides. In some embodiments, a target RNA is a microRNA.
  • the methods comprise detection of expression of one or more microRNAs selected from miR-21, miR-31, miR-182, miR-183, miR-155, miR-9, miR-199a*, miR-199a, miR-199b, miR-145, miR-133a, miR-133b, miR-214, miR-127, miRNAs selected from miR-21, miR-31, miR-182, miR-183, miR-155, miR-9, miR-199a*, miR-199a, miR-199b, miR-145, miR-133a, miR-133b, miR-214, miR-127, mi
  • real-time RT-PCR detection is utilized to detect, in a single multiplex reaction, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, or at least 8 target RNAs.
  • At least one target RNA in some embodiments, is capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211.
  • at least one target RNA comprises at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388.
  • At least one target RNA comprises a sequence that is complementary to at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211.
  • a target RNA in its mature form, comprises fewer than 30 nucleotides.
  • a target RNA is a microRNA.
  • the method comprises detecting expression in a multiplex RT-PCR reaction of at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, or at least 8 target RNAs, wherein each target RNA is capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, and 8.
  • the method comprises detecting greater than normal expression, using a single multiplex RT-PCR reaction, of at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, or at least 12 target RNAs, wherein each target RNA is capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 and 12.
  • the method comprises detecting greater than normal expression, using a single multiplex RT-PCR reaction, of at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, or at least 12 target RNAs, wherein each target RNA is capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 142, 151, 153, 193, 194, 205, 172, 208, 210, and 211.
  • the method comprises detecting expression in a multiplex RT-PCR reaction of at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, or at least 9 target RNAs, wherein each target RNA is capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1, 4, 5, 7, 12, 17, 25, 26, 32.
  • a plurality of probes such as TaqMan® probes, each specific for a different RNA target, is used.
  • each target RNA-specific probe is spectrally distinguishable from the other probes used in the same multiplex reaction.
  • quantitation of real-time RT PCR products is accomplished using a dye that binds to double-stranded DNA products, such as SYBR Green, EvaGreen, thiazole orange, YO-PRO, TO-PRO, etc.
  • the assay is the QuantiTect SYBR Green PCR assay from Qiagen. In this assay, total RNA is first isolated from a sample. Total RNA is subsequently poly-adenylated at the 3 '-end and reverse transcribed using a universal primer with poly-dT at the 5 '-end. In some embodiments, a single reverse transcription reaction is sufficient to assay multiple target RNAs.
  • Real-time RT-PCR is then accomplished using target RNA-specific primers and an miScript Universal Primer, which comprises a poly-dT sequence at the 5 '-end.
  • SYBR Green dye binds non-specifically to double-stranded DNA and upon excitation, emits light.
  • buffer conditions that promote highly-specific annealing of primers to the PCR template e.g., available in the QuantiTect SYBR Green PCR Kit from Qiagen
  • the signal from SYBR Green increases, allowing quantitation of specific products.
  • Real-time RT-PCR is performed using any RT-PCR instrumentation available in the art.
  • instrumentation used in real-time RT-PCR data collection and analysis comprises a thermal cycler, optics for fluorescence excitation and emission collection, and optionally a computer and data acquisition and analysis software.
  • the analytical method used in the methods described herein is a DASL® (cDNA-mediated Annealing, Selection, Extension, and Ligation) Assay, such as the MicroRNA Expression Profiling Assay available from Illumina, Inc. (See http://www.illumina.com/downloads/MicroRNAAssayWorkflow.pdf).
  • total RNA is isolated from a sample to be analyzed by any method.
  • small RNAs are isolated from a sample to be analyzed by any method. Total RNA or isolated small RNAs may then be polyadenylated (> 18 A residues are added to the 3 '-ends of the RNAs in the reaction mixture).
  • the RNA is reverse transcribed using a biotin-labeled DNA primer that comprises from the 5' to the 3' end, a sequence that includes a PCR primer site and a poly-dT region that binds to the poly-dA tail of the sample RNA.
  • the resulting biotinylated cDNA transcripts are then hybridized to a solid support via a biotin- streptavidin interaction and contacted with one or more target RNA-specific polynucleotides.
  • the target RNA-specific polynucleotides comprise, from the 5 '-end to the 3 '-end, a region comprising a PCR primer site, region comprising an address sequence, and a target RNA-specific sequence.
  • the target RNA-specific sequence comprises at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, or at least 24 contiguous nucleotides having a sequence identically present in one of SEQ ID NOs: 1 to 41 and 133 to 211.
  • the target RNA- specific sequence comprises a probe sequence that is complementary to at least a portion of a microRNA of the human miRNome, such as miR-21, miR-31, miR-182, miR-183, miR-155, miR-9, miR-199a*, miR-199a, miR-199b, miR-145, miR-133a, miR-133b, miR-205, miR-214, miR-127, miR-210, miR-146a, miR-301, miR-142-5p, miR-194, miR-215, miR-32, miR-374b, miR-933, miR-769-3p, miR-671, miR-934, miR-935, miR- 937, miR-938, miR-939, miR-940, miR-941, miR-942, miR-943, miR-944, miR-708, miR-874-5p, and miR-874-3p.
  • the target RNA-specific polynucleotide is extended, and the extended products are then eluted from the immobilized cDNA array.
  • a second PCR reaction using a fluorescently-labeled universal primer generates a fluorescently-labeled DNA comprising the target RNA-specific sequence.
  • the labeled PCR products are then hybridized to a microbead array for detection and quantitation.
  • the analytical method used for detecting and quantifying the expression of the at least one target RNA in the methods described herein is a bead-based flow cytometric assay. See Lu J. et al. (2005) Nature 435:834-838, which is incorporated herein by reference in its entirety.
  • An example of a bead-based flow cytometric assay is the xMAP® technology of Luminex, Inc. (See http://www.luminexcorp.com/ technology/index. html).
  • total RNA is isolated from a sample and is then labeled with biotin.
  • RNA-specific capture probes e.g., FlexmiRTM products sold by Luminex, Inc. at http://www.luminexco ⁇ .com/products/assays/index.html
  • a streptavidin-bound reporter molecule e.g., streptavidin- phycoerythrin, also known as "SAPE"
  • SAPE streptavidin- phycoerythrin
  • the RNA sample (total RNA or enriched small RNAs) is first polyadenylated, and is subsequently labeled with a biotinylated 3DNATM dendrimer (i.e., a multiple-arm DNA with numerous biotin molecules bound thereto), such as those sold by Marligen Biosciences as the VantageTM microRNA Labeling Kit, using a bridging polynucleotide that is complementary to the 3 '-end of the poly-dA tail of the sample RNA and to the 5 '-end of the polynucleotide attached to the biotinylated dendrimer.
  • a biotinylated 3DNATM dendrimer i.e., a multiple-arm DNA with numerous biotin molecules bound thereto
  • a bridging polynucleotide that is complementary to the 3 '-end of the poly-dA tail of the sample RNA and to the 5 '-end of the polynucleotide attached to the bio
  • biotin-labeled RNA is first exposed to SAPE, and the RNA/SAPE complex is subsequently exposed to an anti-phycoerythrin antibody attached to a DNA dendrimer, which can be bound to as many as 900 biotin molecules. This allows multiple SAPE molecules to bind to the biotinylated dendrimer through the biotin-streptavidin interaction, thus increasing the signal from the assay.
  • the analytical method used for detecting and quantifying the expression of the at least one target RNA in the methods described herein is by gel electrophoresis and detection with labeled probes (e.g., probes labeled with a radioactive or chemiluminescent label), such as by Northern blotting.
  • labeled probes e.g., probes labeled with a radioactive or chemiluminescent label
  • Northern blotting e.g., total RNA is isolated from the sample, and then is size-separated by SDS polyacrylamide gel electrophoresis. The separated RNA is then blotted onto a membrane and hybridized to radiolabeled complementary probes.
  • exemplary probes contain one or more affinity-enhancing nucleotide analogs as discussed below, such as locked nucleic acid (“LNA”) analogs, which contain a bicyclic sugar moiety instead of deoxyribose or ribose sugars.
  • LNA locked nucleic acid
  • the total RNA sample can be further purified to enrich for small RNAs.
  • target RNAs can be amplified by, e.g., rolling circle amplification using a long probe that is complementary to both ends of a target RNA ("padlocked probes"), ligation to circularize the probe followed by rolling circle replication using the target RNA hybridized to the circularized probe as a primer.
  • rolling circle amplification using a long probe that is complementary to both ends of a target RNA ("padlocked probes")
  • ligation to circularize the probe followed by rolling circle replication using the target RNA hybridized to the circularized probe as a primer.
  • the amplified product can then be detected and quantified using, e.g., gel electrophoresis and Northern blotting.
  • labeled probes are hybridized to isolated total RNA in solution, after which the RNA is subjected to rapid ribonuclease digestion of single-stranded RNA, e.g., unhybridized portions of the probes or unhybridized target RNAs.
  • the ribonuclease treated sample is then analyzed by SDS-PAGE and detection of the radiolabeled probes by, e.g., Northern blotting. See mirVanaTM miRNA Detection Kit sold by Applied Biosystems, Inc. product literature at http://www.ambion.com/catalog/CatNum.php71552.
  • the analytical method used for detecting and quantifying the at least one target RNA in the methods described herein is by hybridization to a microarray. See, e.g., Liu, CG. et al. (2004) Proc. Nat'l Acad. Sci. USA 101:9740-9744; Lim, L.P. et al. (2005) Nature 433:769-773, each of which is incorporated herein by reference in its entirety, and Example 1.
  • RNA using a microarray is accomplished by surface plasmon resonance. See, e.g., Nanotech News (2006), available at http://nano.cancer.gov/news_center/nanotech_news_2006-l 0-30b. asp.
  • total RNA is isolated from a sample being tested.
  • the RNA sample is further purified to enrich the population of small RNAs. After purification, the RNA sample is bound to an addressable microarray containing probes at defined locations on the microarray.
  • Nonlimiting exemplary probes include probes comprising sequences set forth in SEQ ID NOs: 1 to 41 and 133 to 211.
  • Exemplary probes also include, but are not limited to, probes comprising a region that is complementary to at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388.
  • Exemplary probes also include, but are not limited to, probes comprising at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211.
  • the probes contain one or more affinity-enhancing nucleotide analogs as discussed below, such as locked nucleic acid (“LNA”) nucleotide analogs.
  • LNA locked nucleic acid
  • the RNA that is hybridized to the array is first polyadenylated, and the array is then exposed to gold particles having poly-dT bound to them. The amount of bound target RNA is quantitated using surface plasmon resonance.
  • microarrays are utilized in a RNA-primed
  • RNA 12(2): 1-5 each of which is incorporated herein by reference in its entirety.
  • total RNA is isolated from a sample.
  • small RNAs are isolated from a sample. The RNA sample is then hybridized to DNA probes immobilized at the 5 '-end on an addressable array.
  • the DNA probes comprise, in some embodiments, from the 5 '-end to the 3 '-end, a first region comprising a "spacer" sequence which is the same for all probes, a second region comprising three thymidine-containing nucleosides, and a third region comprising a sequence that is complementary to a target RNA of interest.
  • target RNAs of interest include, but are not limited to, target RNAs capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211, and target RNAs comprising at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388, and target RNAs comprising a region that is complementary to at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211.
  • Target RNAs also include target RNAs in the miRNome that do not specifically hybridize to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211.
  • a target RNA in its mature form, comprises fewer than 30 nucleotides.
  • a target RNA is a microRNA.
  • the sample is hybridized to the array, it is exposed to exonuclease I to digest any unhybridized probes.
  • the Klenow fragment of DNA polymerase I is then applied along with biotinylated dATP, allowing the hybridized target RNAs to act as primers for the enzyme with the DNA probe as template.
  • the slide is then washed and a streptavidin-conjugated fluorophore is applied to detect and quantitate the spots on the array containing hybridized and Klenow-extended target RNAs from the sample.
  • the RNA sample is reverse transcribed.
  • the RNA sample is reverse transcribed using a biotin/poly-dA random octamer primer.
  • primer When than primer is used, the RNA template is digested and the biotin-containing cDNA is hybridized to an addressable microarray with bound probes that permit specific detection of target RNAs.
  • the microarray includes at least one probe comprising at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, or at least 24 contiguous nucleotides identically present in, or complementary to a region of, a sequence selected from SEQ ID NOs: 1 to 41, 133 to 211, and 345 to 388.
  • the microarray is exposed to a streptavidin-bound detectable marker, such as a fluorescent dye, and the bound cDNA is detected. See Liu CG. et al. (2008) Methods 44:22-30, which is incorporated herein by reference in its entirety.
  • target RNAs are detected and quantified in an ELISA-like assay using probes bound in the wells of microtiter plates. See Mora J.R. and Getts R.C. (2006) BioTechniques 41 :420-424 and supplementary material in BioTechniques 41(4): 1-5; U.S. Patent Publication No. 2006/0094025 to Getts et al., each of which is incorporated by reference herein in its entirety.
  • a sample of RNA that is enriched in small RNAs is either polyadenylated, or is reverse transcribed and the cDNA is polyadenylated.
  • RNA or cDNA is hybridized to probes immobilized in the wells of a microtiter plates, wherein each of the probes comprises a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1 to 41, 133 to 211, or 345 to 388, or a sequence such as one or more sequences of target RNAs (or the reverse complement thereof) of the human miRNome, depending on whether RNA or cDNA is hybridized to the array.
  • the hybridized RNAs are labeled using a capture sequence, such as a DNA dendrimer (such as those available from Genisphere, Inc., http://www.genisphere.com/about 3dna.html) that is labeled with a plurality of biotin molecules or with a plurality of horseradish peroxidase molecules, and a bridging polynucleotide that contains a poly-dT sequence at the 5 '-end that binds to the poly-dA tail of the captured nucleic acid, and a sequence at the 3 '-end that is complementary to a region of the capture sequence.
  • a capture sequence such as a DNA dendrimer (such as those available from Genisphere, Inc., http://www.genisphere.com/about 3dna.html) that is labeled with a plurality of biotin molecules or with a plurality of horseradish peroxidase molecules, and a bridging polynucleotide that contains a
  • the microarray is then exposed to streptavidin-bound horseradish peroxidase. Hybridization of target RNAs is detected by the addition of a horseradish peroxidase substrate such as tetramethylbenzidine (TMB) and measurement of the absorbance of the solution at 45OnM.
  • a horseradish peroxidase substrate such as tetramethylbenzidine (TMB)
  • total RNA is isolated from a sample.
  • small RNAs are isolated from the sample. The 3 '-ends of the target RNAs are biotinylated using biotin-X- hydrazide.
  • the biotinylated target RNAs are captured on a microarray comprising immobilized probes comprising sequences that are identically present in, or complementary to a region of, one or more of SEQ ID NOs: 1 to 41, 133 to 211, and 345 to 388 and/or probes comprising sequences other than those that are complementary to one or more microRNAs of the human miRNome.
  • the hybridized target RNAs are then labeled with quantum dots via a biotin-streptavidin binding.
  • a confocal laser causes the quantum dots to fluoresce and the signal can be quantified.
  • small RNAs can be detected using a colorimetric assay.
  • small RNAs are labeled with streptavidin-conjugated gold followed by silver enhancement.
  • the gold nanoparticules bound to the hybridized target RNAs catalyze the reduction of silver ions to metallic silver, which can then be detected colorimetrically with a CCD camera
  • target RNAs in a sample of isolated total RNA are hybridized to two probes, one which is complementary to nucleic acids at the 5 '-end of the target RNA and the second which is complementary to the 3 '-end of the target RNA.
  • Each probe comprises, in some embodiments, one or more affinity-enhancing nucleotide analogs, such as LNA nucleotide analogs and each is labeled with a different fluorescent dye having different fluorescence emission spectra.
  • the sample is then flowed through a microfluidic capillary in which multiple lasers excite the fluorescent probes, such that a unique coincident burst of photons identifies a particular target RNA, and the number of particular unique coincident bursts of photons can be counted to quantify the amount of the target RNA in the sample.
  • a microfluidic capillary in which multiple lasers excite the fluorescent probes, such that a unique coincident burst of photons identifies a particular target RNA, and the number of particular unique coincident bursts of photons can be counted to quantify the amount of the target RNA in the sample.
  • a target RNA-specific probe can be labeled with 3 or more distinct labels selected from, e.g., fluorophores, electron spin labels, etc., and then hybridized to an RNA sample, such as total RNA, or a sample that is enriched in small RNAs.
  • Nonlimiting exemplary target RNA-specific probes include probes comprising sequences selected from of SEQ ID NOs: 1 to 41 and 133 to 211.
  • Nonlimiting exemplary target RNA-specific probes include probes comprising sequences that are complementary to sequences selected from of SEQ ID NOs: 1 to 41 and 133 to 211.
  • Nonlimiting exemplary target RNA-specific probes also include probes comprising at least 15 contiguous nucleotides of, or the complement of at least 15 contiguous nucleotides of, a sequence selected from SEQ ID NOs: 1 to 41, 133 to 211, and 345 to 388.
  • the sample RNA is modified before hybridization.
  • the target RNA/probe duplex is then passed through channels in a microfluidic device and that comprise detectors that record the unique signal of the 3 labels. In this way, individual molecules are detected by their unique signal and counted. See U.S. Patent Nos. 7,402,422 and 7,351,538 to Fuchs et al., U.S. Genomics, Inc., each of which is incorporated herein by reference in its entirety.
  • the detection and quantification of one or more target RNAs is accomplished by a solution-based assay, such as a modified Invader assay.
  • a solution-based assay such as a modified Invader assay. See Allawi H.T. et al. (2004) RNA 10:1153-1161, which is incorporated herein by reference in its entirety.
  • the modified invader assay can be performed on unfractionated detergent lysates of cervical cells.
  • the modified invader assay can be performed on total RNA isolated from cells or on a sample enriched in small RNAs. The target RNAs in a sample are annealed to two probes which form hairpin structures.
  • a first probe has a hairpin structure at the 5' end and a region at the 3 '-end that has a sequence that is complementary to the sequence of a region at the 5'-end of a target RNA.
  • the 3'-end of the first probe is the "invasive polynucleotide”.
  • a second probe has, from the 5' end to the 3 '-end a first "flap" region that is not complementary to the target RNA, a second region that has a sequence that is complementary to the 3 '-end of the target RNA, and a third region that forms a hairpin structure.
  • the two probes When the two probes are bound to a target RNA target, they create an overlapping configuration of the probes on the target RNA template, which is recognized by the Cleavase enzyme, which releases the flap of the second probe into solution.
  • the flap region then binds to a complementary region at the 3 '-end of a secondary reaction template ("SRT").
  • SRT secondary reaction template
  • a FRET polynucleotide (having a fluorescent dye bound to the 5 '-end and a quencher that quenches the dye bound closer to the 3 ' end) binds to a complementary region at the 5 '-end of the SRT, with the result that an overlapping configuration of the 3 '-end of the flap and the 5 '-end of the FRET polynucleotide is created.
  • Cleavase recognizes the overlapping configuration and cleaves the 5'-end of the FRET polynucleotide, generates a fluorescent signal when the dye is released into solution.
  • polynucleotides are provided.
  • synthetic polynucleotides are provided.
  • Synthetic polynucleotides refer to polynucleotides that have been synthesized in vitro either chemically or enzymatically.
  • Chemical synthesis of polynucleotides includes, but is not limited to, synthesis using polynucleotide synthesizers, such as OligoPilot (GE Healthcare), ABI 3900 DNA Synthesizer (Applied Biosystems), and the like.
  • Enzymatic synthesis includes, but is not limited, to producing polynucleotides by enzymatic amplification, e.g., PCR.
  • a polynucleotide comprises at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, or at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 1 to 41, 133 to 211, and 345 to 388, and sequences complementary to SEQ ID NOs: 1 to 41, 133 to 211, and 345 to 388.
  • the polynucleotide further comprises a region having a sequence that is not found in, or complementary to, any of SEQ ID NOs: 1 to 41, 133 to 211, and 345 to 388.
  • a polynucleotide comprises at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, or at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 1 to 7, 9 to 37, 133 to 201, and 345 to 388, and sequences complementary to SEQ ID NOs: 1 to 7, 9 to 37, 133 to 201, and 345 to 388.
  • the polynucleotide further comprises a region having a sequence that is not found in, or complementary to, any of SEQ ID NOs: 1 to 7, 9 to 37, 133 to 201, and 345 to 388.
  • a polynucleotide comprises fewer than
  • a polynucleotide is between 8 and 200, between 8 and 150, between 8 and 100, between 8 and 75, between 8 and 50, between 8 and 40, or between 8 and 30 nucleotides long.
  • the polynucleotide is a primer.
  • the primer is labeled with a detectable moiety.
  • a primer is not labeled.
  • a primer is a polynucleotide that is capable of specifically hybridizing to a target RNA or to a cDNA reverse transcribed from the target RNA or to an amplicon that has been amplified from a target RNA or a cDNA (collectively referred to as "template"), and, in the presence of the template, a polymerase and suitable buffers and reagents, can be extended to form a primer extension product.
  • the polynucleotide is a probe.
  • the probe is labeled with a detectable moiety.
  • a detectable moiety includes both directly detectable moieties, such as fluorescent dyes, and indirectly detectable moieties, such as members of binding pairs.
  • the probe can be detectable by incubating the probe with a detectable label bound to the second member of the binding pair.
  • a probe is not extendable, e.g., by a polymerase. In other embodiments, a probe is extendable.
  • the polynucleotide is a FRET probe that in some embodiments is labeled at the 5 '-end with a fluorescent dye (donor) and at the 3'- end with a quencher (acceptor), a chemical group that absorbs (i.e., suppresses) fluorescence emission from the dye when the groups are in close proximity (i.e., attached to the same probe).
  • the donor and acceptor are not at the ends of the FRET probe.
  • the emission spectrum of the donor moiety should overlap considerably with the absorption spectrum of the acceptor moiety.
  • RNA described herein employ one or more polynucleotides that have been modified, such as polynucleotides comprising one or more affinity-enhancing nucleotide analogs.
  • Modified polynucleotides useful in the methods described herein include primers for reverse transcription, PCR amplification primers, and probes.
  • the incorporation of affinity-enhancing nucleotides increases the binding affinity and specificity of a polynucleotide for its target nucleic acid as compared to polynucleotides that contain only deoxyribonucleotides, and allows for the use of shorter polynucleotides or for shorter regions of complementarity between the polynucleotide and the target nucleic acid.
  • affinity-enhancing nucleotide analogs include nucleotides comprising one or more base modifications, sugar modifications and/or backbone modifications.
  • modified bases for use in affinity-enhancing nucleotide analogs include 5-methylcytosine, isocytosine, pseudoisocytosine, 5- bromouracil, 5-propynyluracil, 6-aminopurine, 2-aminopurine, inosine, diaminopurine, 2- chloro-6-aminopurine, xanthine and hypoxanthine.
  • affinity-enhancing nucleotide analogs include nucleotides having modified sugars such as 2 '-substituted sugars, such as 2'-O- alkyl-ribose sugars, 2'-amino-deoxyribose sugars, 2'-fluoro- deoxyribose sugars, T- fluoro-arabinose sugars, and 2'-O-methoxyethyl-ribose (2'MOE) sugars.
  • modified sugars are arabinose sugars, or d-arabino-hexitol sugars.
  • affinity-enhancing nucleotide analogs include backbone modifications such as the use of peptide nucleic acids (PNA; e.g., an oligomer including nucleobases linked together by an amino acid backbone).
  • PNA peptide nucleic acids
  • backbone modifications include phosphorothioate linkages, phosphodiester modified nucleic acids, combinations of phosphodiester and phosphorothioate nucleic acid, methylphosphonate, alkylphosphonates, phosphate esters, alkylphosphonothioates, phosphoramidates, carbamates, carbonates, phosphate triesters, acetamidates, carboxymethyl esters, methylphosphorothioate, phosphorodithioate, p-ethoxy, and combinations thereof.
  • a polynucleotide includes at least one affinity-enhancing nucleotide analog that has a modified base, at least nucleotide (which may be the same nucleotide) that has a modified sugar, and/or at least one internucleotide linkage that is non-naturally occurring.
  • an affinity-enhancing nucleotide analog contains a locked nucleic acid ("LNA") sugar, which is a bicyclic sugar.
  • a polynucleotide for use in the methods described herein comprises one or more nucleotides having an LNA sugar.
  • a polynucleotide contains one or more regions consisting of nucleotides with LNA sugars.
  • a polynucleotide contains nucleotides with LNA sugars interspersed with deoxyribonucleotides. See, e.g., Frieden, M. et al. (2008) Curr. Pharm. Des. 14(11):1138- 1142.
  • a primer is provided.
  • a primer is identical or complementary to at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, or at least 24 contiguous nucleotides of a target RNA.
  • a primer may also comprise portions or regions that are not identical or complementary to the target RNA.
  • a region of a primer that is identical or complementary to a target RNA is contiguous, such that any region of a primer that is not identical or complementary to the target RNA does not disrupt the identical or complementary region.
  • a primer comprises a portion that is identically present in a target RNA.
  • a primer that comprises a region that is identically present in the target RNA is capable of selectively hybridizing to a cDNA that has been reverse transcribed from the RNA, or to an amplicon that has been produced by amplification of the target RNA or cDNA.
  • the primer is complementary to a sufficient portion of the cDNA or amplicon such that it selectively hybridizes to the cDNA or amplicon under the conditions of the particular assay being used.
  • “selectively hybridize” means that a polynucleotide, such as a primer or probe, will hybridize to a particular nucleic acid in a sample with at least 5 -fold greater affinity than it will hybridize to another nucleic acid present in the same sample that has a different nucleotide sequence in the hybridizing region. Exemplary hybridization conditions are discussed in Example 1. In some embodiments, a polynucleotide will hybridize to a particular nucleic acid in a sample with at least 10-fold greater affinity than it will hybridize to another nucleic acid present in the same sample that has a different nucleotide sequence in the hybridizing region.
  • Nonlimiting exemplary primers include primers comprising sequences that are identically present in, or complementary to a region of, sequences selected from SEQ ID NOs: 1 to 41, 133 to 211, and 345 to 388.
  • Exemplary primers also include, but are not limited to, primers comprising regions that are identical or complementary to at least 15 contiguous nucleotides of sequences selected from SEQ ID NOs: 1 to 41, 133 to 211, and 345 to 388.
  • a primer is used to reverse transcribe a target RNA, for example, as discussed herein.
  • a primer is used to amplify a target RNA or a cDNA reverse transcribed therefrom. Such amplification, in some embodiments, is quantitative PCR, for example, as discussed herein.
  • a primer comprises a detectable moiety.
  • methods of detecting the presence of a cervical dysplasia comprise hybridizing nucleic acids of a human cervical sample with a probe.
  • the probe comprises a portion that is complementary to a target RNA.
  • the probe comprises a portion that is identically present in the target RNA.
  • a probe that is complementary to a target RNA is complementary to a sufficient portion of the target RNA such that it selectively hybridizes to the target RNA under the conditions of the particular assay being used.
  • a probe that is complementary to a target RNA is complementary to at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, or at least 24 contiguous nucleotides of the target RNA.
  • a probe that is complementary to a target RNA comprises a region that is complementary to at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, or at least 24 contiguous nucleotides of the target RNA.
  • a probe that is complementary to a target RNA may also comprise portions or regions that are not complementary to the target RNA.
  • a region of a probe that is complementary to a target RNA is contiguous, such that any region of a probe that is not complementary to the target RNA does not disrupt the complementary region.
  • the probe comprises a portion that is identically present in the target RNA.
  • a probe that comprises a region that is identically present in the target RNA is capable of selectively hybridizing to a cDNA that has been reverse transcribed from the RNA, or to an amplicon that has been produced by amplification of the target RNA or cDNA.
  • the probe is complementary to a sufficient portion of the cDNA or amplicon such that it selectively hybridizes to the cDNA or amplicon under the conditions of the particular assay being used.
  • a probe that is complementary to a cDNA or amplicon is complementary to at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, or at least 24 contiguous nucleotides of the cDNA or amplicon.
  • a probe that is complementary to a target RNA comprises a region that is complementary to at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, or at least 24 contiguous nucleotides of the cDNA or amplicon.
  • a probe that is complementary to a cDNA or amplicon may also comprise portions or regions that a ⁇ e not complementary to the cDNA or amplicon.
  • a region of a probe that is complementary to a cDNA or amplicon is contiguous, such that any region of a probe that is not complementary to the cDNA or amplicon does not disrupt the complementary region.
  • Nonlimiting exemplary probes include probes comprising sequences set forth in SEQ ID NOs: 1 to 41 and 133 to 211.
  • Nonlimiting exemplary probes include probes comprising sequences that are identically present in, or complementary to a region of, sequences selected from SEQ ID NOs: 1 to 41, 133 to 211, and 345 to 388.
  • Exemplary probes also include, but are not limited to, probes comprising regions that are identical or complementary to at least 15 contiguous nucleotides of sequences selected from SEQ ID NOs: 1 to 41, 133 to 211, and 345 to 388.
  • the method of detectably quantifying one or more target RNAs comprises: (a) isolating total RNA; (b) reverse transcribing a target RNA to produce a cDNA that is complementary to the target RNA; (c) amplifying the cDNA from (b); and (d) detecting the amount of a target RNA using real time RT-PCR and a detection probe.
  • the real time RT-PCR detection is performed using a FRET probe, which includes, but is not limited to, a TaqMan® probe, a Molecular beacon probe and a Scorpion probe.
  • a FRET probe which includes, but is not limited to, a TaqMan® probe, a Molecular beacon probe and a Scorpion probe.
  • the real time RT-PCR detection and quantification is performed with a TaqMan® probe, i.e., a linear probe that typically has a fluorescent dye covalently bound at one end of the DNA and a quencher molecule covalently bound at the other end of the DNA.
  • the FRET probe comprises a sequence that is complementary to a region of the cDNA such that, when the FRET probe is hybridized to the cDNA, the dye fluorescence is quenched, and when the probe is digested during amplification of the cDNA, the dye is released from the probe and produces a fluorescence signal.
  • the amount of target RNA in the sample is proportional to the amount of fluorescence measured during cDNA amplification.
  • the TaqMan® probe typically comprises a region of contiguous nucleotides having a sequence that is complementary to a region of a target RNA or its complementary cDNA that is reverse transcribed from the target RNA template (i.e., the sequence of the probe region is complementary to or identically present in the target RNA to be detected) such that the probe is specifically hybridizable to the resulting PCR amplicon.
  • the probe comprises a region of at least 6 contiguous nucleotides having a sequence that is fully complementary to or identically present in a region of a cDNA that has been reverse transcribed from a target RNA template, such as comprising a region of at least 8 contiguous nucleotides, at least 10 contiguous nucleotides, at least 12 contiguous nucleotides, at least 14 contiguous nucleotides, or at least 16 contiguous nucleotides having a sequence that is complementary to or identically present in a region of a cDNA reverse transcribed from a target RNA to be detected.
  • the region of the cDNA that has a sequence that is complementary to the TaqMan® probe sequence is at or near the center of the cDNA molecule.
  • Molecular Beacons can be used to detect and quantitate PCR products. Like TaqMan® probes, Molecular Beacons use FRET to detect and quantitate a PCR product via a probe having a fluorescent dye and a quencher attached at the ends of the probe. Unlike TaqMan® probes, Molecular Beacons remain intact during the PCR cycles. Molecular Beacon probes form a stem-loop structure when free in solution, thereby allowing the dye and quencher to be in close enough proximity to cause fluorescence quenching. When the Molecular Beacon hybridizes to a target, the stem-loop structure is abolished so that the dye and the quencher become separated in space and the dye fluoresces. Molecular Beacons are available, e.g., from Gene LinkTM (see http://www.genelink.com/newsite/products/mbintro.asp).
  • Scorpion probes can be used as both sequence-specific primers and for PCR product detection and quantitation. Like Molecular Beacons, Scorpion probes form a stem-loop structure when not hybridized to a target nucleic acid. However, unlike Molecular Beacons, a Scorpion probe achieves both sequence-specific priming and PCR product detection. A fluorescent dye molecule is attached to the 5 '-end of the Scorpion probe, and a quencher is attached to the 3 '-end. The 3' portion of the probe is complementary to the extension product of the PCR primer, and this complementary portion is linked to the 5 '-end of the probe by a non-amplifiable moiety.
  • Scorpion probes are available from, e.g, Premier Biosoft International (see http://www.premierbiosoft.com/tech_notes/Scorpion.html).
  • labels that can be used on the FRET probes include colorimetric and fluorescent labels such as 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 Alexa Fluor dyes, BODIPY dyes, such
  • 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
  • fluorescently labeled ribonucleotides useful in the preparation of RT-PCR probes for use in some embodiments of the methods described herein are available from Molecular Probes (Invitrogen), 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 (GE Healthcare), such as Cy3-UTP and Cy5-UTP.
  • Examples of fluorescently labeled deoxyribonucleotides useful in the preparation of RT-PCR probes for use in the methods described herein include Dinitrophenyl (DNP)- l'-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
  • dyes and other moieties are introduced into polynucleotide used in the methods described herein, such as FRET probes, via modified nucleotides.
  • a "modified nucleotide” refers to a nucleotide that has been chemically modified, but still functions as a nucleotide.
  • the modified nucleotide has a chemical moiety, such as a dye or quencher, covalently attached, and can be introduced into a polynucleotide, for example, by way of solid phase synthesis of the polynucleotide.
  • the modified nucleotide includes one or more reactive groups that can react with a dye or quencher before, during, or after incorporation of the modified nucleotide into the nucleic acid.
  • the modified nucleotide is an amine-modified nucleotide, i.e., a nucleotide that has been modified to have a reactive amine group.
  • the modified nucleotide comprises a modified base moiety, such as uridine, adenosine, guanosine, and/or cytosine.
  • the amine-modified nucleotide is selected from 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.
  • nucleotides with different nucleobase moieties are similarly modified, for example, 5-(3-aminoallyl)-GTP instead of 5-(3- aminoallyl)-UTP.
  • Many amine modified nucleotides are commercially available from, e.g., Applied Biosystems, Sigma, Jena Bioscience and TriLink.
  • Exemplary detectable moieties also include, but are not limited to, members of binding pairs. In some such embodiments, a first member of a binding pair is linked to a polynucleotide. The second member of the binding pair is linked to a detectable label, such as a fluorescent label.
  • binding pairs include, but are not limited to, biotin and streptavidin, antibodies and antigens, etc.
  • each probe that is targeted to a unique cDNA is spectrally distinguishable when released from the probe.
  • each target RNA is detected by a unique fluorescence signal.
  • One skilled in the art can select a suitable detection method for a selected assay, e.g., a real-time RT-PCR assay.
  • the selected detection method need not be a method described above, and may be any method.
  • compositions are provided.
  • compositions are provided for use in the methods described herein.
  • a composition comprises at least one polynucleotide. In some embodiments, a composition comprises at least one primer. In some embodiments, a composition comprises at least one probe. In some embodiments, a composition comprises at least one primer and at least one probe.
  • compositions that comprise at least one target RNA-specific primer.
  • target RNA-specific primer encompasses primers that have a region of contiguous nucleotides having a sequence that is (i) identically present in one of SEQ ID NOs: 1 to 41 or 133 to 211, (ii) complementary to the sequence of a region of contiguous nucleotides found in one of SEQ ID NOs: 1 to 41 or 133 to 211 ; (iii) complementary to the sequence of a region of contiguous nucleotides found in one of SEQ ID NOs: 345 to 388; or (iv) identically present in one of SEQ ID NOs: 345 to 388.
  • compositions that comprise at least one target RNA-specific probe.
  • target RNA-specific probe encompasses probes that have a region of contiguous nucleotides having a sequence that is (i) identically present in one of SEQ ID NOs: 1 to 41 or 133 to 211, (ii) complementary to the sequence of a region of contiguous nucleotides found in one of SEQ ID NOs: 1 to 41 or 133 to 211; (iii) complementary to the sequence of a region of contiguous nucleotides found in one of SEQ ID NOs: 345 to 388; or (iv) identically present in one of SEQ ID NOs: 345 to 388.
  • target RNA-specific primers and probes comprise deoxyribonucleotides.
  • target RNA-specific primers and probes comprise at least one nucleotide analog.
  • Nonlimiting exemplary nucleotide analogs include, but are not limited to, analogs described herein, including LNA analogs and peptide nucleic acid (PNA) analogs.
  • target RNA-specific primers and probes comprise at least one nucleotide analog which increases the hybridization binding energy (e.g., an affinity-enhancing nucleotide analog, discussed above).
  • a target RNA-specific primer or probe in the compositions described herein binds to one target RNA in the sample.
  • a single primer or probe binds to multiple target RNAs, such as multiple isomirs.
  • more than one primer or probe specific for a single target RNA is present in the compositions, the primers or probes capable of binding to overlapping or spatially separated regions of the target RNA.
  • the composition comprises at least one target RNA-specific primer or probe (or region thereof) having a sequence that is identically present in a target RNA (or region thereof).
  • a target RNA is capable of specifically hybridizing to at least one probe comprising a sequence selected from SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7 and 8. In some embodiments, a target RNA is capable of specifically hybridizing to at least one nucleic acid probe comprising a sequence selected from SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 and 12. In some embodiments, a target RNA is capable of specifically hybridizing to at least one nucleic acid probe comprising a sequence selected from SEQ ID NOs: 142, 151, 153, 193, 194, 205, 172, 208, 210, and 211.
  • a target RNA is capable of specifically hybridizing to at least one nucleic acid probe comprising a sequence selected from SEQ ID NOs: 1, 5, 7, and 32. In some embodiments, a target RNA is capable of specifically hybridizing to at least one probe comprising a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211. In some embodiments, a target RNA comprises at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388. In some embodiments, a target RNA comprises a sequence that is complementary to at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211. In some embodiments, a target RNA, in its mature form, comprises fewer than 30 nucleotides. In some embodiments, a target RNA is a microRNA.
  • the composition comprises a plurality of target RNA-specific primers and/or probes for each of at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, or at least 8 target RNAs, the target RNAs comprising a region of contiguous nucleotides having a sequence that is identically present in one of SEQ ID NOs: 42, 43, 44, 45, 46, 47, 48, or 49.
  • the plurality includes a target RNA-specific primer and/or probe specific for each of at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, or at least 12 target RNAs, the target RNAs comprising a region of contiguous nucleotides having a sequence that is identically present in one of SEQ ID NOs: 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, and 53.
  • the plurality includes a target RNA-specific primer and/or probe specific for each of at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 40, at least 50, at least 75, or at least 100 target RNAs comprising a region of contiguous nucleotides having a sequence that is identically present in one of SEQ ID NOs: 42 to 82 or 226 to 317.
  • target RNAs described herein comprise a sequence identically present in a sequence set forth in Table 2 or Table 12, except that thymine (T) bases in the sequences shown in Table 2 or Table 12 are replaced by uracil (U) bases in the target RNAs.
  • a composition is an aqueous composition.
  • the aqueous composition comprises a buffering component, such as phosphate, tris, HEPES, etc., and/or additional components, as discussed below.
  • a composition is dry, for example, lyophilized, and suitable for reconstitution by addition of fluid.
  • a dry composition may include a buffering component and/or additional components.
  • a composition comprises one or more additional components.
  • Additional components include, but are not limited to, salts, such as NaCl , KCl,and MgCl 2 ; polymerases, including thermostable polymerases; dNTPs; RNase inhibitors; bovine serum albumin (BSA) and the like; reducing agents, such as ⁇ - mercaptoethanol; EDTA and the like; etc.
  • salts such as NaCl , KCl,and MgCl 2
  • polymerases including thermostable polymerases
  • dNTPs including RNase inhibitors
  • BSA bovine serum albumin
  • reducing agents such as ⁇ - mercaptoethanol
  • EDTA and the like
  • an addressable microarray component comprises target RNA-specific probes attached to a substrate.
  • Microarrays for use in the methods described herein comprise a solid substrate onto which the probes are covalently or non-covalently attached.
  • probes capable of hybridizing to one or more target RNAs or cDNAs are attached to the substrate at a defined location ("addressable array").
  • Probes can be attached to the substrate in a wide variety of ways, as will be appreciated by those in the art.
  • the probes are synthesized first and subsequently attached to the substrate.
  • the probes are synthesized on the substrate.
  • probes are synthesized on the substrate surface using techniques such as photopolymerization and photolithography.
  • the solid substrate is a material that is modified to contain discrete individual sites appropriate for the attachment or association of the probes and is amenable to at least one detection method.
  • substrates include glass and modified or functionalized glass, plastics (including acrylics, polystyrene and copolymers of styrene and other materials, polypropylene, polyethylene, polybutylene, polyurethanes, TeflonJ, etc.), polysaccharides, nylon or nitrocellulose, resins, silica or silica-based materials including silicon and modified silicon, carbon, metals, inorganic glasses and plastics.
  • the substrates allow optical detection without appreciably fluorescing.
  • the substrate is planar.
  • probes are placed on the inside surface of a tube, such as for flow-through sample analysis to minimize sample volume.
  • probes can be in the wells of multi-well plates.
  • probes can be attached to an addressable microbead array.
  • the probes can be attached to a flexible substrate, such as a flexible foam, including closed cell foams made of particular plastics.
  • the substrate and the probe can each be derivatized with functional groups for subsequent attachment of the two.
  • the substrate is derivatized with one or more chemical functional groups including, but not limited to, amino groups, carboxyl groups, oxo groups and thiol groups.
  • probes are attached directly to the substrate through one or more functional groups.
  • probes are attached to the substrate indirectly through a linker (i.e., a region of contiguous nucleotides that space the probe regions involved in hybridization and detection away from the substrate surface).
  • probes are attached to the solid support through the 5' terminus. In other embodiments, probes are attached through the 3' terminus.
  • probes are attached to the substrate through an internal nucleotide.
  • the probe is attached to the solid support non-covalently, e.g., via a biotin-streptavidin interaction, wherein the probe biotinylated and the substrate surface is covalently coated with streptavidin.
  • the compositions comprise a microarray having probes attached to a substrate, wherein at least one of the probes (or a region thereof) comprises a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1 to 41, 133 to 211, or SEQ ID NOs: 345 to 388.
  • at least 2, at least 5, at least 10, at least 15, at least 20, at least 25, at least 30, at least 40, at least 50, or at least 100 of the probes comprise a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1 to 41, 133 to 211, or 345 to 388.
  • the microarray comprises at least one target RNA-specific probe comprising a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1 to 41, 133 to 211, or 345 to 388 and at least one target RNA-specific probe comprising a sequence that is identically present in, or complementary to a region of, a target RNA set forth in Table 3.
  • the microarray comprises each target RNA-specific probe at only one location on the microarray.
  • the microarray comprises at least one target RNA-specific probe at multiple locations on the microarray.
  • the terms “complementary” or “partially complementary” to a target RNA (or target region thereof), and the percentage of “complementarity” of the probe sequence to that of the target RNA sequence is the percentage “identity” to the reverse complement of the sequence of the target RNA.
  • the degree of “complementarity” is expressed as the percentage identity between the sequence of the probe (or region thereof) and the reverse complement of the sequence of the target RNA that best aligns therewith. The percentage is calculated by counting the number of aligned bases that are identical as between the 2 sequences, dividing by the total number of contiguous nucleotides in the probe, and multiplying by 100.
  • the microarray comprises at least one probe having a region with a sequence that is fully complementary to a target region of a target RNA. In other embodiments, the microarray comprises at least one probe having a region with a sequence that comprises one or more base mismatches when compared to the sequence of the best-aligned target region of a target RNA.
  • the microarray comprises at least one probe having a region of at least 10, at least 11, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, or at least 25 contiguous nucleotides with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1 to 41 or 133 to 211.
  • the microarray comprises at least one probe having a region of at least 10, at least 11, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, or at least 25 contiguous nucleotides with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1 to 41 or 133 to 211, and at least one probe comprising a region of at least 10, at least 11, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, or at least 25 contiguous nucleotides having a sequence that is identically present in, or complementary to a region of, a target RNA set forth in Table 3.
  • the microarray comprises at least one probe having a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, or 8.
  • the microarray comprises at least one, at least two, at least three, at least five, or eight probes that each comprise a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7 or 8.
  • the microarray further comprises additional probes that do not have a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1 , 2, 3, 4, 5, 6, 7 and 8.
  • the microarray comprises at least one probe having a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12.
  • the microarray comprises at least one, at least two, at least three, at least five, at least eight, at least 10, or at least 12 probes that each comprise a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12.
  • the microarray further comprises additional probes that do not have a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12.
  • the microarray comprises at least one probe having a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 142, 151, 153, 193, 194, 205, 172, 208, 210, and 211.
  • the microarray comprises at least one, at least two, at least three, at least five, at least eight, or at least 10 probes that each comprise a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 142, 151, 153, 193, 194, 205, 172, 208, 210, and 211.
  • the microarray further comprises additional probes that do not have a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 142, 151, 153, 193, 194, 205, 172, 208, 210, and 211.
  • the microarray comprises at least one probe having a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1, 5, 7, or 32. In some embodiments, the microarray comprises at least one, at least two, at least three, or at least four probes that each comprise a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1, 5, 7, or 32. In some embodiments, the microarray further comprises additional probes that do not have a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1, 5, 7, or 32.
  • the microarrays comprise probes having a region with a sequence that is complementary to target RNAs that comprise a substantial portion of the human miRNome (i.e., the publicly known microRNAs that have been accessioned by others into miRBase (http://microrna.sanger.ac.uk/ at the time the microarray is fabricated), such as at least about 60%, at least about 70%, at least about 80%, at least about 90%, even at least about 95% of the human miRNome.
  • the human miRNome i.e., the publicly known microRNAs that have been accessioned by others into miRBase (http://microrna.sanger.ac.uk/ at the time the microarray is fabricated
  • the microarrays comprise probes that have a region with a sequence that is identically present in target RNAs that comprise a substantial portion of the human miRNome, such as at least about 60%, at least about 70%, at least about 80%, at least about 90%, even at least about 95% of the human miRNome.
  • components are provided that comprise probes attached to microbeads, such as those sold by Luminex, each of which is internally dyed with red and infrared fluorophores at different intensities to create a unique signal for each bead.
  • the compositions useful for carrying out the methods described herein include a plurality of microbeads, each with a unique spectral signature. Each uniquely labeled microbead is attached to a unique target RNA-specific probe such that the unique spectral signature from the dyes in the bead is associated with a particular probe sequence.
  • Nonlimiting exemplary probe sequences include SEQ ID NOs: 1 to 41 and 133 to 211.
  • Nonlimiting exemplary probe sequences also include probes comprising a region that is identically present in, or complementary to, a sequence selected from SEQ ID NOs: 1 to 41, 133 to 211, and SEQ ID NOs: 345 to 388.
  • a probe sequence comprises at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, or at least 24 contiguous nucleotides that are identically present in, or complementary to a region of, SEQ ID NOs: 1 to 41, 133 to 211, and SEQ ID NOs: 345 to 388.
  • a uniquely labeled microbead has attached thereto a probe having a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1 to 41, 133 to 211, or 345 to 388.
  • the uniquely labeled microbead has attached thereto a probe having a region with a sequence that comprises one or more base mismatches when compared to the most similar sequence selected from SEQ ID NOs: 1 to 41, 133 to 211, and 345 to 388, and sequences complementary to SEQ ID NOs: 1 to 41, 133 to 211.
  • a composition comprises a plurality of uniquely labeled microbeads, wherein at least one microbead has attached thereto a probe having a region of at least 10, at least 11, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, or at least 25 contiguous nucleotides with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1 to 41 , 133 to 211 or 345 to 388.
  • a composition comprises a plurality of uniquely labeled microbeads, wherein at least one of the microbeads has attached thereto a probe having a region of at least 10, at least 11, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, or at least 25 contiguous nucleotides with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1 to 41, 133 to 211, or 345 to 388, and at least a second microbead having attached thereto a probe comprising a region of at least 10, at least 11, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, or at least 25 contiguous nucleotides having a sequence that is identically present in, or complementary to a region of, a target RNA set forth in Table 3.
  • the compositions comprise a plurality of uniquely labeled microbeads, at least one of which has attached thereto a target RNA- specific probe having a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, or 8.
  • the compositions comprise at least two, at least three, at least five, or at least 8 uniquely labeled microbeads that each have attached thereto a unique target RNA- specific probe having a region with a sequence that is identically present in, or complementary to a region of, a different one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7 or 8.
  • the composition comprises at least one uniquely labeled microbead having attached thereto a target RNA-specific probe having a region with a sequence that is not present in, or complementary to a region of, any of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, or 8.
  • the compositions comprise plurality of uniquely labeled microbeads, wherein at least one microbead has attached thereto a probe having a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12.
  • the compositions comprise at least two, at least three, at least five, at least eight, at least 10, or at least 12 uniquely labeled microbeads that each have attached thereto a unique target RNA-specific probe having a region with a sequence that is identically present in, or complementary to a region of, a different one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12.
  • the composition comprises at least one uniquely labeled microbead having attached thereto a target RNA-specific probe having a region with a sequence that is not present in, or complementary to a region of, any of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12.
  • compositions comprise plurality of uniquely labeled microbeads, wherein at least one microbead has attached thereto a probe having a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 142, 151, 153, 193, 194, 205, 172, 208, 210, and 211.
  • compositions comprise at least two, at least three, at least five, at least eight, or at least 10 uniquely labeled microbeads that each have attached thereto a unique target RNA-specific probe having a region with a sequence that is identically present in, or complementary to a region of, a different one of SEQ ID NOs: 142, 151, 153, 193, 194, 205, 172, 208, 210, and 211.
  • the composition comprises at least one uniquely labeled microbead having attached thereto a target RNA- specific probe having a region with a sequence that is not present in, or complementary to a region of, any of SEQ ID NOs: 142, 151, 153, 193, 194, 205, 172, 208, 210, and 211.
  • the compositions comprise plurality of uniquely labeled microbeads, wherein at least one microbead has attached thereto a probe having a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1, 5, 7, or 32.
  • the compositions comprise at least two, at least three, or at least four uniquely labeled microbeads that each have attached thereto a unique target RNA-specific probe having a region with a sequence that is identically present in, or complementary to a region of, a different one of SEQ ID NOs: 1, 5, 7, or 32.
  • the composition comprises at least one uniquely labeled microbead having attached thereto a target RNA-specific probe having a region with a sequence that is not present in, or complementary to a region of, any of SEQ ID NOs: 1 , 5, 7, or 32.
  • compositions comprise a plurality of uniquely labeled microbeads, wherein the plurality comprises at least one microbead having attached thereto a probe having a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1 to 41, 133 to 211, or SEQ ID NOs: 345 to 388.
  • the plurality comprises at least two, at least five, at least 10, at least 15, at least 20, at least 25, at least 30, at least 40, at least 50, at least 60, at least 75, or at least 100 microbeads each of which having attached thereto a probe having a region with a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: I to 41, 133 to 211, or 345 to 388.
  • a composition comprises at least one uniquely labeled microbead having attached thereto a target RNA-specific probe having a region with a sequence that is not present in, or complementary to a region of, any of SEQ ID NOs: 1 to 41 or 133 to 211.
  • the compositions comprise a plurality of uniquely labeled microbeads, at least one of which has attached thereto a probe having a region with a sequence that identically present in, or complementary to a region of, one of SEQ ID NOs: 1 to 41, 133 to 211, or 345 to 388, and at least a second bead that has attached thereto a probe having a region with a sequence that is identically present in, or complementary to a region of, a target RNA set forth in Table 3.
  • compositions comprise a plurality of uniquely labeled microbeads, each of which has attached thereto a unique probe having a region that is complementary to target RNAs that comprise a substantial portion of the human miRNome, such as at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 95% of the human miRNome.
  • compositions comprise a plurality of uniquely labeled microbeads having attached thereto a unique probe having a region with a sequence that is identically present in target RNAs that comprise a substantial portion of the human miRNome, such as at least about 60%, at least about 70%, at least about 80%, at least about 90%, or at least about 95% of the human miRNome.
  • compositions are provided that comprise at least one polynucleotide for detecting at least one target RNA.
  • the polynucleotide is used as a primer for a reverse transcriptase reaction.
  • the polynucleotide is used as a primer for amplification.
  • the polynucleotide is used as a primer for RT-PCR.
  • the polynucleotide is used as a probe for detecting at least one target RNA.
  • the polynucleotide is detectably labeled.
  • the polynucleotide is a FRET probe.
  • the polynucleotide is a TaqMan® probe, a Molecular Beacon, or a Scorpion probe.
  • a composition comprises at least one FRET probe having a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1 to 41, 133 to 211, or 345 to 388.
  • a composition comprises at least two, at least five, at least 10, at least 15, at least 20, at least 25, at least 30, at least 40, at least 50, at least 60, at least 75, or at least 100 FRET probes, each of which has a sequence that is identically present in, or complementary to a region of, a different one of SEQ ID NOs: 1 to 41, 133 to 211, or 345 to 388.
  • a FRET probe is labeled with a donor/acceptor pair such that when the probe is digested during the PCR reaction, it produces a unique fluorescence emission that is associated with a specific target RNA.
  • each probe is labeled with a different donor/acceptor pair such that when the probe is digested during the PCR reaction, each one produces a unique fluorescence emission that is associated with a specific probe sequence and/or target RNA.
  • the sequence of the FRET probe is complementary to a target region of a target RNA.
  • the FRET probe has a sequence that comprises one or more base mismatches when compared to the sequence of the best- aligned target region of a target RNA.
  • a composition comprises a FRET probe consisting of at least 8, at least 9, at least 10, at least 11, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, or at least 25 nucleotides, wherein at least a portion of the sequence is identically present in, or complementary to a region of, one of SEQ ID NOs: 1 to 41, 133 to 211, and 345 to 388.
  • At least 8, at least 9, at least 10, at least 11, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, or at least 25 nucleotides of the FRET probe are identically present in, or complementary to a region of, one of SEQ ID NOs: 1 to 41, 133 to 211, and 345 to 388.
  • the FRET probe has a sequence with one, two or three base mismatches when compared to the sequence or complement of one of SEQ ID NOs: 1 to 41 or 133 to 211.
  • compositions further comprise a FRET probe consisting of at least 10, at least 11 , at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, or at least 25 contiguous nucleotides, wherein the FRET probe comprises a sequence that is identically present in, or complementary to a region of, a region of a target RNA set forth in Table 3.
  • the FRET probe is identically present in, or complementary to a region of, at least at least 10, at least 11, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, or at least 24 contiguous nucleotides of a target RNA set forth in Table 3.
  • the compositions comprise at least one target RNA-specific FRET probe comprising a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, or 8.
  • the compositions comprise at least two, at least three, at least five, or at least 8 uniquely labeled target RNA-specific FRET probes, each comprising a sequence that is identically present in, or complementary to a region of, a different one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7 and 8.
  • the compositions comprise at least one target RNA-specific FRET probe comprising a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12.
  • the compositions comprise at least two, at least three, at least five, at least eight, at least 10, or at least 12 uniquely labeled target RNA-specific FRET probes, each of which comprises a sequence that is identically present in, or complementary to a region of, a different one of SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 and 12.
  • the compositions comprise at least one target RNA-specific FRET probe comprising a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 142, 151, 153, 193, 194, 205, 172, 208, 210, and 211.
  • the compositions comprise at least two, at least three, at least five, at least eight, at least nine, or at least 10 uniquely labeled target RNA- specific FRET probes, each of which comprises a sequence that is identically present in, or complementary to a region of, a different one of SEQ ID NOs: 142, 151, 153, 193, 194, 205, 172, 208, 210, and 211.
  • the compositions comprise at least one target RNA-specific FRET probe comprising a sequence that is identically present in, or complementary to a region of, one of SEQ ID NOs: 1 , 5, 7, or 32.
  • the compositions comprise at least two, at least three, or at least four uniquely labeled target RNA-specific FRET probes, each of which comprises a sequence that is identically present in, or complementary to a region of, a different one of SEQ ID NOs: 1, 5, 7, or 32.
  • kits comprises a polynucleotide discussed above. In some embodiments, a kit comprises at least one primer and/or probe discussed above. In some embodiments, a kit comprises at least one polymerase, such as a thermostable polymerase. In some embodiments, a kit comprises dNTPs. In some embodiments, kits for use in the real time RT-PCR methods described herein comprise one or more target RNA-specific FRET probes and/or one or more primers for reverse transcription of target RNAs and/or one or more primers for amplification of target RNAs or cDNAs reverse transcribed therefrom.
  • one or more of the primers and/or probes is
  • linear primer refers to a polynucleotide that is a single stranded molecule, and typically does not comprise a short region of, for example, at least 3, 4 or 5 contiguous nucleotides, which are complementary to another region within the same polynucleotide such that the primer forms an internal duplex.
  • the primers for use in reverse transcription comprise a region of at least 4, such as at least 5, such as at least 6, such as at least 7 or more contiguous nucleotides at the 3 '-end that has a sequence that is complementary to region of at least 4, such as at least 5, such as at least 6, such as at least 7 or more contiguous nucleotides at the 5 '-end of a target RNA.
  • a kit comprises one or more pairs of linear primers (a "forward primer” and a “reverse primer”) for amplification of a cDNA reverse transcribed from a target RNA.
  • a first primer comprises a region of at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 contiguous nucleotides having a sequence that is identical to the sequence of a region of at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 contiguous nucleotides at the 5 '-end of a target RNA.
  • a second primer comprises a region of at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 contiguous nucleotides having a sequence that is complementary to the sequence of a region of at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 contiguous nucleotides at the 3 '-end of a target RNA.
  • the kit comprises at least a first set of primers for amplification of a cDNA that is reverse transcribed from a target RNA capable of specifically hybridizing to a nucleic acid comprising a sequence identically present in one of SEQ ID NOs: 1 to 41 and 133 to 211 and/or a cDNA that is reverse transcribed from a target RNA that comprises at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388.
  • the kit further comprises at least a second set of primers for amplification of a cDNA that is reverse transcribed from a target RNA set forth in Table 3.
  • the kit comprises at least two, at least five, at least 10, at least 15, at least 20, at least 25, at least 30, at least 40, at least 50, at least 60, at least 75, or at least 100 sets of primers, each of which is for amplification of a cDNA that is reverse transcribed from a different target RNA capable of specifically hybridizing to a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211 and/or a cDNA that is reverse transcribed from a target RNA that comprises at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388.
  • the kit comprises at least one set of primers that is capable of amplifying more than one cDNA reverse transcribed from a target RNA in a sample.
  • probes and/or primers for use in the compositions described herein comprise deoxyribonucleotides.
  • probes and/or primers for use in the compositions described herein comprise deoxyribonucleotides and one or more nucleotide analogs, such as LNA analogs or other duplex-stabilizing nucleotide analogs described above.
  • probes and/or primers for use in the compositions described herein comprise all nucleotide analogs.
  • the probes and/or primers comprise one or more duplex- stabilizing nucleotide analogs, such as LNA analogs, in the region of complementarity.
  • compositions described herein also comprise probes, and in the case of RT-PCR, primers, that are specific to one or more housekeeping genes for use in normalizing the quantities of target RNAs.
  • probes include those that are specific for one or more products of housekeeping genes selected from U6 snRNA, ACTB, B2M, GAPDH, GUSB, HPRTl, PPIA, RPLP, RRNl 8S, TBP, TUBB, UBC, YWHA (TATAA), PGKl, and RPL4.
  • the kits for use in real time RT-PCR methods described herein further comprise reagents for use in the reverse transcription and amplification reactions.
  • kits comprise enzymes such as reverse transcriptase, and a heat stable DNA polymerase, such as Taq polymerase.
  • the kits further comprise deoxyribonucleotide triphosphates (dNTP) for use in reverse transcription and amplification.
  • the kits comprise buffers optimized for specific hybridization of the probes and primers.
  • quantitation of target RNA expression levels requires assumptions to be made about the total RNA per cell and the extent of sample loss during sample preparation. In order to correct for differences between different samples or between samples that are prepared under different conditions, the quantities of target RNAs in some embodiments are normalized to the expression of at least one endogenous housekeeping gene.
  • Appropriate genes for use as reference genes in the methods described herein include those as to which the quantity of the product does not vary between normal and dysplastic or cancerous cervical cells, or between different cell lines or under different growth and sample preparation conditions.
  • endogenous housekeeping genes useful as normalization controls in the methods described herein include, but are not limited to, U6 snRNA, RNU44, RNU 48, and U47.
  • the at least one endogenous housekeeping gene for use in normalizing the measured quantity of microRNAs is selected from U6 snRNA, U6 snRNA, RNU44, RNU 48, and U47.
  • one housekeeping gene is used for normalization.
  • more than one housekeeping gene is used for normalization.
  • methods comprise detecting a qualitative change in a target RNA profile generated from a clinical sample of human cervical cells as compared to a normal target RNA profile (in some exemplary embodiments, a target RNA profile of a control sample). Some qualitative changes in the expression profile are indicative of the presence of cervical dysplasia in a sample of human cervical cells. Various qualitative changes in the expression profile are indicative of the propensity to proceed to cervical cancer.
  • target RNA profile refers to a set of data regarding the concurrent expression of a plurality of target RNAs in the same sample.
  • At least one, at least two, at least three, at least four, at least five, at least six, at least seven, or at least eight of the target RNAs of the plurality of target RNAs are capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7 and 8.
  • At least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least 10, at least 11, or at least 12 of the target RNAs of the plurality of target RNAs is capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 and 12.
  • At least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, or at least 10 of the target RNAs of the plurality of target RNAs is capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 142, 151, 153, 193, 194, 205, 172, 208, 210, and 211.
  • at least one, at least two, at least three, or at least four of the target RNAs of the plurality of target RNAs is capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1, 5, 7, and 32.
  • At least one, at least two, at least five, at least 10, at least 15, at least 20, at least 25, at least 30, at least 40, at least 50, at least 60, at least 75, or at least 100 of the plurality of target RNAs is capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211.
  • at least one, at least two, at least five, at least 10, at least 15, at least 20, at least 25, at least 30, at least 40 of the plurality of target RNAs comprises at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388.
  • At least one, at least two, at least five, at least 10, at least 15, at least 20, at least 25, at least 30, at least 40, at least 50, at least 60, at least 75, or at least 100 of the plurality of target RNAs comprises a sequence that is complementary to at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211.
  • a target RNA in its mature form, comprises fewer than 30 nucleotides.
  • a target RNA is a microRNA.
  • concurrent expression data are obtained using, e.g., a microarray, as described above.
  • a microarray comprising probes having sequences that are complementary to a substantial portion of the miRNome may be employed to carry out target RNA gene expression profiling, for analysis of target RNA expression patterns.
  • distinct target RNA signatures are associated with established markers for cervical dysplasia, or directly with the presence of cervical dysplasia. In some embodiments, distinct target RNA signatures are associated with established markers for CIN-I, CIN-2 or CIN-3 cervical dysplasia, or directly with the level of severity of cervical dysplasia. In some embodiments, distinct target RNA signatures are associated with established markers for cervical dysplasia likely to progress to carcinoma, or directly with cervical dysplasia that is likely to progress to carcinoma. In some embodiments, distinct target RNA signatures are associated with HPV infection and/or integration into the genome of the host cell. In some embodiments, distinct target RNA signatures are associated with established markers for cervical cancer, or directly with the presence of cervical cancer.
  • total RNA from a sample from a subject suspected of having cervical dysplasia is quantitatively reverse transcribed to provide a set of labeled polynucleotides complementary to the RNA in the sample.
  • the polynucleotides are then hybridized to a microarray comprising target RNA-speciflc probes to provide a hybridization profile for the sample.
  • the result is a hybridization profile for the sample representing the expression pattern of target RNAs in the sample.
  • the hybridization profile comprises the signal from the binding of the polynucleotides reverse transcribed from the sample to the target RNA- specific probes in the microarray.
  • the profile is recorded as the presence or absence of binding (signal vs. zero signal). In some embodiments, the profile recorded includes the intensity of the signal from each hybridization. The profile is compared to the hybridization profile generated from a normal, i.e., noncancerous, or in some embodiments, a control sample. An alteration in the signal is indicative of the presence of cervical dysplasia or cervical cancer in the subject.
  • methods herein in combination with detecting one or more target RNAs that are capable of specifically hybridizing to a nucleic acid comprising a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211 and/or detecting one or more target RNAs comprising at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 345 to 388 and/or detecting one or more target RNAs that comprise a sequence that is complementary to at least 15 contiguous nucleotides of a sequence selected from SEQ ID NOs: 1 to 41 and 133 to 211, methods herein further comprise detecting the level(s) of expression of at least one other marker associated with cervical dysplasia or HPV integration.
  • the methods described herein further comprise detecting increased expression of any one or more of miR-21, miR-31, miR-182, miR-183, miR-146a, miR-155, and miR-205.
  • the methods described herein further comprise detecting increased expression of any one or more of miR-663, miR-765, miR-92b*, miR-936, miR-9, miR-199a*, miR-199a, miR- 199b, miR-145, miR-133a, miR-133b, miR-214, miR-127, miR-210, miR-301, miR-142- 3p, miR-142-5p, miR-194, miR-215 and miR-32.
  • the methods described herein further comprise detecting altered expression of target RNAs associated with HPV integration sites.
  • the term "associated with" a given HPV integration site means that the target RNA gene is located in close proximity to the HPV integration site; i.e., when the target RNA is located within the same chromosomal band or within 3 megabases (3 Mb), preferably within 2.5 Mb, of the HPV integration site.
  • the methods further comprise detecting increased expression of target RNAs associated with HPV integration sites, such as fragile sites which are preferential targets for HPVl 6 associated with cervical tumors.
  • Such target RNAs include: miR-186, miR-101 (associated with FRAlA on chromosome Ip36 and FRAlC on chromosome Ip31); miR- 194 and miR-215 (associated with FRAlF on chromosome Iq21 and FRAlH on chromosome Iq42.1); miR-106b, miR-25 and miR93 (associated with FRA7F on chromosome 7q22); miR-29b, miR-29a, miR-96, miR-182-5p, miR-182-3p, miR-183, and miR- 129-1 (associated with FRA7G on chromosome 7q31.2 and FRA7H on chromosome 7q32.3); Iet7-la, Iet7-d, let-7f-l, miR-23b, miR-24-1, and miR-27b (associated with FRA9D on chromosome 9q22.1); miR-32 (associated with FRA9E on
  • the methods described herein further comprise detecting altered expression of cervical cancer-associated small RNAs with non-canonical hairpins.
  • the methods described herein further comprise detecting increased expression of the mRNA of one or more of the following genes: BIRC5, IGF2BP3, TERC, CDKN2A, MCM5, TOP2A, MYBL2, PIK3CA, DROSHA, MKI67, MMP9, and MCM2.
  • the methods described herein further comprise detecting increased expression of the mRNA of one or more of the following genes: CDKN2A, MKI67, TOP2A, and MCM5.
  • the methods described herein further comprise detecting increased expression of the mRNA of one or more of the following genes: CDKN2A, MKI67, TOP2A, MCM5, BIRC5, MMP9, and MCM2.
  • Appropriate genes for use as reference genes when detecting mRNA expression include those as to which the quantity of the product does not vary between normal and cancerous cervical cells, or between different cell lines or under different growth and sample preparation conditions.
  • endogenous housekeeping genes useful as normalization controls in the methods described herein include, but are not limited to, ACTB, B2M, GAPDH, GUSB, HPRTl, PPIA, RPLP,TBP, TUBB, UBC, PGKl and RPL4.
  • the at least one endogenous housekeeping gene for use in normalizing the measured quantity of mRNAs is selected from GAPDH, TBP and ACTB. In some embodiments, one housekeeping gene is used for normalization. In some embodiments, more than one housekeeping gene is used for normalization.
  • the methods described herein further comprise detecting chromosomal codefendants, i.e., target RNAs clustered near each other in the human genome which tend to be regulated together. Accordingly, in further embodiments, the methods comprise detecting the expression of one or more target microRNAs, each situated within the chromosome no more than 50,000 bp from the chromosomal location of the pre-microRNA sequences in Table 2.
  • RNA was prepared from eight different cell lines of cervical origin that are commonly used in studies of cervical dysplasia and/or carcinoma. The RNA was used for both microRNA array profiling, further described below, and mRNA expression studies.
  • cell lines were selected for diversity, deriving from various squamous cervical cancers (SCC) and adenocarcinomas (AC) and, in most cases, chronically infected with HPV.
  • Cell line C-33A appears to be HPV negative, but likely originally contained HPV.
  • seven of the eight cell lines chosen were derived from primary lesions.
  • One cell line, ME- 180 was derived from a metastatic source. All cell lines were purchased from LGC Promochem (ATCC) and cultured according to ATCCs guidelines. Table 5
  • RNA samples were prepared using TRIzol® (InvitrogenTM) according to the manufacturer's protocol. All RNA samples were diluted in RNase-free water and stored in -80°C (-112°F). OD260/280 was measured on a spectrophotometer.
  • CaSki (CRL-1550) 1000 300 300 1 ,8 sw756 (CRL-10302) 2716 150 407 4 1 5
  • RNA quality was assessed by calculating OD 260/280 ratios, and by electrophoresis on agarose gels under denaturing conditions. The quality of all RNA samples was high as assessed using an Agilent Bioanalyser 2100, as exemplified by the electropherogram shown in Figure 1 obtained for total RNA from cell line CaSki. Figure 2 shows denaturing gel electrophoresis of total RNA from the cell lines. The quantity was sufficient for microRNA array profiling and quantitative RT-PCR of both microRNA and mRNA.
  • RNA from normal cervix was purchased for use as a control from Ambion (Applied Biosystems).
  • RNA fractionator (Ambion). The gel purification protocol enriches for small RNAs less than about 40 nucleotides (nt) long, including microRNAs. Briefly, a total RNA sample (prepared as above) was loaded onto a pre-cast gel using the Flash PAGE Fractionator. The total RNA fraction smaller than 40 nt (the "microRNA fraction") was recovered after gel migration and resuspended into nuclease free water.
  • the polynucleotide probes used for microarray preparation had the configuration 5'-NH 2 -(C) 6 -(spacer)-(oligomer probe sequence)-3'.
  • the 5 '-amino group allowed chemical bonding onto the array support.
  • the probes were spotted in duplicate using the Nexterion spotting buffer provided with the array glass support by Schott with 1 % SDS (sodium dodecyl sulfate) added to allow larger spot sizes (e.g., 100-150 microns compared to 70-100 microns without SDS).
  • the spotter used was the QArray mini (Genetix) equipped with Stealth SMP3 pins (Telechem). After deposition of one series of spots, the spotting needle was washed 5 times with 6OmM NaOH before spotting the next series of probes. Each slide is designed with 32 blocks of spotted probes, with each block being a 20x20 square of spotted probes. Each probe was spotted in duplicate. Spotted glass slides were stored at 4°C until use.
  • microRNA fraction was adapted from a published protocol developed at EMBL (Heidelburg, Germany) by the European Molecular Biology Group (Castoldi et al, "A sensitive array for microRNA expression profiling (miChip) based on locked nucleic acids (LNA),” RNA 2006 May;12(5):913-20. Epub 2006 Mar 15, incorporated herein by reference in its entirety).
  • the microRNA fraction was incubated for 6 hours at 4°C with a mixture containing 10 ⁇ M of dye-labelled tetra-nucleotide (5'-rUrUrUrU -Cy5- 3') (or alternatively, 5'-rUrUrUrU - Cy3-3') (Biospring, Germany) in Ambion buffer diluted to IX with RNase free water, 8% polyethylene glycol (PEG), 2 mM adenosine triphosphate (ATP), and T4 RNA ligase (0.7U/ ⁇ l).
  • PEG polyethylene glycol
  • ATP 2 mM adenosine triphosphate
  • T4 RNA ligase 0.7U/ ⁇ l
  • the labelled microRNA fraction was hybridized to the spotted arrays using a Discovery hybridization station (Ventana, Arlington, AZ). Briefly, 2 mL of a mixture of 1% BSA, 2X SSC, and 0.2 % SDS was incubated with the chips for 30 min at 42 0 C. Then the chips were washed once using EZ Prep buffer (Ventana) and then three more times with Ribowash (Ventana). Next, 20 ⁇ l of the labelled microRNA mixture and 180 ⁇ l of ChipHybe Reagent (Ventana) were added to the array. The arrays were heated for 6 minutes at 37°C, then were incubated at 42°C for 8 hours, after which the heating was stopped.
  • a Discovery hybridization station Ventana, Arlington, AZ.
  • the chips were washed once with Ribowash (Ventana) and then heated for 2 minutes at 37°C.
  • the chips were washed again with Ribowash (Ventana) with one drop of CheapClean (Ventana) added, and incubated for 2 minutes at 37°C.
  • the chips were washed two more times using Ribowash (Ventana).
  • the chips were then stored dry at room temperature overnight. On the following day, the final washes were done according to Ventana's instructions for the Discovery hybridization station.
  • the slides were washed twice with 2X SSC + 0.2X SDS buffer and then one more time with 0.1X SSC. All the slides were dried using a speed centrifuge from Arrayit (TeleChem International, Sunnyvale, CA) at room temperature and kept in the dark before scanning.
  • the following solution may be used for array hybridization (solution 2) to form probe:target RNA hybrids by mixing 2 parts of 1.5X TMAC Hybridization Solution to 1 part (v:v) sample, so that the final component concentrations are 3M TMAC, 0.10% Sarkosyl, 50 mM Tris, and 4 mM EDTA, and incubating on the array at 42 0 C for 8 h:
  • the image was formatted in tif format, defined by an image color depth of 16 bits/pixel (1600*1600). At such setting, pixels can assume intensity values ranging from 0 to 65,535. Pixels exhibiting the maximum intensity value are "saturated” and were assigned the value of 65,535. The resolution of the array scan was set at 10 ⁇ m/pixel. For hybridization experiments using different fluorescent dyes (e.g., Cy5 and Cy3) the photomultiplier tube (PMT) was adjusted to the higher intensity spot (Cy3 is scanned at lower PMT settings than Cy5).
  • the PMT of the laser scanner digitized the captured fluorescence intensity for each given "point" of a slide and stored the numerical value as a pixel corresponding to that point. A picture composed of such pixels was then analyzed.
  • the first task for image analysis was to detect the spot position, using a process called segmentation. Spots were segmented by circles of adaptable or fixed radius. To be reliably segmented and quantified, the spot diameter was required to be more than 5 - 6 pixels. Before segmentation an indexing grid was provided giving the approximate positions of the spots. The segmentation itself detected the limits of spots near the grid circles. Briefly, the Genepix software assigns a circle to each spot on the array (segmentation). The segmentation had to be conducted in a somewhat flexible way due to spotting imperfections and/or support deformation, as the spots were almost never on a perfectly rectangular grid.
  • the second task of image analysis was to quantify spots and export the data into a result file. This was a relatively easy and well-defined task once the spots were located on the image.
  • the statistical approach used most frequently to quantify spot intensity was the mean or median of pixels belonging to a spot.
  • the median approach was more robust than the mean value in the presence of outlier pixels. In practice, however, there was little difference in the results obtained using mean or median.
  • Log2ratio where the Log2ratio equals average intensity signal of the duplicated spots/median intensity of all positives controls for the block.
  • the normalization was done per block to avoid non-homogenous labelling of all blocks of the array. This block-by- block normalization has been shown to be more efficient then using overall normalization of the slide.
  • the obtained values are Log2 values.
  • Log2ratio is the ratio between the two conditions compared, or log2(Xcell-line/Xnormal), which is the same as (log2Xcell-line - log2Xnormal), where X is the measured intensity value. In cases where there was no signal from the "normal" condition, the lowest measured intensity value in the experiment was used as the baseline from which a fold- change expression value was calculated. A fold-change value of less than zero corresponds to a down-regulation of (1 /fold-change) times.
  • Luminex technology (Luminex Corp., Austin, TX) is based on liquid phase hybridization to probe-labelled beads, followed by flow cytometry detection of beads with differing ratios of fluorescent dyes. Beads with up to 100 different dye ratios are available, making it possible to interrogate a single sample for up to 100 analytes simultaneously.
  • RNAs used for the array controls are added to the total RNA fraction isolated from the biological samples. Prior to hybridization with Luminex beads, the total RNA preparation is treated to avoid the formation of dendrimers, which result from the circularization of a single RNA molecule, or concatenation to another RNA molecule. To avoid the formation of dendrimers, the RNA is pre-treated with calf intestinal phosphatase (CIP) to remove the 5 '-phosphate groups.
  • CIP calf intestinal phosphatase
  • the CIP reagent can be obtained from Invitrogen (Carlsbad, CA) and the CIP reaction is run according to the manufacturer's protocol.
  • the total RNA fraction is then labelled with biotin using the Vantage microRNA Labelling Kit (Marligen).
  • the labelled fraction is hybridized to the Luminex beads using the Marligen protocol. Briefly, the polynucleotide beads are mixed with the Marligen hybridization solution (1.5 X TMAC) and the labelled total RNA. The hybridization is performed at 60°C for an hour in the dark. After hybridization, the beads are washed using the Luminex standard 6X SSPET wash buffer (sodium phosphate, sodium chloride, EDTA, Triton X-100, pH 7.4).
  • Luminex beads The detection of the Luminex beads is done using streptavidin phycoerythrin (SAPE) (Europa Bioproducts, Cambridge, UK). The SAPE is added to the washed beads according to the Luminex protocol. The beads are then read using the Luminex IS-200 instrument using the high gain setting for better resolution.
  • SAPE streptavidin phycoerythrin
  • the Luminex IS-200 reads at least 25 beads of each dye-ratio in the reaction mix. Each dye-ratio bead corresponds to a particular probe sequence, and the intensity value is returned as an average value of all read beads.
  • the mean fluorescence intensity (MFI) data is normalized using synthetic RNA controls, and fold changes between normal and diseased samples are computed using the Bioplex software (Bio-Rad, Hercules, CA) and the R bioconductor package (Bioconductor: open software development for computational biology and bioinformatics, Genome Biol. 2004;5(10):R80. Epub 2004 Sep 15).
  • FFPE paraffin-embedded
  • RNA samples were diluted in RNase-free water and stored in -80°C (-112°F).
  • RNA fraction was recovered after gel migration and resuspended into nuclease free water.
  • the polynucleotide probes used for microarray preparation had the configuration 5'-NH 2 -(C) 6 -(spacer)-(oligomer probe sequence)-3'.
  • the 5 '-amino group allowed chemical bonding onto the array support.
  • the probes were spotted in duplicate using the Nexterion spotting buffer provided with the array glass support by Schott with 1 % SDS (sodium dodecyl sulfate) added to allow larger spot sizes (e.g., 100-150 microns compared to 70-100 microns without SDS).
  • the spotter used was the QArray mini (Genetix) equipped with Stealth SMP3 pins (Telechem). After deposition of one series of spots, the spotting needle was washed 5 times with 6OmM NaOH before spotting the next series of probes. Each slide is designed with 48 blocks of spotted probes, with each block being a 20x18 square of spotted probes. Each probe was spotted in duplicate. Spotted glass slides were stored at 4°C until use.
  • microRNA fraction was adapted from a published protocol developed at EMBL (Heidelburg, Germany) by the European Molecular Biology Group (Castoldi et al. , "A sensitive array for microRNA expression profiling (miChip) based on locked nucleic acids (LNA),” RNA 2006 May; 12(5):913-20. Epub 2006 Mar 15, incorporated herein by reference in its entirety).
  • the microRNA fraction was incubated for 6 hours at 4°C with a mixture containing 10 ⁇ M of dye-labelled tetra-nucleotide (5'-rUrUrUrU -Cy5- 3') (or alternatively, 5'-rUrUrUrU - Cy3-3') (Biospring, Germany) in Ambion buffer diluted to IX with RNase free water, 8% polyethylene glycol (PEG), 2 mM adenosine triphosphate (ATP), and T4 RNA ligase (0.7U/ ⁇ l).
  • PEG polyethylene glycol
  • ATP 2 mM adenosine triphosphate
  • T4 RNA ligase 0.7U/ ⁇ l
  • the labelled microRNA fraction was hybridized to the spotted arrays using a Discovery hybridization station (Ventana, Arlington, AZ). Briefly, 2 mL of a mixture of 1% BSA, 2X SSC, and 0.2 % SDS was incubated with the chips for 30 min at 42°C. Then the chips were washed once using EZ Prep buffer (Ventana) and then three more times with Ribowash (Ventana). Next, 20 ⁇ l of the labelled microRNA mixture and 180 ⁇ l of ChipHybe Reagent (Ventana) were added to the array. The arrays were heated for 6 minutes at 37°C, then were incubated at 42°C for 8 hours, after which the heating was stopped.
  • EZ Prep buffer Ventana
  • ChipHybe Reagent ChipHybe Reagent
  • the chips were washed once with Ribowash (Ventana) and then heated for 2 minutes at 37 0 C.
  • the chips were washed again with Ribowash (Ventana) with one drop of CheapClean (Ventana) added, and incubated for 2 minutes at 37°C.
  • the chips were washed two more times using Ribowash (Ventana).
  • the final washes were done according to Ventana' s instructions for the Discovery hybridization station.
  • the slides were washed twice with 2X SSC + 0.2X SDS buffer and then one more time with 0.1X SSC. All the slides were dried using a speed centrifuge from Arrayit (TeleChem International, Sunnyvale, CA) at room temperature and kept in the dark before scanning.
  • the image was formatted in tif format, defined by an image color depth of 16 bits/pixel (1600*1600). At such setting, pixels can assume intensity values ranging from 0 to 65,535. Pixels exhibiting the maximum intensity value are "saturated” and were assigned the value of 65,535. The resolution of the array scan was set at 10 ⁇ m/pixel. For hybridization experiments using different fluorescent dyes (e.g., Cy5 and Cy3) the photomultiplier tube (PMT) was adjusted to the higher intensity spot (Cy3 is scanned at lower PMT settings than Cy5).
  • the PMT of the laser scanner digitized the captured fluorescence intensity for each given "point" of a slide and stored the numerical value as a pixel corresponding to that point. A picture composed of such pixels was then analyzed.
  • the first task for image analysis was to detect the spot position, using a process called segmentation.
  • Spots were segmented by circles of adaptable or fixed radius. To be reliably segmented and quantified, the spot diameter was required to be more than 5 - 6 pixels. Before segmentation an indexing grid was provided giving the approximate positions of the spots. The segmentation itself detected the limits of spots near the grid circles.
  • the Genepix software assigns a circle to each spot on the array (segmentation). The segmentation had to be conducted in a somewhat flexible way due to spotting imperfections and/or support deformation, as the spots were almost never on a perfectly rectangular grid.
  • the second task of image analysis was to quantify spots and export the data into a result file. This was a relatively easy and well-defined task once the spots were located on the image.
  • the statistical approach used most frequently to quantify spot intensity was the mean or median of pixels belonging to a spot.
  • the median approach was more robust than the mean value in the presence of outlier pixels. In practice, however, there was little difference in the results obtained using mean or median.
  • Log2ratio where the Log2ratio equals average intensity signal of the duplicated spots/median intensity of all positives controls for the block.
  • the normalization was done per block to avoid non-homogenous labelling of all blocks of the array. This block-by- block normalization has been shown to be more efficient then using overall normalization of the slide.
  • the obtained values are Log2 values.
  • Log2ratio is the ratio between the two conditions compared, or log2(Xcell-line/Xnormal), which is the same as (log2Xcell-line - log2Xnormal), where X is the measured intensity value. In cases where there was no signal from the "normal" condition, the lowest measured intensity value in the experiment was used as the baseline from which a fold- change expression value was calculated. A fold-change value of less than zero corresponds to a down-regulation of (1 /fold-change) times.
  • miR-21 was up-regulated in all of the tumors tested compared to the normal samples.
  • certain tumor samples for example, ADC- Ia, ADC-Ib, ASCC-Ib, SCC-4a, and SCC-7
  • a particularly high or low number of sequences was detected.
  • Two microRNAs, miR-145 and miR-143 which have previously been reported to be down-regulated in certain cancer tissues (Wang et al., PLoS One (2008) 3: e2557), were also found to be downregulated in this experiment.
  • Table 10 shows a list of the microRNAs that were found to be upregulated in at least one of the tumor samples tested.
  • Table 11 shows the probe sequences that were used to detect the microRNAs listed in Table 10.
  • Table 12 shows the microRNA precursor sequences and their chromosomal location.
  • Table 12 microRNA precursor sequences and chromosomal locations
  • Control HeatMap allows verification, after normalization, of: a. the low signal of the negative control, b. the specificity (CTL26 versus CTL26_MUT), c. the approximate equality between positive control signals (without CTL30, which is a purification and labelling control), d. the approximate equality between block signal medians (based only on positive controls), e. the approximate equality between array signal medians (based on all sequences detected) f. the purification and labelling control (CTL30).
  • Control HeatMap The positive, block, and labeling/purification controls gave approximately the same normalized signals. We found that the specificity of all of the arrays was excellent.
  • RNA from the cell lines was prepared as described in Example 1.
  • RNA from two normal cervix and one squamous cervical tumor (SCC) were purchased from Applied Biosystems (Foster City, CA). See Table 14.
  • RNA samples showed a good A260/280 ratio.
  • Reverse transcription was performed using 1 ⁇ g total RNA.
  • the cDNA preparation was performed in a 50 ⁇ L reaction volume using random hexamers and TaqMan® reverse transcription reagents (Applied Biosystems; Foster City, CA) according to manufacturer's instructions. In parallel, the same reactions were performed without the reverse transcriptase as no-RT controls.
  • ACTB and TBP were used as reference genes.
  • ACTB ACTB
  • TBP TBP
  • GAPDH GAPDH
  • CDKN2A SEQID forward 5 ' -CATAGATGCCGCGGAAGGT-S ' 318 reverse 5 ' -CCCGAGGTTTCTCAGAGCCT-B ' 319 probe FAM-CCTCAGACATCCCCGATTGAAAGAACC-TAMRA 320
  • GenEx 4.4.2 software was used (multiD analysis, Gothenburg, Sweden).
  • the GenEx software uses the ⁇ Ct formula, compensating for differences in PCR efficiency. All fold-change values are calculated relative to one normal sample from Ambion (AM6992).
  • the GenEx statistical module was used for descriptive statistics and t-test analysis.
  • PCR Specificity was determined using no-RT controls and analysis of amplicon size on an agarose gel. All amplicons tested were of the expected size, and we observed no amplification in no-RT controls, except for the no-RT controls for PCNA and RPSA, which was at very low levels and may have been due to contaminating DNA.
  • PCR Efficiency In order to estimate the PCR efficiency for target and control mRNAs, a pool of cDNA from all of the cell lines was diluted in three-fold steps to generate a standard curve. PCR efficiency was calculated by the MX3000p software (Stratagene). Most assays had a similar efficiency, as shown in Table 20, except for MMP2, IGF2BP3, PIK3CA and POU4F1, which had very low expression levels in the cell lines, so it was not possible to generate a standard curve. We found that MMP9 was not expressed in cell lines at all.
  • Table 20 PCR efficiency in pooled cDNA from cell lines
  • IGF2BP3 showed low to absent expression in all tumor an normal samples. Interestingly, IGF2BP3 was only detected in tumor samples, although with very high Ct values, and not in any of the normal samples.
  • TOP2A, and MCM5 were clearly elevated, and that elevation was statistically significant. Increased expression of at least those four mRNAs, and possibly others, correlate with tumors.
  • PreservCyt transport media (Cytec) were used in this study. Four mL of each cell suspension was centrifuged at 2200xg for 15 minutes. The cell pellet was mixed with 700 ⁇ L QIAzol lysis reagent (Qiagen; Hilden, Germany). Total RNA was extracted using miRNeasy RNA extraction kit (Qiagen; Hilden, Germany), according to manufacturer's protocol. The RNA concentration of each sample was measured with a NanoDrop instrument (Thermo Scientific; Wilmington, DE).
  • cDNA Synthesis Reverse transcription was performed using 10 ⁇ L total RNA (0.1 - 0.5 ⁇ g) in a 20 ⁇ L reaction volume using random hexamers and the High Capacity cDNA RT kit (Applied Biosystems, Inc.; Foster City, CA) according to manufacturer's protocol. In parallel, the same reactions were performed without the reverse transcriptase for no RT controls.
  • GAPDH and TBP were selected as references for this experiment.
  • CDKN2A and MKI67 were selected as target mRNAs.
  • PCR Reactions All PCR reactions were performed in triplicate in a 25 ⁇ L reaction volume on a Stratagene MX3000p instrument using a cDNA concentration corresponding to about 2 ng total RNA/reaction. TaqMan® Universal PCR Master Mix (Applied Biosystems, Inc; Foster City, CA) was used for all reactions. Primer and probe sequences, concentrations and thermal cycling conditions were the same as discussed in Example 4. All probes were FAM-labeled and all reactions were run in singleplex.
  • GenEx software uses the ⁇ Ct formula, compensating for differences in PCR efficiency.
  • RNA yield varied between 0.3 ⁇ g - 4 ⁇ g.
  • Four of the samples yielded insufficient concentrations of RNA to be used for cDNA synthesis.
  • Ten out of twenty-one cDNAs were further analyzed for mRNA expression; the remaining samples resulted in Ct values that were too high to be reliable.
  • Ct values for the analyzed samples are shown in Table 26.
  • the relative Iog2 fold-changes are plotted in Figure 4.
  • LSIL low grade squamous intraepithelial lesion
  • HSIL high grade squamous intraepithelial lesion
  • RNA from cervical tumor and normal specimens were prepared as in Example 3.
  • MicroRNAs Selected for Analysis miR-21 was analyzed for each of the samples shown in Table 27. miR-1290 was analyzed for a subset of the samples, as shown in Table 27. RNU44, U47, and RNU48 were used for normalization.
  • RNA samples and microRNAs selected for expression analysis are listed in Table 27.
  • SCC Squamous Cervical Carcinoma
  • ADC Cervical Adenocarcinoma
  • ASCC Adeno-Squamous Cervical Carcinoma
  • AIS Adenocarcinoma in situ
  • RT-PCR Reactions All microRNA RT-PCR assays were purchased from Applied Biosystems Inc (Foster City, CA), including those for normalization genes. The cDNA synthesis and PCR reactions were performed according to manufacturer's protocols. All PCR reactions were run on an MX3000 instrument (Stratagene).
  • GenEx 4.4.2 software (multiD analysis, Gothenburg, Sweden) was used.
  • the GenEx software uses the ⁇ Ct formula, compensating for differences in PCR efficiency.
  • the GenEx statistical module was used for t-test analysis.
  • MiR-205 and miR-1290 show a statistically significant difference in expression levels between tumor and normal samples. See Figures 5 and 6. Interestingly, miR-205 is markedly down-regulated in adeno-carcinoma specimens, which originate from glandular endothelial cells, while it is upregulated in squamous carcinoma specimens, which are derived from epithelial cells. This result is consistent with reports discussing other forms of cancer, such as lung cancer.
  • microRNA sequencing (smRNASeq) datasets were analysed using the probe sequences to identify expressed microRNAs detected by those sequences.
  • the analysis identified 44 sequences with precise ends, corresponding to 37 arms (i.e., some of the sequences appear to be isomirs, or multiple candidate microRNAs from a single arm).
  • Those 44 candidate microRNA sequences are show in Table 28.
  • Table 28 microRNA candidate sequences corresponding to probes
EP10703542A 2009-01-16 2010-01-15 Verfahren zum nachweis von gebärmutterhalskrebs Withdrawn EP2387616A2 (de)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP13150278.3A EP2641976A3 (de) 2009-01-16 2010-01-15 Verfahren zur Erkennung von Gebärmutterhalskrebs

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US14543909P 2009-01-16 2009-01-16
US16583509P 2009-04-01 2009-04-01
PCT/US2010/021274 WO2010083464A2 (en) 2009-01-16 2010-01-15 Methods of detecting cervical cancer

Publications (1)

Publication Number Publication Date
EP2387616A2 true EP2387616A2 (de) 2011-11-23

Family

ID=42340309

Family Applications (2)

Application Number Title Priority Date Filing Date
EP13150278.3A Withdrawn EP2641976A3 (de) 2009-01-16 2010-01-15 Verfahren zur Erkennung von Gebärmutterhalskrebs
EP10703542A Withdrawn EP2387616A2 (de) 2009-01-16 2010-01-15 Verfahren zum nachweis von gebärmutterhalskrebs

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP13150278.3A Withdrawn EP2641976A3 (de) 2009-01-16 2010-01-15 Verfahren zur Erkennung von Gebärmutterhalskrebs

Country Status (3)

Country Link
EP (2) EP2641976A3 (de)
CA (1) CA2749841A1 (de)
WO (1) WO2010083464A2 (de)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2825522A1 (en) 2011-01-26 2012-08-02 Cepheid Methods of detecting lung cancer
WO2014164753A1 (en) 2013-03-11 2014-10-09 Emory University Methods and compositions for managing vascular conditions

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2004121218A (ja) * 2002-08-06 2004-04-22 Jenokkusu Soyaku Kenkyusho:Kk 気管支喘息または慢性閉塞性肺疾患の検査方法
CN101031658A (zh) * 2004-05-26 2007-09-05 罗塞塔金诺米克斯有限公司 病毒和病毒相关mirna及其用途
FR2873625B1 (fr) 2004-08-02 2008-05-30 Michelin Soc Tech Assemblage pneu/roue pourvu d'une composition lubrifiante
EP1786932A4 (de) 2004-08-23 2010-10-27 Us Genomics Inc Systeme und verfahren zur detektion und analyse von polymeren
US20060094025A1 (en) 2004-11-02 2006-05-04 Getts Robert C Methods for detection of microrna molecules
DE602006016739D1 (de) 2005-01-25 2010-10-21 Rosetta Inpharmatics Llc Verfahren zur quantifizierung kleiner rna-moleküle
US20070054287A1 (en) 2005-05-31 2007-03-08 Applera Corporation Method for identifying medically important cell populations using micro rna as tissue specific biomarkers
WO2006130761A1 (en) 2005-05-31 2006-12-07 Applera Corporation Linear amplification of short nucleic acids
AU2006341607B2 (en) 2005-05-31 2011-03-17 Applied Biosystems, Llc. Multiplexed amplification of short nucleic acids
US20060292616A1 (en) 2005-06-23 2006-12-28 U.S. Genomics, Inc. Single molecule miRNA-based disease diagnostic methods
EP2308990B1 (de) 2005-07-15 2012-09-26 Life Technologies Corporation Analyse von Boten-RNA und Mikro-RNA im gleichen Reaktionsansatz
EP1924713B1 (de) 2005-08-24 2011-11-09 Life Technologies Corporation Verfahren zur quantifizierung von sirnas, mirnas und polymorphen mirnas
EP2126141A4 (de) * 2007-03-15 2010-08-11 Univ Cleveland Hospitals Screening, diagnose, behandlung und prognose pathophysiologischer zustände durch rna-regulation
WO2008125883A1 (en) * 2007-04-16 2008-10-23 Cancer Research Technology Limited Cancer markers for prognosis and screening of anti-cancer agents
EP2198050A1 (de) * 2007-09-14 2010-06-23 Asuragen, INC. Mikrornas mit unterschiedlicher expression bei zervikalkarzinom und verwendungen davon
EP2260110B1 (de) * 2008-02-08 2014-11-12 Asuragen, INC. In lymphknoten von krebspatienten differentiell exprimierte mirnas

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2010083464A2 *

Also Published As

Publication number Publication date
EP2641976A2 (de) 2013-09-25
CA2749841A1 (en) 2010-07-22
EP2641976A3 (de) 2014-01-08
WO2010083464A2 (en) 2010-07-22
WO2010083464A3 (en) 2011-05-12

Similar Documents

Publication Publication Date Title
US10000808B2 (en) Methods of detecting cervical cancer
EP2391738B1 (de) Verfahren für den nachweis von sepsis
JP5755569B2 (ja) 肺癌を検出する方法
US9365903B2 (en) Compositions comprising polynucleotides for detecting lung cancer
Guerau-de-Arellano et al. miRNA profiling for biomarker discovery in multiple sclerosis: from microarray to deep sequencing
US7993831B2 (en) Methods of normalization in microRNA detection assays
US20160024586A1 (en) Methods of detecting cancer
AU2012308365A1 (en) Methods of detecting sepsis
EP2641976A2 (de) Verfahren zur Erkennung von Gebärmutterhalskrebs
EP2839028B1 (de) Verfahren zur erkennung von blasenkrebs
US20160108478A1 (en) Methods of detecting lung cancer
US20160122834A1 (en) Methods of detecting ebola

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20110816

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20120627

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20130108