miR-10 Regulated Genes and Pathways as Targets for Therapeutic Intervention

Abstract
The present invention concerns methods and compositions for identifying genes or genetic pathways modulated by miR-10, using miR-10 to modulate a gene or gene pathway, using this profile in assessing the condition of a patient and/or treating the patient with an appropriate miRNA.
Description
BACKGROUND OF THE INVENTION

I. Field of the Invention


The present invention relates to the fields of molecular biology and medicine. More specifically, the invention relates to methods and compositions for the treatment of diseases or conditions that are affected by microRNA (miRNA) miR-10 expression or lack thereof, and genes and cellular pathways directly and indirectly modulated by such.


II. Background


In 2001, several groups used a cloning method to isolate and identify a large group of “microRNAs” (miRNAs) from C. elegans, Drosophila, and humans (Lau et al., 2001; Lee and Ambros, 2001; Lagos-Quintana et al., 2003). Several hundreds of miRNAs have been identified in plants and animals—including humans—which do not appear to have endogenous siRNAs. Thus, while similar to siRNAs, miRNAs are distinct.


miRNAs thus far observed have been approximately 17-24 nucleotides in length, and they arise from longer precursors, which are transcribed from non-protein-encoding genes (Carrington and Ambros, 2003). The precursors form structures that fold back on themselves in self-complementary regions; they are then processed by the nuclease Dicer (in animals) or DCL1 (in plants) to generate the short double-stranded miRNA. One of the miRNA strands is incorporated into a complex of proteins and miRNA called the RNA-induced silencing complex (RISC). The miRNA guides the RISC complex to a target mRNA, which is then cleaved or translationally silenced, depending on the degree of sequence complementarity of the miRNA to its target mRNA. Currently, it is believed that perfect or nearly perfect complementarity leads to mRNA degradation, as is most commonly observed in plants. In contrast, imperfect base pairing, as is primarily found in animals, leads to translational silencing. However, recent data suggest additional complexity (Bagga et al., 2005; Lim et al., 2005), and mechanisms of gene silencing by miRNAs remain under intense study (Chendrimada et al., 2007; Kiriakidou et al., 2007).


Recent studies have shown that changes in the expression levels of numerous miRNAs are associated with various cancers (reviewed in Calin and Croce, 2006; Esquela-Kerscher and Slack, 2006; Wiemer, 2007). miRNAs have also been implicated in regulating cell growth and cell and tissue differentiation—cellular processes that are associated with the development of cancer.


The inventors previously demonstrated that hsa-miR-10 family members are involved with the regulation of numerous cell activities that represent intervention points for cancer therapy and for therapy of other diseases and disorders (U.S. patent application Ser. No. 11/141,707 filed May 31, 2005 and Ser. No. 11/273,640 filed Nov. 14, 2005, each of which is incorporated herein by reference). When transfected into a human leukemic T cell line (Jurkat), synthetic miR-10a increased the viability of those cells. The same effect was observed when synthetic miR-10a or miR-10b was transfected into normal, primary human T cells. However, transfection of synthetic miR-10b into Jurkat cells resulted in a reduction in cell viability. Synthetic inhibitors of miR-10 and miR-10b increased the proliferation of basal cell carcinoma cells (TE354T) and of normal human breast epithelial cells (MCF12A); whereas, an inhibitor of miR-10b was shown to increase proliferation of human prostate cancer cells (22Rv1). Upon transfection, miR-10a caused an increase in the programmed cell death (apoptosis) of 22Rv1 cells and miR-10b caused an increase in apoptosis in Jurkat cells. Apoptosis is a natural cellular process that helps control cancer by inducing death in cells with oncogenic potential. Many oncogenes function by altering induction of apoptosis. The inventors also previously observed that miR-10a is expressed at lower levels in 5 of 6 colon tumors than in adjacent normal colon tissue samples.


Others have observed that miR-10b is upregulated in metastatic, human breast cancer cells but not in breast cancer cell lines that have little if any metastatic properties (Ma et al., 2007). In primary breast tumors (independent of their clinical aggressiveness), miR-10b was downregulated relative to normal breast tissue (Iorio et al., 2005). Using a rat model for inflammatory muscular pain, others observed a downregulation of miR-10a, upon the induction of pain with intramuscular injection of complete Freund's adjuvant (Bai et al., 2007). Hsa-miR-10a was also observed to be down-regulated during the in vitro megakaryocytic differentiation from bone marrow CD34+ progenitor cells (Garzon et al., 2006).


Bioinformatics analyses suggest that any given miRNA may bind to and alter the expression of up to several hundred different genes. In addition, a single gene may be regulated by several miRNAs. Thus, each miRNA may regulate a complex interaction among genes, gene pathways, and gene networks. Mis-regulation or alteration of these regulatory pathways and networks involving miRNAs are likely to contribute to the development of disorders and diseases such as cancer. Although bioinformatics tools are helpful in predicting miRNA binding targets, all have limitations. Because of the imperfect complementarity with their target binding sites, it is difficult to accurately predict the mRNA targets of miRNAs with bioinformatics tools alone. Furthermore, the complicated interactive regulatory networks among miRNAs and target genes make it difficult to accurately predict which genes will actually be mis-regulated in response to a given miRNA.


Correcting gene expression errors by manipulating miRNA expression or by repairing miRNA mis-regulation represent promising methods to repair genetic disorders and cure diseases like cancer. A current, disabling limitation of this approach is that, as mentioned above, the details of the regulatory pathways and gene networks that are affected by any given miRNA, have been largely unknown. This represents a significant limitation for treatment of cancers in which a specific miRNA may play a role. A need exists to identify the genes, genetic pathways, and genetic networks that are regulated by or that may regulate expression of miRNAs.


SUMMARY OF THE INVENTION

The present invention provides additional compositions and methods by identifying genes that are direct targets for miR-10 regulation or that are indirect or downstream targets of regulation following the miR-10-mediated modification of another gene(s) expression. Furthermore, the invention describes gene, disease, and/or physiologic pathways and networks that are influenced by miR-10 and its family members. In certain aspects, compositions of the invention are administered to a subject having, suspected of having, or at risk of developing a metabolic, an immunologic, an infectious, a cardiovascular, a digestive, an endocrine, an ocular, a genitourinary, a blood, a musculoskeletal, a nervous system, a congenital, a respiratory, a skin, or a cancerous disease or condition.


In particular aspects, a subject or patient may be selected for treatment based on expression and/or aberrant expression of one or more miRNA or mRNA. In a further aspect, a subject or patient may be selected for treatment based on aberrations in one or more biologic or physiologic pathway(s), including aberrant expression of one or more gene associated with a pathway, or the aberrant expression of one or more protein encoded by one or more gene associated with a pathway. In still a further aspect, a subject or patient may be selected based on aberrations in miRNA expression, or biologic and/or physiologic pathway(s). A subject may be assessed for sensitivity, resistance, and/or efficacy of a therapy or treatment regime based on the evaluation and/or analysis of miRNA or mRNA expression or lack thereof. A subject may be evaluated for amenability to certain therapy prior to, during, or after administration of one or more therapy to a subject or patient. Typically, evaluation or assessment may be done by analysis of miRNA and/or mRNA, as well as combination of other assessment methods that include but are not limited to histology, immunohistochemistry, blood work, etc.


Many of these genes and pathways are associated with various cancers and other diseases. Cancerous conditions include, but are not limited to astrocytoma, acute lymphoblastic leukemia, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, chronic lymphoblastic leukemia, chronic myeloid leukemia, colorectal carcinoma, endometrial carcinoma, Ewing's sarcoma, fibrosarcoma, glioma, glioblastoma, glioblastoma multiforme, gastric carcinoma, hepatoblastoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, lung carcinoma, leiomyosarcoma, liposarcoma, melanoma, mantle cell lymphoma, multiple myeloma, neuroblastoma, non-Hodgkin lymphoma, nasopharyngeal carcinoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, retinoblastoma, renal cell carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma, Wilm's tumor, wherein the modulation of one or more gene is sufficient for a therapeutic response. In certain aspects, a cancerous condition is includes or is selected from the group that includes astrocytoma, acute lymphoblastic leukemia, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, chronic lymphoblastic leukemia, chronic myeloid leukemia, colorectal carcinoma, chondrosarcoma, endometrial carcinoma, Ewing's sarcoma, fibrosarcoma, glioma, glioblastoma, glioblastoma multiforme, gastric carcinoma, hepatoblastoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, lung carcinoma, leiomyoma, liposarcoma melanoma, mantle cell lymphoma, multiple myeloma, mesothelioma, neuroblastoma, non-Hodgkin lymphoma, nasopharyngeal carcinoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, retinoblastoma, renal cell carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma, T-cell leukemia, and/or Wilm's tumor. Typically a cancerous condition is an aberrant hyperproliferative condition associated with the uncontrolled growth or inability to undergo cell death, including apoptosis.


In some embodiments, an infectious disease or condition includes a bacterial, viral, parasite, or fungal infection. In certain aspects, a disease or condition can include blood disorders such as alpha thalassemia, and neurologic disorders such as schizophrenia, Alzheimer disease, or Parkinson disease.


The present invention provides methods and compositions for identifying genes that are direct targets for miR-10 regulation or that are downstream targets of regulation following the miR-10-mediated modification of upstream gene expression. Furthermore, the invention describes gene pathways and networks that are influenced by miR-10 expression in biological samples. Many of these genes and pathways are associated with various cancers and other diseases and disorders. The altered expression or function of miR-10 in cells can lead to changes in the expression of these genes and contribute to the development of disease or other conditions. Introducing miR-10 (for diseases where the miRNA is down-regulated) or a miR-10 inhibitor (for diseases where the miRNA is up-regulated) into disease cells or tissues or subjects would result in a therapeutic response. The identities of key genes that are regulated directly or indirectly by miR-10 and the disease with which they are associated are provided herein.


In certain aspects a cell may be an epithelial, an endothelial, a mesothelial, a glial, a stromal, or a mucosal cell. The cell can be, but is not limited to a brain, a neuronal, a blood, an endometrial, a meninges, an esophageal, a lung, a cardiovascular, a liver, a lymphoid, a breast, a bone, a connective tissue, a fat, a retinal, a thyroid, a glandular, an adrenal, a pancreatic, a stomach, an intestinal, a kidney, a bladder, a colon, a prostate, a uterine, an ovarian, a cervical, a testicular, a splenic, a skin, a smooth muscle, a cardiac muscle, or a striated muscle cell. A cell, tissue, or subject may be a cancer cell, a cancerous tissue, harbor cancerous tissue, or be a subject or patient diagnosed or at risk of developing a disease or condition.


In still a further aspect cancer includes, but is not limited to astrocytoma, acute lymphoblastic leukemia, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, chronic lymphoblastic leukemia, chronic myeloid leukemia, colorectal carcinoma, chondrosarcoma, endometrial carcinoma, Ewing's sarcoma, fibrosarcoma, glioma, glioblastoma, glioblastoma multiforme, gastric carcinoma, hepatoblastoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, lung carcinoma, leiomyoma, liposarcoma melanoma, mantle cell lymphoma, multiple myeloma, mesothelioma, neuroblastoma, non-Hodgkin lymphoma, nasopharyngeal carcinoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, retinoblastoma, renal cell carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma, T-cell leukemia, or Wilm's tumor.


In certain aspects, the cell, tissue, or target may not be defective in miRNA expression yet may still respond therapeutically to expression or over expression of a miRNA. miR-10 could be used as a therapeutic target for any of these diseases or conditions. In certain embodiments miR-10 or its compliment can be used to modulate the activity of miR-10 or a miR-10 regulated gene in a subject, organ, tissue, or cell.


A cell, tissue, or subject may be a cancer cell, a cancerous tissue, harbor cancerous tissue, or be a subject or patient diagnosed or at risk of developing a disease or condition. In certain aspects a cancer cell is an epithelial, an endothelial, a mesothelial, a stromal, a mucosal, a brain, a glial, a neuronal, a blood, a leukemic, an endometrial, a meninges, an esophageal, a lung, a cardiovascular, a liver, a lymphoid, a breast, a bone, a connective tissue, a fat, a retinal, a thyroid, a glandular, a salivary gland, an adrenal, a pancreatic, a stomach, an intestinal, a kidney, a bladder, a colon, a colorectal, a prostate, a uterine, an ovarian, a cervical, a testicular, a splenic, a skin, a smooth muscle, a cardiac muscle, or a striated muscle cell


Embodiments of the invention include methods of modulating gene expression, or biologic or physiologic pathways in a cell, a tissue, or a subject comprising administering to the cell, tissue, or subject an amount of an isolated nucleic acid or mimetic thereof comprising a miR-10 nucleic acid, mimetic, or inhibitor sequence in an amount sufficient to modulate the expression of a gene positively or negatively modulated by a miR-10 miRNA. A “miR-10 nucleic acid sequence” or “miR-10 inhibitor” includes the full length precursor of miR-10, or complement thereof or processed (i.e., mature) sequence of miR-10 and related sequences set forth herein, as well as 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or more nucleotides of a precursor miRNA or its processed sequence, or complement thereof, including all ranges and integers there between. In certain embodiments, the miR-10 nucleic acid sequence or miR-10 inhibitor contains the full-length processed miRNA sequence or complement thereof and is referred to as the “miR-10 full-length processed nucleic acid sequence” or “miR-10 full-length processed inhibitor sequence.” In still further aspects, the miR-10 nucleic acid comprises at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 50 nucleotide segment (including all ranges and integers there between) or complementary segment of a miR-10 that is at least 75, 80, 85, 90, 95, 98, 99 or 100% identical to SEQ ID NO:1 to SEQ ID NO:87. The general term miR-10 includes all members of the miR-10 family that share at least part of a mature miR-10 sequence. Mature miR-10 sequences include ame-miR-10 (ACCCUGUAGAUCCGAAUUUGU, SEQ ID NO:1 (MIMAT0004419)), dre-miR-10b (UACCCUGUAGAACCGAAUUUGUG, SEQ ID NO:2 (MIMAT0001268)), age-miR-10a (UACCCUGUAGAUCCGAAUUUGUG, SEQ ID NO:3 (MIMAT0002489)), gga-miR-10b (UACCCUGUAGAACCGAAUUUGU, SEQ ID NO:4 (MIMAT0001148)), dps-miR-10 (ACCCUGUAGAUCCGAAUUUGU, SEQ ID NO:5 (MIMAT0001214)), mmu-miR-10b (UACCCUGUAGAACCGAAUUUGUG, SEQ ID NO:6 (MIMAT0000208)), rno-miR-10b (CCCUGUAGAACCGAAUUUGUGU, SEQ ID NO:7 (MIMAT0000783)), ppy-miR-10a (UACCCCGUAGAUCCGAAUUUGUG, SEQ ID NO:8 (MIMAT0002487)), hsa-miR-10b* ACAGAUUCGAUUCUAGGGGAAU, SEQ ID NO:9 (MIMAT0004556)), ppa-miR-10b (UACCCUGUAGAACCGAAUUUGU, SEQ ID NO:10 (MIMAT0002493)), tni-miR-10b (UACCCUGUAGAACCGAAUUUGUG, SEQ ID NO:11 (MIMAT0002964)), hsa-miR-10a* (CAAAUUCGUAUCUAGGGGAAUA, SEQ ID NO:12 (MIMAT0004555)), bta-miR-10a (UACCCUGUAGAUCCGAAUUUGUG, SEQ ID NO:13 (MIMAT0003786)), rno-miR-10a-3p (CAAAUUCGUAUCUAGGGGAAUA, SEQ ID NO:14 (MIMAT0004709)), ppa-miR-10a (UACCCUGUAGAUCCGAAUUUGUG, SEQ ID NO:15 (MIMAT0002490)), dme-miR-10 (ACCCUGUAGAUCCGAAUUUGU, SEQ ID NO:16 (MIMAT0000115)), tni-miR-10d (UACCCUGUAGAACCGAAUGUGUGUG, SEQ ID NO:17 (MIMAT0003772)), mmu-miR-10a (UACCCUGUAGAUCCGAAUUUGUG, SEQ ID NO:18 (MIMAT0000648)), ggo-miR-10b (UACCCUGUAGAACCGAAUUUGU, SEQ ID NO:19 (MIMAT0002491)), bta-miR-10b (UACCCUGUAGAACCGAAUUUGUG, SEQ ID NO:20 (MIMAT0003839)), dre-miR-10a (UACCCUGUAGAUCCGAAUUUGU, SEQ ID NO:21 (MIMAT0001267)), dre-miR-10d* (CAGAUUCGGUUUUAGGGGAGUA, SEQ ID NO:22 (MIMAT0003394)), hsa-miR-10a (UACCCUGUAGAUCCGAAUUUGUG, SEQ ID NO:23 (MIMAT0000253)), fru-miR-10c (UACCCUGUAGAUCCGGAUUUGU, SEQ ID NO:24 (MIMAT0003087)), dre-miR-10a* (CAAAUUCGUGUCUUGGGGAAUA, SEQ ID NO:25 (MIMAT0003391)), dre-miR-10d (UACCCUGUAGAACCGAAUGUGUG, SEQ ID NO:26 (MIMAT0001771)), mdo-miR-10a (UACCCUGUAGAUCCGAAUUUGUG, SEQ ID NO:27 (MIMAT0004089)), rno-miR-10a-5p (UACCCUGUAGAUCCGAAUUUGUG, SEQ ID NO:28 (MIMAT0000782)), bmo-miR-10 (ACCCUGUAGAUCCGAAUUUGU, SEQ ID NO:29 (MIMAT0004195)), hsa-miR-10b UACCCUGUAGAACCGAAUUUGUG, SEQ ID NO:30 (MIMAT0000254)), mne-miR-10b UACCCUGUAGAACCGAAUUUGU, SEQ ID NO:31 (MIMAT0002492)), xtr-miR-10b (UACCCUGUAGAACCGAAUUUGU, SEQ ID NO:32 (MIMAT0003558)), dre-miR-10c (UACCCUGUAGAUCCGGAUUUGU, SEQ ID NO:33 (MIMAT0001770)), mmu-miR-10b* (CAGAUUCGAUUCUAGGGGAAUA, SEQ ID NO:34 (MIMAT0004538)), xtr-miR-10c (CACCCUGUAGAAUCGAAUUUGU, SEQ ID NO:35 (MIMAT0003559)), sla-miR-10a (UACCCUGUAGAUCCGAAUUUGUG, SEQ ID NO:36 (MIMAT0002488)), tni-miR-10c (UACCCUGUAGAUCCGGAUUUGU, SEQ ID NO:37 (MIMAT0003088)), fru-miR-10d (UACCCUGUAGAACCGAAUGUGUGUG, SEQ ID NO:38 (MIMAT0003773)), xtr-miR-10a (UACCCUGUAGAUCCGAAUUUGUG, SEQ ID NO:39 (MIMAT0003557)), fru-miR-10b (UACCCUGUAGAACCGAAUUUGUG, SEQ ID NO:40 (MIMAT0002963)), ggo-miR-10a (UACCCUGUAGAUCCGAAUUUGUG, SEQ ID NO:41 (MIMAT0002486)), aga-miR-10 (ACCCUGUAGAUCCGAAUUUGU, SEQ ID NO:42 (MIMAT0001497)), mmu-miR-10a* (CAAAUUCGUAUCUAGGGGAAUA, SEQ ID NO:43 (MIMAT0004659)), mdo-miR-10b (AUACCCUGUAGAACCGAAUUUGU, SEQ ID NO:44 (MIMAT0004090)), or a complement thereof. In certain aspects, a subset of these miRNAs will be used that include some but not all of the listed miR-10 family members. In one aspect, miR-10 sequences have a core consensus sequence of [A][U/C][A/-]CCC[U/C][G/A][U/C]A[G/A]A[U/A][C/U]CG[G/A][A/U]U[U/C][U/G/C][G/A][U/G][G/-][-/U][-/G][-/G][-/G][-/A][-/A][-/U][-/A] (SEQ ID NO:45, wherein the bracketed nucleotides are optional). In one embodiment only sequences comprising the consensus sequence of CCCUGUAGA[A/U]CCG[A/G]AU[U/G]UGU (SEQ ID NO:46)) will be included with all other miRNAs excluded. The term miR-10 includes all members of the miR-10 family unless specifically identified. In certain aspects, a subset of these miRNAs will be used that include some but not all of the listed miR-10 family members. For instance, in one embodiment only sequences comprising the consensus sequence of SEQ ID NO:46 will be included with all other miRNAs excluded.


In a further aspect, a “miR-10 nucleic acid sequence” includes all or a segment of the full length precursor of miR-10 family members. Stem-loop sequences of miR-10 family members include aga-mir-10 (MI0001602, GUCGAUUUAUGUUCUACAUCCACCCUGUAGAUCCGAAUUUGUUUGAAUUUAUAU UAAUAACAAAUUCGGUUCUAGAGAGGUUUGUGUGGGGCAUUUGUUAAC SEQ ID NO:47), age-mir-10a (MI0002791, GAUCUGUCUGUCUUCUGUAUAUACCCUGUAGAUCCGAAUUUGUGUAAGGAAUUU UGUGGUCACAAAUUCGUAUCUAGGGGAAUAUGUAGUUGACAUAAACACUCCGCU CA SEQ ID NO:48), ame-mir-10 (MI0005727, CCCAGUUAAUGCUCUACAUCUACCCUGUAGAUCCGAAUUUGUUUGAUAAGAGGC GACAAAUUCGGUUCUAGAGAGGUUUGUGUGGUGCAUACAGAGCUAC SEQ ID NO:49), bmo-mir-10 (MI0004973, AGUGCCCUACAUCUACCCUGUAGAUCCGAAUUUGUUUGAAGUGAGGCGACAAAU UCGGUUCUAGAGAGGUUUGUGUGGUGCACG SEQ ID NO:50), bta-mir-10a (MI0005007, GAUCUGUCUGUCUUCUGUAUAUACCCUGUAGAUCCGAAUUUGUGUAAGGAAUUU UGUGAUCACAAAUUCGUAUCUAGGGGAAUAUGUAGUUGACAUAAACACUCCGCU C SEQ ID NO:51), bta-mir-10b (MI0005052, CAGUGACGUUGUCUAUAUAUACCCUGUAGAACCGAAUUUGUGUGGUAUCCAUGU AGUCACAGAUUCGAUUCUAGGGGAAUAUAUGGUCGAUGCAAAAAC SEQ ID NO:52), dme-mir-10 (MI0000130, CCACGUCUACCCUGUAGAUCCGAAUUUGUUUUAUACUAGCUUUAAGGACAAAUU CGGUUCUAGAGAGGUUUGUGUGG SEQ ID NO:53), dps-mir-10 (MI0001307, CCACGUCUACCCUGUAGAUCCGAAUUUGUUUUACAUUAGCUUUAAGGACAAAUU CGGUUCUAGAGAGGUUUGUGUGG SEQ ID NO:54), dre-mir-10a (MI0001363, UGUCUGUCAUCUAUAUAUACCCUGUAGAUCCGAAUUUGUGUGAAUAUACAGUCG CAAAUUCGUGUCUUGGGGAAUAUGUAGUUGACAUAAACACAACGC SEQ ID NO:55), dre-mir-10b-1 (MI0001364, GUCUAUAUAUACCCUGUAGAACCGAAUUUGUGUGAAAAAAUAACAUUCACAGAU UCGAUUCUAGGGGAGUAUAUGGUC SEQ ID NO:56), dre-mir-10b-2 (MI0001887, GUAGUCGUCUAUAUGUACCCUGUAGAACCGAAUUUGUGUCCAAAACAUCAAAAU CGCAAAUACGUCUCUACAGGAAUACAUGGGCGACGUAA SEQ ID NO:57), dre-mir-10c (MI0001888, CCUGUCAUCUAUAUAUACCCUGUAGAUCCGGAUUUGUGUAAACAGACGCACAGUC ACAAAUUCGUAUCUAGGGGAGUAUGUAGUUGAUGUAUAGG SEQ ID NO:58), dre-mir-10d-1 (MI0001889, UGGAAGCUUUGUUCCGUCGUCUAUAUAUACCCUGUAGAACCGAAUGUGUGUUUA CACAGCAAAUUCACAGAUUCGGUUUUAGGGGAGUAUAUGGACGAUGCAAAAACG UCUGCUUUCA SEQ ID NO:59), dre-mir-10d-2 (MI0001890, UGGAAGCUUUGUUCCGUCGUCUAUAUAUACCCUGUAGAACCGAAUGUGUGUUUA CACAGCAAAUUCACAGAUUCGGUUUUAGGGGAGUAUAUGGACGAUGCAAAAACG UCUGCUUUCA SEQ ID NO:60), fru-mir-10b-1 (MI0003279, AUAUAUACCCUGUAGAACCGAAUUUGUGUGAUGGCGUCAAAGUCACAGAUUCGA UUCUAGGGGAGUAUAU SEQ ID NO:61), fru-mir-10b-2 (MI0003297, GUUGUCUAUAUGUACCCUGUAGAACCGAAUUUGUGUGAGUUCCAGACAGUCGCA AGUACGUCUCUACAGGAAUACAUGGGCAAC SEQ ID NO:62), fru-mir-10c (MI0003449, CUGUCUUCUAUAUCUACCCUGUAGAUCCGGAUUUGUGUAAAAAUCAUUAAAGCA AUCACAAAUUCGCUUCUAGGGGAGUAUAUAGUGGAUUUAUACACGACG SEQ ID NO:63), fru-mir-10d (MI0004967, CCGGUGAGGUGGAUCGUCGUCUAUAAAUACCCUGUAGAACCGAAUGUGUGUGCA GCUGACUUGAUCACAGAUUGGGUUCUAGGGGAGUCUAUGGGCGAUGAAUAAUCA CUGA SEQ ID NO:64), gga-mir-10b (MI0001216, CAGAACGUUAUUACGUUGUCUAUAUAUACCCUGUAGAACCGAAUUUGUGUGAUA UUCAUAUAGUCACAGAUUCGAUUCUAGGGGAAUAUAUGGUCGAUGCAAAAACUU CA SEQ ID NO:65), ggo-mir-10a (MI0002788, GAUCUGUCUGUCUUCUGUAUAUACCCUGUAGAUCCGAAUUUGUGUAAGGAAUUU UGUGGUCACAAAUUCGUAUCUAGGGGAAUAUGUAGUUGACAUAAACACUCCGCU CU SEQ ID NO:66), ggo-mir-10b (MI0002793, CCAGACAUUGUAACGUUGUCUAUAUAUACCCUGUAGAACCGAAUUUGUGUGGUA UCCAUAUAGUCACAGAUUCGAUUCUAGGGGAAUAUAUGGUCGAUGCAAAAACUU CA SEQ ID NO:67), hsa-mir-10a (MI0000266, GAUCUGUCUGUCUUCUGUAUAUACCCUGUAGAUCCGAAUUUGUGUAAGGAAUUU UGUGGUCACAAAUUCGUAUCUAGGGGAAUAUGUAGUUGACAUAAACACUCCGCU CU SEQ ID NO:68), hsa-mir-10b MI0000267, CCAGAGGUUGUAACGUUGUCUAUAUAUACCCUGUAGAACCGAAUUUGUGUGGUA UCCGUAUAGUCACAGAUUCGAUUCUAGGGGAAUAUAUGGUCGAUGCAAAAACUU CA SEQ ID NO:69), mdo-mir-10a MI0005273, CUGUCUUCUGUAUAUACCCUGUAGAUCCGAAUUUGUGUAAGGAAUUUUGUGGUC ACAAAUUCGUAUCUAGGGGAAUAUGUAGUUGACAUA SEQ ID NO:70), mdo-mir-10b MI0005274, CAGAAUGUUAUUACGUUGUCUAUAUAUACCCUGUAGAACCGAAUUUGUGUGGUA UUUACAUAGUCACAGAUUCGAUUCUAGGGGAAUAUAUGGUCGAUGCAAAAACUU CAC SEQ ID NO:71), mmu-mir-10a MI0000685, GACCUGUCUGUCUUCUGUAUAUACCCUGUAGAUCCGAAUUUGUGUAAGGAAUUU UGUGGUCACAAAUUCGUAUCUAGGGGAAUAUGUAGUUGACAUAAACACUCCGCU CA SEQ ID NO:72), mmu-mir-10b MI0000221, UAUAUACCCUGUAGAACCGAAUUUGUGUGGUACCCACAUAGUCACAGAUUCGAU UCUAGGGGAAUAUA SEQ ID NO:73), mne-mir-10b MI0002794, CAGAGGUUGUAACGUUGUCUAUAUAUACCCUGUAGAACCGAAUUUGUGUGGUAU CCAUAUAGUCACAGAUUCGAUUCUAGGGGAAUAUAUGGUCGAUGCAAAAACUUC A SEQ ID NO:74), ppa-mir-10a MI0002792, GAUCUGUCUGUCUUCUGUAUAUACCCUGUAGAUCCGAAUUUGUGUAAGGAAUUU UGUGGUCACACAUUCGUAUCUAGGGGAAUAUGUAGUUGACAUACACACUCCGCU CU SEQ ID NO:75), ppa-mir-10b MI0002795, CCAGAGGUUGUAACGUUGUCUAUAUAUACCCUGUAGAACCGAAUUUGUGUGGUA UCCGUAUAGUCACAGAUUCGAUUCUAGGGGAAUAUAUGGUCGAUGCAAAAACUU CA SEQ ID NO:76), ppy-mir-10a (MI0002789, GAUCUGUCUGUCUUCUGUAUAUACCCCGUAGAUCCGAAUUUGUGUAAGGAAUUU UGUGGUCACAAAUUCGUAUUUAGGGGAAUAUGUAGUUGACAUAAACACUCCGCU CG SEQ ID NO:77), rno-mir-10a MI0000841, GACCUGUCUGUCUUCUGUAUAUACCCUGUAGAUCCGAAUUUGUGUAAGGAAUUU UGUGGUCACAAAUUCGUAUCUAGGGGAAUAUGUAGUUGACAUAAACACUCCGCU CA SEQ ID NO:78), rno-mir-10b MI0000842, CCAAAGUUGUAACGUUGUCUAUAUAUACCCUGUAGAACCGAAUUUGUGUGGUAC CCACAUAGUCACAGAUUCGAUUCUAGGGGAAUAUAUGGUCGAUGCAAAAACUUC A SEQ ID NO:79), sla-mir-10a (MI0002790, GAUCUGUCUGUCUUCUGUAUAUACCCUGUAGAUCCGAAUUUGUGUAAGGAAUUU UGUGGUCACAAAUUCGUAUCUAGGGGAAUAUGUAGUUGACAUAAACACUCCGCU CA SEQ ID NO:80), tni-mir-10b-1 MI0003298, GUUGUCUAUAUGUACCCUGUAGAACCGAAUUUGUGUGAGUUCAGACAGUCACAA GUACGUCUCUACAGGAAUACAUGGGCAAC SEQ ID NO:81), tni-mir-10b-2 MI0003280, AUAUAUACCCUGUAGAACCGAAUUUGUGUGAUCAAGUCACAGUCACAGAUUCGA UUCUAGGGGAGUAUAU SEQ ID NO:82), tni-mir-10c (MI0003450, GAGCCGCUGUCUUCUAUAUCUACCCUGUAGAUCCGGAUUUGUGUAACGAUCAUU AAAGCAAUCACAAAUUCGCUUCUAGGGGAGUAUAUAGUGGAUUUAUACACGACG SEQ ID NO:83), tni-mir-10d (MI0004966, GCCGGUGAGGUGCUCGUCGUCUAUACAUACCCUGUAGAACCGAAUGUGUGUGCA GCUGACUUGAUCACAGAUUGGGUUCUAGGGGAGUCUAUGGGCGCUGAAUAAUCA UCGAUGAACGGC SEQ ID NO:84), xtr-mir-10a (MI0004796, GAUUUGCCUGUCCUCUGUAUGUACCCUGUAGAUCCGAAUUUGUGUGAGCGCAAU CAUAUCACAAAUUCGUGUCUGGGGGGAUAUGCAGUUGACACAAACG SEQ ID NO:85), xtr-mir-10b (MI0004797, AACGUUGUCUAUAUGUACCCUGUAGAACCGAAUUUGUGUGGUUCGUACAGUCAC AGAUUCGAUUCUAGGGGGAUAUAUGGUCGAUGCA SEQ ID NO:86), xtr-mir-10c (MI0004798, UAUAUGCACCCUGUAGAAUCGAAUUUGUGUGAGUUCUGAACCACAGAUUCGUCU CUAGGGGGGUAUAUGGG SEQ ID NO:87), or a complement thereof.


In certain aspects, a miR-10 nucleic acid, or a segment or a mimetic thereof, will comprise 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or more nucleotides of the precursor miRNA or its processed sequence, including all ranges and integers there between. In certain embodiments, the miR-10 nucleic acid sequence contains the full-length processed miRNA sequence and is referred to as the “miR-10 full-length processed nucleic acid sequence.” In still further aspects, a miR-10 comprises at least one 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 50 nucleotide (including all ranges and integers there between) segment of miR-10 that is at least 75, 80, 85, 90, 95, 98, 99 or 100% identical to SEQ ID NOs provided herein.


In specific embodiments, a miR-10 or miR-10 inhibitor containing nucleic acid is hsa-miR-10 or hsa-miR-10 inhibitor, or a variation thereof. In a further aspect, a miR-10 nucleic acid or miR-10 inhibitor can be administered with 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more miRNAs or miRNA inhibitors. miRNAs or their complements can be administered concurrently, in sequence, or in an ordered progression. In certain aspects, a miR-10 or miR-10 inhibitor can be administered in combination with one or more of let-7, miR-15, miR-16, miR-20, miR-21, miR-26a, miR-34a, miR-124a, miR-126, miR-143, miR-147, miR-188, miR-200b/c, miR-215, miR-216, miR-292-3p, and/or miR-331. All or combinations of miRNAs or inhibitors thereof may be administered in a single formulation. Administration may be before, during or after a second therapy.


miR-10 nucleic acids or complements thereof may also include various heterologous nucleic acid sequence, i.e., those sequences not typically found operatively coupled with miR-10 in nature, such as promoters, enhancers, and the like. The miR-10 nucleic acid is a recombinant nucleic acid, and can be a ribonucleic acid and/or a deoxyribonucleic acid. The recombinant nucleic acid may comprise a miR-10 or miR-10 inhibitor expression cassette, i.e., a nucleic acid segment that expresses a nucleic acid when introduce into an environment containing components for nucleic acid synthesis. In a further aspect, the expression cassette is comprised in a viral vector, or plasmid DNA vector or other therapeutic nucleic acid vector or delivery vehicle, including liposomes and the like. In a particular aspect, the miR-10 nucleic acid is a synthetic nucleic acid. Moreover, nucleic acids of the invention may be fully or partially synthetic. In certain aspects, viral vectors can be administered at 1×102, 1×103, 1×104 1×105, 1×106, 1×107, 1×108, 1×109, 1×1010, 1×1011, 1×1012, 1×1013, 1×1014 pfu or viral particle (vp).


In a particular aspect, the miR-10 nucleic acid or miR-10 inhibitor is a synthetic nucleic acid. Moreover, nucleic acids of the invention may be fully or partially synthetic. In still further aspects, a DNA encoding such a nucleic acid of the invention can be administered at 0.001, 0.01, 0.1, 1, 10, 20, 30, 40, 50, 100, 200, 400, 600, 800, 1000, 2000, to 4000 μg or mg, including all values and ranges there between. In yet a further aspect, nucleic acids of the invention, including synthetic nucleic acid, can be administered at 0.001, 0.01, 0.1, 1, 10, 20, 30, 40, 50, 100, to 200 μg or mg per kilogram (kg) of body weight. Each of the amounts described herein may be administered over a period of time, including 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, minutes, hours, days, weeks, months or years, including all values and ranges there between.


In certain embodiments, administration of the composition(s) can be enteral or parenteral. In certain aspects, enteral administration is oral. In further aspects, parenteral administration is intralesional, intravascular, intracranial, intrapleural, intratumoral, intraperitoneal, intramuscular, intralymphatic, intraglandular, subcutaneous, topical, intrabronchial, intratracheal, intranasal, inhaled, or instilled. Compositions of the invention may be administered regionally or locally and not necessarily directly into a lesion.


In certain aspects, the gene or genes modulated comprises 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 100, 150, 200 or more genes or combinations of genes identified in Tables 1, 3, 4, and/or 5. In still further aspects, the gene or genes modulated may exclude 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 100, 150, 175 or more genes or combinations of genes identified in Tables 1, 3, 4, and/or 5. Modulation includes modulating transcription, mRNA levels, mRNA translation, and/or protein levels in a cell, tissue, or organ. In certain aspects the expression of a gene or level of a gene product, such as mRNA or encoded protein, is down-regulated or up-regulated. In a particular aspect the gene modulated comprises or is selected from (and may even exclude) 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26. 27, 28, or all of the genes identified in Tables 1, 3, 4, and/or 5, or any combinations thereof. In certain embodiments a gene modulated or selected to be modulated is from Table 1. In further embodiments a gene modulated or selected to be modulated is from Table 3. In still further embodiments a gene modulated or selected to be modulated is from Table 4. In yet further embodiments a gene modulated or selected to be modulated is from Table 5.


Embodiments of the invention may also include obtaining or assessing a gene expression profile or miRNA profile of a target cell prior to selecting the mode of treatment, e.g., administration of a miR-10 nucleic acid, inhibitor of miR-10, or mimetics thereof. The database content related to all nucleic acids and genes designated by an accession number or a database submission are incorporated herein by reference as of the filing date of this application. In certain aspects of the invention one or more miRNA or miRNA inhibitor may modulate a single gene. In a further aspect, one or more genes in one or more genetic, cellular, or physiologic pathways can be modulated by one or more miRNAs or complements thereof, including miR-10 nucleic acids and miR-10 inhibitors in combination with other miRNAs.


miR-10 nucleic acids may also include various heterologous nucleic acid sequence, i.e., those sequences not typically found operatively coupled with miR-10 in nature, such as promoters, enhancers, and the like. The miR-10 nucleic acid is a recombinant nucleic acid, and can be a ribonucleic acid or a deoxyribonucleic acid. The recombinant nucleic acid may comprise a miR-10 expression cassette. In a further aspect, the expression cassette is comprised in a viral, or plasmid DNA vector or other therapeutic nucleic acid vector or delivery vehicle, including liposomes and the like. In a particular aspect, the miR-10 nucleic acid is a synthetic nucleic acid. Moreover, nucleic acids of the invention may be fully or partially synthetic.


A further embodiment of the invention is directed to methods of modulating a cellular pathway comprising administering to the cell an amount of an isolated nucleic acid comprising a miR-10 nucleic acid sequence in an amount sufficient to modulate the expression, function, status, or state of a cellular pathway, in particular those pathways described in Table 2 or the pathways known to include one or more genes from Table 1, 3, 4, and/or 5. Modulation of a cellular pathway includes, but is not limited to modulating the expression of one or more gene. Modulation of a gene can include inhibiting the function of an endogenous miRNA or providing a functional miRNA to a cell, tissue, or subject. Modulation refers to the expression levels or activities of a gene or its related gene product or protein, e.g., the mRNA levels may be modulated or the translation of an mRNA may be modulated, etc. Modulation may increase or up regulate a gene or gene product or it may decrease or down regulate a gene or gene product.


Still a further embodiment includes methods of treating a patient with a pathological condition comprising one or more of step of (a) administering to the patient an amount of an isolated nucleic acid comprising a miR-10 nucleic acid sequence in an amount sufficient to modulate the expression of a cellular pathway; and (b) administering a second therapy, wherein the modulation of the cellular pathway sensitizes the patient to the second therapy. A cellular pathway may include, but is not limited to one or more pathway described in Table 2 below or a pathway that is know to include one or more genes of Tables 1, 3, 4, and/or 5. A second therapy can include administration of a second miRNA or therapeutic nucleic acid, or may include various standard therapies, such as chemotherapy, radiation therapy, drug therapy, immunotherapy, and the like. Embodiments of the invention may also include the determination or assessment of a gene expression profile for the selection of an appropriate therapy.


Embodiments of the invention include methods of treating a subject with a pathological condition comprising one or more of the steps of (a) determining an expression profile of one or more genes selected from Table 1, 3, 4, and/or 5; (b) assessing the sensitivity of the subject to therapy based on the expression profile; (c) selecting a therapy based on the assessed sensitivity; and (d) treating the subject using selected therapy. Typically, the pathological condition will have as a component, indicator, or result the mis-regulation of one or more gene of Table 1, 3, 4, and/or 5.


Further embodiments include the identification and assessment of an expression profile indicative of miR-10 status in a cell or tissue comprising expression assessment of one or more gene from Table 1, 3, 4, and/or 5, or any combination thereof.


The term “miRNA” is used according to its ordinary and plain meaning and refers to a microRNA molecule found in eukaryotes that is involved in RNA-based gene regulation. See, e.g., Carrington et al., 2003, which is hereby incorporated by reference. The term can be used to refer to the single-stranded RNA molecule processed from a precursor or in certain instances the precursor itself.


In some embodiments, it may be useful to know whether a cell expresses a particular miRNA endogenously or whether such expression is affected under particular conditions or when it is in a particular disease state. Thus, in some embodiments of the invention, methods include assaying a cell or a sample containing a cell for the presence of one or more marker gene or mRNA or other analyte indicative of the expression level of a gene of interest. Consequently, in some embodiments, methods include a step of generating an RNA profile for a sample. The term “RNA profile” or “gene expression profile” refers to a set of data regarding the expression pattern for one or more gene or genetic marker in the sample (e.g., a plurality of nucleic acid probes that identify one or more markers from Tables 1, 3, 4, and/or 5); it is contemplated that the nucleic acid profile can be obtained using a set of RNAs, using for example nucleic acid amplification or hybridization techniques well know to one of ordinary skill in the art. The difference in the expression profile in the sample from the patient and a reference expression profile, such as an expression profile from a normal or non-pathologic sample, is indicative of a pathologic, disease, or cancerous condition. A nucleic acid or probe set comprising or identifying a segment of a corresponding mRNA can include all or part of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 100, 200, 500, or more nucleotides, including any integer or range derivable there between, of a gene, genetic marker, a nucleic acid, mRNA or a probe representative thereof that is listed in Tables 1, 3, 4, and/or 5 or identified by the methods described herein.


Certain embodiments of the invention are directed to compositions and methods for assessing, prognosing, or treating a pathological condition in a patient comprising measuring or determining an expression profile of one or more marker(s) in a sample from the patient, wherein a difference in the expression profile in the sample from the patient and an expression profile of a normal sample or reference expression profile is indicative of pathological condition and particularly cancer (e.g., In certain aspects of the invention, the cellular pathway, gene, or genetic marker is or is representative of one or more pathway or marker described in Table 1, 2, 3, 4, and/or 5, including any combination thereof.


Aspects of the invention include diagnosing, assessing, or treating a pathologic condition or preventing a pathologic condition from manifesting. For example, the methods can be used to screen for a pathological condition; assess prognosis of a pathological condition; stage a pathological condition; assess response of a pathological condition to therapy; or to modulate the expression of a gene, genes, or related pathway as a first therapy or to render a subject sensitive or more responsive to a second therapy. In particular aspects, assessing the pathological condition of the patient can be assessing prognosis of the patient. Prognosis may include, but is not limited to an estimation of the time or expected time of survival, assessment of response to a therapy, and the like. In certain aspects, the altered expression of one or more gene or marker is prognostic for a patient having a pathologic condition, wherein the marker is one or more of Table 1, 3, 4, and/or 5, including any combination thereof.


A further embodiment of the invention is directed to methods of modulating a cellular pathway comprising administering to the cell an amount of an isolated nucleic acid comprising a miR-10 nucleic acid sequence or a miR-10 inhibitor. A cell, tissue, or subject may be a cancer cell, a cancerous tissue or harbor cancerous tissue, or a cancer patient. The database content related to all nucleic acids and genes designated by an accession number or a database submission are incorporated herein by reference as of the filing date of this application.


A further embodiment of the invention is directed to methods of modulating a cellular pathway comprising administering to the cell an amount of an isolated nucleic acid comprising a miR-10 nucleic acid sequence in an amount sufficient to modulate the expression, function, status, or state of a cellular pathway, in particular those pathways described in Table 2 or the pathways known to include one or more genes from Table 1, 3, 4, and/or 5. Modulation of a cellular pathway includes, but is not limited to modulating the expression of one or more gene(s). Modulation of a gene can include inhibiting the function of an endogenous miRNA or providing a functional miRNA to a cell, tissue, or subject. Modulation refers to the expression levels or activities of a gene or its related gene product (e.g., mRNA) or protein, e.g., the mRNA levels may be modulated or the translation of an mRNA may be modulated. Modulation may increase or up regulate a gene or gene product or it may decrease or down regulate a gene or gene product (e.g., protein levels or activity).


Still a further embodiment includes methods of administering an miRNA or mimic thereof, and/or treating a subject or patient having, suspected of having, or at risk of developing a pathological condition comprising one or more of step (a) administering to a patient or subject an amount of an isolated nucleic acid comprising a miR-10 nucleic acid sequence or a miR-10 inhibitor in an amount sufficient to modulate expression of a cellular pathway; and (b) administering a second therapy, wherein the modulation of the cellular pathway sensitizes the patient or subject, or increases the efficacy of a second therapy. An increase in efficacy can include a reduction in toxicity, a reduced dosage or duration of the second therapy, or an additive or synergistic effect. A cellular pathway may include, but is not limited to one or more pathway described in Table 2 below or a pathway that is know to include one or more genes of Tables 1, 3, 4, and/or 5. The second therapy may be administered before, during, and/or after the isolated nucleic acid or miRNA or inhibitor is administered


A second therapy can include administration of a second miRNA or therapeutic nucleic acid such as a siRNA or antisense oligonucleotide, or may include various standard therapies, such as pharmaceuticals, chemotherapy, radiation therapy, drug therapy, immunotherapy, and the like. Embodiments of the invention may also include the determination or assessment of gene expression or gene expression profile for the selection of an appropriate therapy. In a particular aspect, a second therapy is a chemotherapy. A chemotherapy can include, but is not limited to paclitaxel, cisplatin, carboplatin, doxorubicin, oxaliplatin, larotaxel, taxol, lapatinib, docetaxel, methotrexate, capecitabine, vinorelbine, cyclophosphamide, gemcitabine, amrubicin, cytarabine, etoposide, camptothecin, dexamethasone, dasatinib, tipifarnib, bevacizumab, sirolimus, temsirolimus, everolimus, lonafarnib, cetuximab, erlotinib, gefitinib, imatinib mesylate, rituximab, trastuzumab, nocodazole, sorafenib, sunitinib, bortezomib, alemtuzumab, gemtuzumab, tositumomab or ibritumomab.


Embodiments of the invention include methods of treating a subject with a disease or condition comprising one or more of the steps of (a) determining an expression profile of one or more genes selected from Table 1, 3, 4, and/or 5; (b) assessing the sensitivity of the subject to therapy based on the expression profile; (c) selecting a therapy based on the assessed sensitivity; and (d) treating the subject using a selected therapy. Typically, the disease or condition will have as a component, indicator, or resulting mis-regulation of one or more gene of Table 1, 3, 4, and/or 5.


In certain aspects, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more miRNA may be used in sequence or in combination; for instance, any combination of miR-10 or a miR-10 inhibitor with another miRNA. Further embodiments include the identification and assessment of an expression profile indicative of miR-10 status in a cell or tissue comprising expression assessment of one or more gene from Table 1, 3, 4, and/or 5, or any combination thereof.


In certain aspects, miR-10 or miR-10 inhibitor and let-7 or let-7 inhibitor can be administered to patients with acute lymphoblastic leukemia, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, chronic lymphoblastic leukemia, chronic myeloid leukemia, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, lung carcinoma, melanoma, multiple myeloma, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma.


Further aspects include administering miR-10 or miR-10 inhibitor and miR-15 or miR-15 inhibitor to patients with astrocytoma, acute myeloid leukemia breast carcinoma, bladder carcinoma, cervical carcinoma, chronic myeloid leukemia, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatoblastoma, hepatocellular carcinoma, Hodgkin lymphoma, lung carcinoma, melanoma, mantle cell lymphoma, multiple myeloma, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, pancreatic carcinoma, prostate carcinoma, renal cell carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma.


In still further aspects, miR-10 or miR-10 inhibitor and miR-16 or miR-16 inhibitor are administered to patients with astrocytoma, breast carcinoma, bladder carcinoma, colorectal carcinoma, endometrial carcinoma, glioblastoma, gastric carcinoma, hepatoblastoma, hepatocellular carcinoma, Hodgkin lymphoma, melanoma, mantle cell lymphoma, multiple myeloma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, pancreatic carcinoma, prostate carcinoma, renal cell carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma.


In certain aspects, miR-10 or miR-10 inhibitor and miR-20 or miR-20 inhibitor are administered to patients with astrocytoma, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, melanoma, mantle cell lymphoma, multiple myeloma, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma.


Aspects of the invention include methods where miR-10 or miR-10 inhibitor and miR-21 or miR-21 inhibitor are administered to patients with astrocytoma, acute lymphoblastic leukemia, acute myeloid leukemia, breast carcinoma, bladder carcinoma, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, melanoma, mantle cell lymphoma, neuroblastoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, pancreatic carcinoma, prostate carcinoma, renal cell carcinoma, squamous cell carcinoma of the head and neck.


In still further aspects, miR-10 or miR-10 inhibitor and miR-26 or miR-26 inhibitor are administered to patients with acute lymphoblastic leukemia, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, chronic lymphoblastic leukemia, chronic myeloid leukemia, colorectal carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, leukemia, lung carcinoma, melanoma, multiple myeloma, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, renal cell carcinoma.


In yet further aspects, miR-10 or miR-10 inhibitor and miR-34 or miR-34 inhibitor are administered to patients with astrocytoma, acute lymphoblastic leukemia, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, chronic lymphoblastic leukemia, chronic myeloid leukemia, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatoblastoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, lung carcinoma, melanoma, mantle cell lymphoma, multiple myeloma, mesothelioma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma.


In yet another aspect, miR-10 or miR-10 inhibitor and miR-124 or miR-124 inhibitor are administered to patients with astrocytoma, acute lymphoblastic leukemia, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, chronic lymphoblastic leukemia, chronic myeloid leukemia, colorectal carcinoma, endometrial carcinoma, Ewing's sarcoma, glioma, glioblastoma, glioblastoma multiforme, gastric carcinoma, hepatoblastoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, lung carcinoma, liposarcoma, melanoma, mantle cell lymphoma, multiple myeloma, neuroblastoma, non-Hodgkin lymphoma, nasopharyngeal carcinoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, retinoblastoma, renal cell carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma, Wilm's tumor.


In yet further aspects, miR-10 or miR-10 inhibitor and miR-126 or miR-126 inhibitor are administered to patients with astrocytoma, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, Ewing's sarcoma, glioma, glioblastoma, gastric carcinoma, hepatoblastoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, lung carcinoma, melanoma, mantle cell lymphoma, mesothelioma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, renal cell carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma.


In a further aspect, miR-10 or miR-10 inhibitor and miR-143 or miR-143 inhibitor are administered to patients with astrocytoma, acute lymphoblastic leukemia, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, chronic lymphoblastic leukemia, chronic myeloid leukemia, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, lung carcinoma, melanoma, mantle cell lymphoma, multiple myeloma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, renal cell carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma.


In still a further aspect, miR-10 or miR-10 inhibitor and miR-147 or miR-147 inhibitor are administered to patients with astrocytoma, breast carcinoma, bladder carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, melanoma, mantle cell lymphoma, multiple myeloma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, renal cell carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma.


In yet another aspect, miR-10 or miR-10 inhibitor and miR-188 or miR-188 inhibitor are administered to patients with astrocytoma, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, chronic lymphoblastic leukemia, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, leukemia, lung carcinoma, melanoma, multiple myeloma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, pancreatic carcinoma, prostate carcinoma, renal cell carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma.


In yet a further aspect, miR-10 or miR-10 inhibitor and miR-200 or miR-200 inhibitor are administered to patients with breast carcinoma, cervical carcinoma, chronic lymphoblastic leukemia, colorectal carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, leukemia, lung carcinoma, multiple myeloma, mesothelioma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma.


In other aspects, miR-10 or miR-10 inhibitor and miR-215 or miR-215 inhibitor are administered to patients with astrocytoma, acute lymphoblastic leukemia, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, chronic lymphoblastic leukemia, chronic myeloid leukemia, colorectal carcinoma, endometrial carcinoma, Ewing's sarcoma, glioma, glioblastoma, gastric carcinoma, hepatoblastoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, lung carcinoma, liposarcoma, melanoma, mantle cell lymphoma, multiple myeloma, mesothelioma, neuroblastoma, non-Hodgkin lymphoma, nasopharyngeal carcinoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, retinoblastoma, renal cell carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma, Wilm's tumor.


In certain aspects, miR-10 or miR-10 inhibitor and miR-216 or miR-216 inhibitor are administered to patients with astrocytoma, breast carcinoma, cervical carcinoma, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, lung carcinoma, non-Hodgkin lymphoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, prostate carcinoma, squamous cell carcinoma of the head and neck.


In a further aspect, miR-10 or miR-10 inhibitor and miR-292-3p or miR-292-3p inhibitor are administered to patients with astrocytoma, acute lymphoblastic leukemia, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, chronic myeloid leukemia, colorectal carcinoma, endometrial carcinoma, Ewing's sarcoma, fibrosarcoma, glioma, glioblastoma, gastric carcinoma, hepatoblastoma, hepatocellular carcinoma, leukemia, lung carcinoma, liposarcoma, melanoma, multiple myeloma, neuroblastoma, non-Hodgkin lymphoma, nasopharyngeal carcinoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, renal cell carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma, Wilm's tumor.


In still a further aspect, miR-10 or miR-10 inhibitor and miR-331 or miR-331 inhibitor are administered to patients with astrocytoma, acute lymphoblastic leukemia, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, chronic lymphoblastic leukemia, colorectal carcinoma, endometrial carcinoma, glioma, glioblastoma, gastric carcinoma, hepatocellular carcinoma, leukemia, lung carcinoma, melanoma, multiple myeloma, neuroblastoma, non-Hodgkin lymphoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, renal cell carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma.


It is contemplated that when miR-10 or a miR-10 inhibitor is given in combination with one or more other miRNA molecules, the two different miRNAs or inhibitors may be given at the same time or sequentially. In some embodiments, therapy proceeds with one miRNA or inhibitor and that therapy is followed up with therapy with the other miRNA or inhibitor 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55 minutes, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 hours, 1, 2, 3, 4, 5, 6, 7 days, 1, 2, 3, 4, 5 weeks, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months or any such combination later.


The term “miRNA” is used according to its ordinary and plain meaning and refers to a microRNA molecule found in eukaryotes that is involved in RNA-based gene regulation. See, e.g., Carrington et al., 2003, which is hereby incorporated by reference. The term can be used to refer to the single-stranded RNA molecule processed from a precursor or in certain instances the precursor itself.


In some embodiments, it may be useful to know whether a cell expresses a particular miRNA endogenously or whether such expression is affected under particular conditions or when it is in a particular disease state. Thus, in some embodiments of the invention, methods include assaying a cell or a sample containing a cell for the presence of one or more marker gene or mRNA or other analyte indicative of the expression level of a gene of interest. Consequently, in some embodiments, methods include a step of generating an RNA profile for a sample. The term “RNA profile” or “gene expression profile” refers to a set of data regarding the expression pattern for one or more gene or genetic marker or miRNA in the sample (e.g., a plurality of nucleic acid probes that identify one or more markers from Tables 1, 3, 4, and/or 5); it is contemplated that the nucleic acid profile can be obtained using a set of RNAs, using for example nucleic acid amplification or hybridization techniques well know to one of ordinary skill in the art. The difference in the expression profile in the sample from the patient and a reference expression profile, such as an expression profile of one or more genes or miRNAs, are indicative of which miRNAs to be administered.


Further embodiments include the identification and assessment of an expression profile indicative of miR-10 status in a cell or tissue comprising expression assessment of one or more gene from Table 1, 3, 4, and/or 5, or any combination thereof.


The term “miRNA” is used according to its ordinary and plain meaning and refers to a microRNA molecule found in eukaryotes that is involved in RNA-based gene regulation. See, e.g., Carrington et al., 2003, which is hereby incorporated by reference. The term can be used to refer to the single-stranded RNA molecule processed from a precursor or in certain instances the precursor itself or a mimetic thereof.


In some embodiments, it may be useful to know whether a cell expresses a particular miRNA endogenously or whether such expression is affected under particular conditions or when it is in a particular disease state. Thus, in some embodiments of the invention, methods include assaying a cell or a sample containing a cell for the presence of one or more miRNA marker gene or mRNA or other analyte indicative of the expression level of a gene of interest. Consequently, in some embodiments, methods include a step of generating an RNA profile for a sample. The term “RNA profile” or “gene expression profile” refers to a set of data regarding the expression pattern for one or more gene or genetic marker in the sample (e.g., a plurality of nucleic acid probes that identify one or more markers or genes from Tables 1, 3, 4, and/or 5); it is contemplated that the nucleic acid profile can be obtained using a set of RNAs, using for example nucleic acid amplification or hybridization techniques well know to one of ordinary skill in the art. The difference in the expression profile in the sample from a patient and a reference expression profile, such as an expression profile from a normal or non-pathologic sample, or a digitized reference, is indicative of a pathologic, disease, or cancerous condition. In certain aspects the expression profile is an indicator of a propensity to or probability of (i.e., risk factor for a disease or condition) developing such a condition(s). Such a risk or propensity may indicate a treatment, increased monitoring, prophylactic measures, and the like. A nucleic acid or probe set may comprise or identify a segment of a corresponding mRNA and may include all or part of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 100, 200, 500, or more segments, including any integer or range derivable there between, of a gene or genetic marker, or a nucleic acid, mRNA or a probe representative thereof that is listed in Tables 1, 3, 4, and/or 5 or identified by the methods described herein.


Certain embodiments of the invention are directed to compositions and methods for assessing, prognosing, or treating a pathological condition in a patient comprising measuring or determining an expression profile of one or more miRNA or marker(s) in a sample from the patient, wherein a difference in the expression profile in the sample from the patient and an expression profile of a normal sample or reference expression profile is indicative of pathological condition and particularly cancer (e.g., In certain aspects of the invention, the miRNAs, cellular pathway, gene, or genetic marker is or is representative of one or more pathway or marker described in Table 1, 2, 3, 4, and/or 5, including any combination thereof.


Aspects of the invention include diagnosing, assessing, or treating a pathologic condition or preventing a pathologic condition from manifesting. For example, the methods can be used to screen for a pathological condition; assess prognosis of a pathological condition; stage a pathological condition; assess response of a pathological condition to therapy; or to modulate the expression of a gene, genes, or related pathway as a first therapy or to render a subject sensitive or more responsive to a second therapy. In particular aspects, assessing the pathological condition of the patient can be assessing prognosis of the patient. Prognosis may include, but is not limited to an estimation of the time or expected time of survival, assessment of response to a therapy, and the like. In certain aspects, the altered expression of one or more gene or marker is prognostic for a patient having a pathologic condition, wherein the marker is one or more of Table 1, 3, 4, and/or 5, including any combination thereof.


Predicted gene targets are shown in Table 3. Target genes whose mRNA expression levels are affected by hsa-miR-10 represent particularly useful candidates for cancer therapy and therapy of other diseases or conditions through manipulation of their expression levels.


Certain embodiments of the invention include determining expression of one or more marker, gene, or nucleic acid segment representative of one or more genes, by using an amplification assay, a hybridization assay, or protein assay, a variety of which are well known to one of ordinary skill in the art. In certain aspects, an amplification assay can be a quantitative amplification assay, such as quantitative RT-PCR or the like. In still further aspects, a hybridization assay can include array hybridization assays or solution hybridization assays. The nucleic acids from a sample may be labeled from the sample and/or hybridizing the labeled nucleic acid to one or more nucleic acid probes. Nucleic acids, mRNA, and/or nucleic acid probes may be coupled to a support. Such supports are well known to those of ordinary skill in the art and include, but are not limited to glass, plastic, metal, or latex. In particular aspects of the invention, the support can be planar or in the form of a bead or other geometric shapes or configurations known in the art. Proteins are typically assayed by immunoblotting, chromatography, or mass spectrometry or other methods known to those of ordinary skill in the art.


The present invention also concerns kits containing compositions of the invention or compositions to implement methods of the invention. In some embodiments, kits can be used to evaluate one or more marker molecules, and/or express one or more miRNA or miRNA inhibitor. In certain embodiments, a kit contains, contains at least or contains at most 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 100, 150, 200 or more probes, recombinant nucleic acid, or synthetic nucleic acid molecules related to the markers to be assessed or an miRNA or miRNA inhibitor to be expressed or modulated, and may include any range or combination derivable therein. Kits may comprise components, which may be individually packaged or placed in a container, such as a tube, bottle, vial, syringe, or other suitable container means. Individual components may also be provided in a kit in concentrated amounts; in some embodiments, a component is provided individually in the same concentration as it would be in a solution with other components. Concentrations of components may be provided as 1×, 2×, 5×, 10×, or 20× or more. Kits for using probes, synthetic nucleic acids, recombinant nucleic acids, or non-synthetic nucleic acids of the invention for therapeutic, prognostic, or diagnostic applications are included as part of the invention. Specifically contemplated are any such molecules corresponding to any miRNA reported to influence biological activity or expression of one or more marker gene or gene pathway described herein. In certain aspects, negative and/or positive controls are included in some kit embodiments. The control molecules can be used to verify transfection efficiency and/or control for transfection-induced changes in cells.


Certain embodiments are directed to a kit for assessment of a pathological condition or the risk of developing a pathological condition in a patient by nucleic acid profiling of a sample comprising, in suitable container means, two or more nucleic acid hybridization or amplification reagents. The kit can comprise reagents for labeling nucleic acids in a sample and/or nucleic acid hybridization reagents. The hybridization reagents typically comprise hybridization probes. Amplification reagents include, but are not limited to amplification primers, reagents, and enzymes.


In some embodiments of the invention, an expression profile is generated by steps that include: (a) labeling nucleic acid in the sample; (b) hybridizing the nucleic acid to a number of probes, or amplifying a number of nucleic acids, and (c) determining and/or quantitating nucleic acid hybridization to the probes or detecting and quantitating amplification products, wherein an expression profile is generated. See U.S. Provisional Patent Application 60/575,743 and the U.S. Provisional Patent Application 60/649,584, and U.S. patent application Ser. No. 11/141,707 and U.S. patent application Ser. No. 11/273,640, all of which are hereby incorporated by reference.


Methods of the invention involve diagnosing and/or assessing the prognosis of a patient based on a miRNA and/or a marker nucleic acid expression profile. In certain embodiments, the elevation or reduction in the level of expression of a particular gene or genetic pathway or set of nucleic acids in a cell is correlated with a disease state or pathological condition compared to the expression level of the same in a normal or non-pathologic cell or tissue sample. This correlation allows for diagnostic and/or prognostic methods to be carried out when the expression level of one or more nucleic acid is measured in a biological sample being assessed and then compared to the expression level of a normal or non-pathologic cell or tissue sample. It is specifically contemplated that expression profiles for patients, particularly those suspected of having or having a propensity for a particular disease or condition such as cancer, can be generated by evaluating any of or sets of the miRNAs and/or nucleic acids discussed in this application. The expression profile that is generated from the patient will be one that provides information regarding the particular disease or condition. In many embodiments, the profile is generated using nucleic acid hybridization or amplification, (e.g., array hybridization or RT-PCR). In certain aspects, an expression profile can be used in conjunction with other diagnostic and/or prognostic tests, such as histology, protein profiles in the serum and/or cytogenetic assessment.


The methods can further comprise one or more of the steps including: (a) obtaining a sample from the patient, (b) isolating nucleic acids from the sample, (c) labeling the nucleic acids isolated from the sample, and (d) hybridizing the labeled nucleic acids to one or more probes. Nucleic acids of the invention include one or more nucleic acid comprising at least one segment having a sequence or complementary sequence of to a nucleic acid representative of one or more of genes or markers in Table 1, 3, 4, and/or 5.


It is contemplated that any method or composition described herein can be implemented with respect to any other method or composition described herein and that different embodiments may be combined. It is specifically contemplated that any methods and compositions discussed herein with respect to miRNA molecules, miRNA, genes, and Certain embodiments of the invention include determining expression of one or more marker, gene, or nucleic acid representative thereof, by using an amplification assay, a hybridization assay, or protein assay, a variety of which are well known to one of ordinary skill in the art. In certain aspects, an amplification assay can be a quantitative amplification assay, such as quantitative RT-PCR or the like. In still further aspects, a hybridization assay can include array hybridization assays or solution hybridization assays. The nucleic acids from a sample may be labeled from the sample and/or hybridizing the labeled nucleic acid to one or more nucleic acid probes. Nucleic acids, mRNA, and/or nucleic acid probes may be coupled to a support. Such supports are well known to those of ordinary skill in the art and include, but are not limited to glass, plastic, metal, or latex. In particular aspects of the invention, the support can be planar or in the form of a bead or other geometric shapes or configurations known in the art. Protein are typically assayed by immunoblotting, chromatography, or mass spectrometry or other methods known to those of ordinary skill in the art.


The present invention also concerns kits containing compositions of the invention or compositions to implement methods of the invention. In some embodiments, kits can be used to evaluate one or more marker molecules, and/or express one or more miRNA. In certain embodiments, a kit contains, contains at least or contains at most 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 100, 150, 200 or more probes, recombinant nucleic acid, or synthetic nucleic acid molecules related to the markers to be assessed or an miRNA to be expressed or modulated, and may include any range or combination derivable therein. Kits may comprise components, which may be individually packaged or placed in a container, such as a tube, bottle, vial, syringe, or other suitable container means. Individual components may also be provided in a kit in concentrated amounts; in some embodiments, a component is provided individually in the same concentration as it would be in a solution with other components. Concentrations of components may be provided as 1×, 2×, 5×, 10×, or 20× or more. Kits for using probes, synthetic nucleic acids, recombinant nucleic acids, or non-synthetic nucleic acids of the invention for therapeutic, prognostic, or diagnostic applications are included as part of the invention. Specifically contemplated are any such molecules corresponding to any miRNA reported to influence biological activity or expression of one or more marker gene or gene pathway described herein. In certain aspects, negative and/or positive controls are included in some kit embodiments. The control molecules can be used to verify transfection efficiency and/or control for transfection-induced changes in cells.


Certain embodiments are directed to a kit for assessment of a pathological condition or the risk of developing a pathological condition in a patient by nucleic acid profiling of a sample comprising, in suitable container means, two or more nucleic acid hybridization or amplification reagents. The kit can comprise reagents for labeling nucleic acids in a sample and/or nucleic acid hybridization reagents. The hybridization reagents typically comprise hybridization probes. Amplification reagents include, but are not limited to amplification primers, reagents, and enzymes.


In some embodiments of the invention, an expression profile is generated by steps that include: (a) labeling nucleic acid in the sample; (b) hybridizing the nucleic acid to a number of probes, or amplifying a number of nucleic acids, and (c) determining and/or quantitating nucleic acid hybridization to the probes or detecting and quantitating amplification products, wherein an expression profile is generated. See U.S. Provisional Patent Application 60/575,743 and the U.S. Provisional Patent Application 60/649,584, and U.S. patent application Ser. No. 11/141,707 and U.S. patent application Ser. No. 11/273,640, all of which are hereby incorporated by reference.


Methods of the invention involve diagnosing and/or assessing the prognosis of a patient based on a miRNA and/or a marker nucleic acid expression profile. In certain embodiments, the elevation or reduction in the level of expression of a particular gene or genetic pathway or set of nucleic acids in a cell is correlated with a disease state or pathological condition compared to the expression level of the same in a normal or non-pathologic cell or tissue sample. This correlation allows for diagnostic and/or prognostic methods to be carried out when the expression level of one or more nucleic acid is measured in a biological sample being assessed and then compared to the expression level of a normal or non-pathologic cell or tissue sample. It is specifically contemplated that expression profiles for patients, particularly those suspected of having or having a propensity for a particular disease or condition such as cancer, can be generated by evaluating any of or sets of the miRNAs and/or nucleic acids discussed in this application. The expression profile that is generated from the patient will be one that provides information regarding the particular disease or condition. In many embodiments, the profile is generated using nucleic acid hybridization or amplification, (e.g., array hybridization or RT-PCR). In certain aspects, an expression profile can be used in conjunction with other diagnostic and/or prognostic tests, such as histology, protein profiles in the serum and/or cytogenetic assessment.


The methods can further comprise one or more of the steps including: (a) obtaining a sample from the patient, (b) isolating nucleic acids from the sample, (c) labeling the nucleic acids isolated from the sample, and (d) hybridizing the labeled nucleic acids to one or more probes. Nucleic acids of the invention include one or more nucleic acid comprising at least one segment having a sequence or complementary sequence of to a nucleic acid representative of one or more of genes or markers in Table 1, 3, 4, and/or 5.


It is contemplated that any method or composition described herein can be implemented with respect to any other method or composition described herein and that different embodiments may be combined. It is specifically contemplated that any methods and compositions discussed herein with respect to miRNA molecules, miRNA, genes and nucleic acids representative of genes may be implemented with respect to synthetic nucleic acids. In some embodiments the synthetic nucleic acid is exposed to the proper conditions to allow it to become a processed or mature nucleic acid, such as a miRNA under physiological circumstances. The claims originally filed are contemplated to cover claims that are multiply dependent on any filed claim or combination of filed claims.


Also, any embodiment of the invention involving specific genes (including representative fragments there of), mRNA, or miRNAs by name is contemplated also to cover embodiments involving miRNAs whose sequences are at least 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99% identical to the mature sequence of the specified miRNA.


It will be further understood that shorthand notations are employed such that a generic description of a gene or marker thereof, or of a miRNA refers to any of its gene family members (distinguished by a number) or representative fragments thereof, unless otherwise indicated. It is understood by those of skill in the art that a “gene family” refers to a group of genes having the same coding sequence or miRNA coding sequence. Typically, miRNA members of a gene family are identified by a number following the initial designation. For example, miR-16-1 and miR-16-2 are members of the miR-16 gene family and “mir-7” refers to miR-7-1, miR-7-2 and miR-7-3. Moreover, unless otherwise indicated, a shorthand notation refers to related miRNAs (distinguished by a letter). Exceptions to these shorthand notations will be otherwise identified.


Other embodiments of the invention are discussed throughout this application. Any embodiment discussed with respect to one aspect of the invention applies to other aspects of the invention as well and vice versa. The embodiments in the Example and Detailed Description section are understood to be embodiments of the invention that are applicable to all aspects of the invention.


The terms “inhibiting,” “reducing,” or “prevention,” or any variation of these terms, when used in the claims and/or the specification includes any measurable decrease or complete inhibition to achieve a desired result.


The use of the word “a” or “an” when used in conjunction with the term “comprising” in the claims and/or the specification may mean “one,” but it is also consistent with the meaning of “one or more,” “at least one,” and “one or more than one.”


Throughout this application, the term “about” is used to indicate that a value includes the standard deviation of error for the device or method being employed to determine the value.


The use of the term “or” in the claims is used to mean “and/or” unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and “and/or.”


As used in this specification and claim(s), the words “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”) or “containing” (and any form of containing, such as “contains” and “contain”) are inclusive or open-ended and do not exclude additional, unrecited elements or method steps.


Other objects, features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.







DETAILED DESCRIPTION OF THE INVENTION

The present invention is directed to compositions and methods relating to the identification and characterization of genes and biological pathways related to these genes as represented by the expression of the identified genes, as well as use of miRNAs related to such, for therapeutic, prognostic, and diagnostic applications, particularly those methods and compositions related to assessing and/or identifying pathological conditions directly or indirectly related to miR-10 expression or the aberrant expression thereof.


In certain aspects, the invention is directed to methods for the assessment, analysis, and/or therapy of a cell or subject where certain genes have a reduced or increased expression (relative to normal) as a result of an increased or decreased expression of any one or a combination of miR-10 family members (including, but not limited to SEQ ID NO:1 to SEQ ID NO:87) and/or genes with an increased expression (relative to normal) as a result of an increased or decreased expression of one or a combination of miR-10 family members. The expression profile and/or response to miR-10 expression or inhibition may be indicative of a disease or an individual with a condition, e.g., cancer.


Prognostic assays featuring any one or combination of the miRNAs listed or the markers listed (including nucleic acids representative thereof) could be used in assessment of a patient to determine what if any treatment regimen is justified. As with the diagnostic assays mentioned above, the absolute values that define low expression will depend on the platform used to measure the miRNA(s). The same methods described for the diagnostic assays could be used for prognostic assays.


I. THERAPEUTIC METHODS

Embodiments of the invention concern nucleic acids that perform the activities of or inhibit endogenous miRNAs when introduced into cells. In certain aspects, nucleic acids are synthetic or non-synthetic miRNA. Sequence-specific miRNA inhibitors can be used to inhibit sequentially or in combination the activities of one or more endogenous miRNAs in cells, as well those genes and associated pathways modulated by the endogenous miRNA.


The present invention concerns, in some embodiments, short nucleic acid molecules that function as miRNAs or as inhibitors of miRNA in a cell. The term “short” refers to a length of a single polynucleotide that is 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 50, 100, or 150 nucleotides or fewer, including all integers or ranges derivable there between. The nucleic acid molecules are typically synthetic. The term “synthetic” refers to nucleic acid molecule that is isolated and not produced naturally in a cell. In certain aspects the sequence (the entire sequence) and/or chemical structure deviates from a naturally-occurring nucleic acid molecule, such as an endogenous precursor miRNA or miRNA molecule or complement thereof. While in some embodiments, nucleic acids of the invention do not have an entire sequence that is identical or complementary to a sequence of a naturally-occurring nucleic acid, such molecules may encompass all or part of a naturally-occurring sequence or a complement thereof. It is contemplated, however, that a synthetic nucleic acid administered to a cell may subsequently be modified or altered in the cell such that its structure or sequence is the same as non-synthetic or naturally occurring nucleic acid, such as a mature miRNA sequence. For example, a synthetic nucleic acid may have a sequence that differs from the sequence of a precursor miRNA, but that sequence may be altered once in a cell to be the same as an endogenous, processed miRNA or an inhibitor thereof. The term “isolated” means that the nucleic acid molecules of the invention are initially separated from different (in terms of sequence or structure) and unwanted nucleic acid molecules such that a population of isolated nucleic acids is at least about 90% homogenous, and may be at least about 95, 96, 97, 98, 99, or 100% homogenous with respect to other polynucleotide molecules. In many embodiments of the invention, a nucleic acid is isolated by virtue of it having been synthesized in vitro separate from endogenous nucleic acids in a cell. It will be understood, however, that isolated nucleic acids may be subsequently mixed or pooled together. In certain aspects, synthetic miRNA of the invention are RNA or RNA analogs. miRNA inhibitors may be DNA or RNA, or analogs thereof. miRNA and miRNA inhibitors of the invention are collectively referred to as “synthetic nucleic acids.”


In some embodiments, there is a miRNA or a synthetic miRNA having a length of between 17 and 130 residues. The present invention concerns miRNA or synthetic miRNA molecules that are, are at least, or are at most 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 140, 145, 150, 160, 170, 180, 190, 200 or more residues in length, including any integer or any range there between.


In certain embodiments, synthetic miRNA have (a) a “miRNA region” whose sequence or binding region from 5′ to 3′ is identical or complementary to all or a segment of a mature miRNA sequence, and (b) a “complementary region” whose sequence from 5′ to 3′ is between 60% and 100% complementary to the miRNA sequence in (a). In certain embodiments, these synthetic miRNA are also isolated, as defined above. The term “miRNA region” refers to a region on the synthetic miRNA that is at least 75, 80, 85, 90, 95, or 100% identical, including all integers there between, to the entire sequence of a mature, naturally occurring miRNA sequence or a complement thereof. In certain embodiments, the miRNA region is or is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9 or 100% identical to the sequence of a naturally-occurring miRNA or complement thereof.


The term “complementary region” or “complement” refers to a region of a nucleic acid or mimetic that is or is at least 60% complementary to the mature, naturally occurring miRNA sequence. The complementary region is or is at least 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9 or 100% complementary, or any range derivable therein. With single polynucleotide sequences, there may be a hairpin loop structure as a result of chemical bonding between the miRNA region and the complementary region. In other embodiments, the complementary region is on a different nucleic acid molecule than the miRNA region, in which case the complementary region is on the complementary strand and the miRNA region is on the active strand.


In other embodiments of the invention, there are synthetic nucleic acids that are miRNA inhibitors. A miRNA inhibitor is between about 17 to 25 nucleotides in length and comprises a 5′ to 3′ sequence that is at least 90% complementary to the 5′ to 3′ sequence of a mature miRNA. In certain embodiments, a miRNA inhibitor molecule is 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleotides in length, or any range derivable therein. Moreover, an miRNA inhibitor may have a sequence (from 5′ to 3′) that is or is at least 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, 99.9 or 100% complementary, or any range derivable therein, to the 5′ to 3′ sequence of a mature miRNA, particularly a mature, naturally occurring miRNA. One of skill in the art could use a portion of the miRNA sequence that is complementary to the sequence of a mature miRNA as the sequence for a miRNA inhibitor. Moreover, that portion of the nucleic acid sequence can be altered so that it is still comprises the appropriate percentage of complementarity to the sequence of a mature miRNA.


In certain embodiments, a synthetic miRNA has a nucleotide at its 5′ end of the complementary region in which the phosphate and/or hydroxyl group has been replaced with another chemical group (referred to as the “replacement design”). In some cases, the phosphate group is replaced, while in others, the hydroxyl group has been replaced. In particular embodiments, the replacement group is biotin, an amine group, a lower alkylamine group, an aminohexyl phosphate group, an acetyl group, 2′O-Me (2′oxygen-methyl), DMTO (4,4′-dimethoxytrityl with oxygen), fluoroscein, a thiol, or acridine, though other replacement groups are well known to those of skill in the art and can be used as well. This design element can also be used with a miRNA inhibitor.


In certain embodiments, a synthetic miRNA has a nucleotide at its 5′ end of the complementary region in which the phosphate and/or hydroxyl group has been replaced with another chemical group (referred to as the “replacement design”). In some cases, the phosphate group is replaced, while in others, the hydroxyl group has been replaced. In particular embodiments, the replacement group is biotin, an amine group, a lower alkylamine group, an acetyl group, 2′O-Me (2′oxygen-methyl), DMTO (4,4′-dimethoxytrityl with oxygen), fluoroscein, a thiol, or acridine, though other replacement groups are well known to those of skill in the art and can be used as well. This design element can also be used with a miRNA inhibitor.


Additional embodiments concern a synthetic miRNA having one or more sugar modifications in the first or last 1 to 6 residues of the complementary region (referred to as the “sugar replacement design”). In certain cases, there is one or more sugar modifications in the first 1, 2, 3, 4, 5, 6 or more residues of the complementary region, or any range derivable therein. In additional cases, there is one or more sugar modifications in the last 1, 2, 3, 4, 5, 6 or more residues of the complementary region, or any range derivable therein, have a sugar modification. It will be understood that the terms “first” and “last” are with respect to the order of residues from the 5′ end to the 3′ end of the region. In particular embodiments, the sugar modification is a 2′O-Me modification, a 2° F. modification, a 2′H modification, a 2′amino modification, a 4′thioribose modification or a phosphorothioate modification on the carboxy group linked to the carbon at position 6′. In further embodiments, there is one or more sugar modifications in the first or last 2 to 4 residues of the complementary region or the first or last 4 to 6 residues of the complementary region. This design element can also be used with an miRNA inhibitor. Thus, an miRNA inhibitor can have this design element and/or a replacement group on the nucleotide at the 5′ terminus, as discussed above.


In other embodiments of the invention, there is a synthetic miRNA or inhibitor in which one or more nucleotides in the last 1 to 5 residues at the 3′ end of the complementary region are not complementary to the corresponding nucleotides of the miRNA region (“noncomplementarity”) (referred to as the “noncomplementarity design”). The noncomplementarity may be in the last 1, 2, 3, 4, and/or 5 residues of the complementary miRNA. In certain embodiments, there is noncomplementarity with at least 2 nucleotides in the complementary region.


It is contemplated that synthetic miRNA of the invention have one or more of the replacement, sugar modification, or noncomplementarity designs. In certain cases, synthetic RNA molecules have two of them, while in others these molecules have all three designs in place.


The miRNA region and the complementary region may be on the same or separate polynucleotides. In cases in which they are contained on or in the same polynucleotide, the miRNA molecule will be considered a single polynucleotide. In embodiments in which the different regions are on separate polynucleotides, the synthetic miRNA will be considered to be comprised of two polynucleotides.


When the RNA molecule is a single polynucleotide, there can be a linker region between the miRNA region and the complementary region. In some embodiments, the single polynucleotide is capable of forming a hairpin loop structure as a result of bonding between the miRNA region and the complementary region. The linker constitutes the hairpin loop. It is contemplated that in some embodiments, the linker region is, is at least, or is at most 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 residues in length, or any range derivable therein. In certain embodiments, the linker is between 3 and 30 residues (inclusive) in length.


In addition to having a miRNA or inhibitor region and a complementary region, there may be flanking sequences as well at either the 5′ or 3′ end of the region. In some embodiments, there is or is at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 nucleotides or more, or any range derivable therein, flanking one or both sides of these regions.


Methods of the invention include reducing or eliminating activity of one or more miRNAs in a cell comprising introducing into a cell a miRNA inhibitor (which may be described generally herein as an miRNA, so that a description of miRNA, where appropriate, also will refer to a miRNA inhibitor); or supplying or enhancing the activity of one or more miRNAs in a cell. The present invention also concerns inducing certain cellular characteristics by providing to a cell a particular nucleic acid, such as a specific synthetic miRNA molecule or a synthetic miRNA inhibitor molecule. However, in methods of the invention, the miRNA molecule or miRNA inhibitor need not be synthetic. They may have a sequence that is identical to a naturally occurring miRNA or they may not have any design modifications. In certain embodiments, the miRNA molecule and/or the miRNA inhibitor are synthetic, as discussed above.


The particular nucleic acid molecule provided to the cell is understood to correspond to a particular miRNA in the cell, and thus, the miRNA in the cell is referred to as the “corresponding miRNA.” In situations in which a named miRNA molecule is introduced into a cell, the corresponding miRNA will be understood to be the induced or inhibited miRNA function. It is contemplated, however, that the miRNA molecule introduced into a cell is not a mature miRNA but is capable of becoming or functioning as a mature miRNA under the appropriate physiological conditions. In cases in which a particular corresponding miRNA is being inhibited by a miRNA inhibitor, the particular miRNA will be referred to as the “targeted miRNA.” It is contemplated that multiple corresponding miRNAs may be involved. In particular embodiments, more than one miRNA molecule is introduced into a cell. Moreover, in other embodiments, more than one miRNA inhibitor is introduced into a cell. Furthermore, a combination of miRNA molecule(s) and miRNA inhibitor(s) may be introduced into a cell. The inventors contemplate that a combination of miRNA may act at one or more points in cellular pathways of cells with aberrant phenotypes and that such combination may have increased efficacy on the target cell while not adversely effecting normal cells. Thus, a combination of miRNA may have a minimal adverse effect on a subject or patient while supplying a sufficient therapeutic effect, such as amelioration of a condition, growth inhibition of a cell, death of a targeted cell, alteration of cell phenotype or physiology, slowing of cellular growth, sensitization to a second therapy, sensitization to a particular therapy, and the like.


Methods include identifying a cell or patient in need of inducing those cellular characteristics. Also, it will be understood that an amount of a synthetic nucleic acid that is provided to a cell or organism is an “effective amount,” which refers to an amount needed (or a sufficient amount) to achieve a desired goal, such as inducing a particular cellular characteristic(s).


In certain embodiments of the methods include providing or introducing to a cell a nucleic acid molecule corresponding to a mature miRNA in the cell in an amount effective to achieve a desired physiological result.


Moreover, methods can involve providing synthetic or nonsynthetic miRNA molecules. It is contemplated that in these embodiments, that methods may or may not be limited to providing only one or more synthetic miRNA molecules or only one or more nonsynthetic miRNA molecules. Thus, in certain embodiments, methods may involve providing both synthetic and nonsynthetic miRNA molecules. In this situation, a cell or cells are most likely provided a synthetic miRNA molecule corresponding to a particular miRNA and a nonsynthetic miRNA molecule corresponding to a different miRNA. Furthermore, any method articulated using a list of miRNAs using Markush group language may be articulated without the Markush group language and a disjunctive article (i.e., or) instead, and vice versa.


In some embodiments, there is a method for reducing or inhibiting cell proliferation in a cell comprising introducing into or providing to the cell an effective amount of (i) an miRNA inhibitor molecule or (ii) a synthetic or nonsynthetic miRNA molecule that corresponds to a miRNA sequence. In certain embodiments the methods involves introducing into the cell an effective amount of (i) a miRNA inhibitor molecule having a 5′ to 3′ sequence that is at least 90% complementary to the 5′ to 3′ sequence of one or more mature miRNA.


Certain embodiments of the invention include methods of treating a pathologic condition, in particular cancer, e.g., lung or liver cancer. In one aspect, the method comprises contacting a target cell with one or more nucleic acid, synthetic miRNA, or miRNA comprising at least one nucleic acid segment having all or a portion of a miRNA sequence. The segment may be 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30 or more nucleotides or nucleotide analog, including all integers there between. An aspect of the invention includes the modulation of gene expression, miRNA expression or function or mRNA expression or function within a target cell, such as a cancer cell.


Typically, an endogenous gene, miRNA or mRNA is modulated in the cell. In particular embodiments, the nucleic acid sequence comprises at least one segment that is at least 70, 75, 80, 85, 90, 95, or 100% identical in nucleic acid sequence to one or more miRNA or gene sequence. Modulation of the expression or processing of an endogenous gene, miRNA, or mRNA can be through modulation of the processing of a mRNA, such processing including transcription, transportation and/or translation with in a cell. Modulation may also be effected by the inhibition or enhancement of miRNA activity with a cell, tissue, or organ. Such processing may affect the expression of an encoded product or the stability of the mRNA. In still other embodiments, a nucleic acid sequence can comprise a modified nucleic acid sequence. In certain aspects, one or more miRNA sequence may include or comprise a modified nucleobase or nucleic acid sequence.


It will be understood in methods of the invention that a cell or other biological matter such as an organism (including patients) can be provided a miRNA or miRNA molecule corresponding to a particular miRNA by administering to the cell or organism a nucleic acid molecule that functions as the corresponding miRNA once inside the cell. The form of the molecule provided to the cell may not be the form that acts a miRNA once inside the cell. Thus, it is contemplated that in some embodiments, a synthetic miRNA or a nonsynthetic miRNA is provided such that it becomes processed into a mature and active miRNA once it has access to the cell's miRNA processing machinery. In certain embodiments, it is specifically contemplated that the miRNA molecule provided is not a mature miRNA molecule but a nucleic acid molecule that can be processed into the mature miRNA once it is accessible to miRNA processing machinery. The term “nonsynthetic” in the context of miRNA means that the miRNA is not “synthetic,” as defined herein. Furthermore, it is contemplated that in embodiments of the invention that concern the use of synthetic miRNAs, the use of corresponding nonsynthetic miRNAs is also considered an aspect of the invention, and vice versa. It will be understand that the term “providing” an agent is used to include “administering” the agent to a patient.


In certain embodiments, methods also include targeting a miRNA to modulate in a cell or organism. The term “targeting a miRNA to modulate” means a nucleic acid of the invention will be employed so as to modulate the selected miRNA. In some embodiments the modulation is achieved with a synthetic or non-synthetic miRNA that corresponds to the targeted miRNA, which effectively provides the targeted miRNA to the cell or organism (positive modulation). In other embodiments, the modulation is achieved with a miRNA inhibitor, which effectively inhibits the targeted miRNA in the cell or organism (negative modulation).


In some embodiments, the miRNA targeted to be modulated is a miRNA that affects a disease, condition, or pathway. In certain embodiments, the miRNA is targeted because a treatment can be provided by negative modulation of the targeted miRNA. In other embodiments, the miRNA is targeted because a treatment can be provided by positive modulation of the targeted miRNA or its targets.


In certain methods of the invention, there is a further step of administering the selected miRNA modulator to a cell, tissue, organ, or organism (collectively “biological matter”) in need of treatment related to modulation of the targeted miRNA or in need of the physiological or biological results discussed herein (such as with respect to a particular cellular pathway or result like decrease in cell viability). Consequently, in some methods of the invention there is a step of identifying a patient in need of treatment that can be provided by the miRNA modulator(s). It is contemplated that an effective amount of a miRNA modulator can be administered in some embodiments. In particular embodiments, there is a therapeutic benefit conferred on the biological matter, where a “therapeutic benefit” refers to an improvement in the one or more conditions or symptoms associated with a disease or condition or an improvement in the prognosis, duration, or status with respect to the disease. It is contemplated that a therapeutic benefit includes, but is not limited to, a decrease in pain, a decrease in morbidity, a decrease in a symptom. For example, with respect to cancer, it is contemplated that a therapeutic benefit can be inhibition of tumor growth, prevention of metastasis, reduction in number of metastases, inhibition of cancer cell proliferation, induction of cell death in cancer cells, inhibition of angiogenesis near cancer cells, induction of apoptosis of cancer cells, reduction in pain, reduction in risk of recurrence, induction of chemo- or radiosensitivity in cancer cells, prolongation of life, and/or delay of death directly or indirectly related to cancer.


Furthermore, it is contemplated that the miRNA compositions may be provided as part of a therapy to a patient, in conjunction with traditional therapies or preventative agents. Moreover, it is contemplated that any method discussed in the context of therapy may be applied as preventatively, particularly in a patient identified to be potentially in need of the therapy or at risk of the condition or disease for which a therapy is needed.


In addition, methods of the invention concern employing one or more nucleic acids corresponding to a miRNA and a therapeutic drug. The nucleic acid can enhance the effect or efficacy of the drug, reduce any side effects or toxicity, modify its bioavailability, and/or decrease the dosage or frequency needed. In certain embodiments, the therapeutic drug is a cancer therapeutic. Consequently, in some embodiments, there is a method of treating cancer in a patient comprising administering to the patient the cancer therapeutic and an effective amount of at least one miRNA molecule that improves the efficacy of the cancer therapeutic or protects non-cancer cells. Cancer therapies also include a variety of combination therapies with both chemical and radiation based treatments. Combination chemotherapies include but are not limited to, for example, 5-fluorouracil, alemtuzumab, amrubicin, bevacizumab, bleomycin, bortezomib, busulfan, camptothecin, capecitabine, cisplatin (CDDP), carboplatin, cetuximab, chlorambucil, cisplatin (CDDP), cyclophosphamide, camptothecin, COX-2 inhibitors (e.g., celecoxib), cyclophosphamide, cytarabine, dactinomycin, dasatinib, daunorubicin, dexamethasone, docetaxel, doxorubicin (adriamycin), EGFR inhibitors (gefitinib and cetuximab), erlotinib, estrogen receptor binding agents, etoposide (VP16), everolimus, farnesyl-protein transferase inhibitors, gefitinib, gemcitabine, gemtuzumab, ibritumomab, ifosfamide, imatinib mesylate, larotaxel, lapatinib, lonafamib, mechlorethamine, melphalan, methotrexate, mitomycin, navelbine, nitrosurea, nocodazole, oxaliplatin, paclitaxel, plicomycin, procarbazine, raloxifene, rituximab, sirolimus, sorafenib, sunitinib, tamoxifen, taxol, taxotere, temsirolimus, tipifarnib, tositumomab, transplatinum, trastuzumab, vinblastin, vincristin, or vinorelbine or any analog or derivative variant of the foregoing.


Generally, inhibitors of miRNAs can be given to decrease the activity of an endogenous miRNA. Similarly, nucleic acid molecules corresponding to the mature miRNA can be given to achieve the opposite effect as compared to when inhibitors of the miRNA are given. For example, inhibitors of miRNA molecules that increase cell proliferation can be provided to cells to increase proliferation or decrease cell proliferation. The present invention contemplates these embodiments in the context of the different physiological effects observed with the different miRNA molecules and miRNA inhibitors disclosed herein. These include, but are not limited to, the following physiological effects: increase and decreasing cell proliferation, increasing or decreasing apoptosis, increasing transformation, increasing or decreasing cell viability, activating or inhibiting a kinase (e.g., Erk), activating/inducing or inhibiting hTert, inhibit stimulation of growth promoting pathway (e.g., Stat 3 signaling), reduce or increase viable cell number, and increase or decrease number of cells at a particular phase of the cell cycle. Methods of the invention are generally contemplated to include providing or introducing one or more different nucleic acid molecules corresponding to one or more different miRNA molecules. It is contemplated that the following, at least the following, or at most the following number of different nucleic acid or miRNA molecules may be provided or introduced: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, or any range derivable therein. This also applies to the number of different miRNA molecules that can be provided or introduced into a cell.


II. PHARMACEUTICAL FORMULATIONS AND DELIVERY

Methods of the present invention include the delivery of an effective amount of a miRNA or an expression construct encoding the same. An “effective amount” of the pharmaceutical composition, generally, is defined as that amount sufficient to detectably and repeatedly to achieve the stated desired result, for example, to ameliorate, reduce, minimize or limit the extent of the disease or its symptoms. Other more rigorous definitions may apply, including elimination, eradication or cure of disease.


A. Administration


In certain embodiments, it is desired to kill cells, inhibit cell growth, inhibit metastasis, decrease tumor or tissue size, and/or reverse or reduce the malignant or disease phenotype of cells. The routes of administration will vary, naturally, with the location and nature of the lesion or site to be targeted, and include, e.g., intradermal, subcutaneous, regional, parenteral, intravenous, intramuscular, intranasal, systemic, and oral administration and formulation. Direct injection, intratumoral injection, or injection into tumor vasculature is specifically contemplated for discrete, solid, accessible tumors, or other accessible target areas. Local, regional, or systemic administration also may be appropriate. For tumors of >4 cm, the volume to be administered will be about 4-10 ml (preferably 10 ml), while for tumors of <4 cm, a volume of about 1-3 ml will be used (preferably 3 ml).


Multiple injections delivered as a single dose comprise about 0.1 to about 0.5 ml volumes. Compositions of the invention may be administered in multiple injections to a tumor or a targeted site. In certain aspects, injections may be spaced at approximately 1 cm intervals.


In the case of surgical intervention, the present invention may be used preoperatively, to render an inoperable tumor subject to resection. Alternatively, the present invention may be used at the time of surgery, and/or thereafter, to treat residual or metastatic disease. For example, a resected tumor bed may be injected or perfused with a formulation comprising a miRNA or combinations thereof. Administration may be continued post-resection, for example, by leaving a catheter implanted at the site of the surgery. Periodic post-surgical treatment also is envisioned. Continuous perfusion of an expression construct or a viral construct also is contemplated.


Continuous administration also may be applied where appropriate, for example, where a tumor or other undesired affected area is excised and the tumor bed or targeted site is treated to eliminate residual, microscopic disease. Delivery via syringe or catherization is contemplated. Such continuous perfusion may take place for a period from about 1-2 hours, to about 2-6 hours, to about 6-12 hours, to about 12-24 hours, to about 1-2 days, to about 1-2 wk or longer following the initiation of treatment. Generally, the dose of the therapeutic composition via continuous perfusion will be equivalent to that given by a single or multiple injections, adjusted over a period of time during which the perfusion occurs.


Treatment regimens may vary as well and often depend on tumor type, tumor location, immune condition, target site, disease progression, and health and age of the patient. Certain tumor types will require more aggressive treatment. The clinician will be best suited to make such decisions based on the known efficacy and toxicity (if any) of the therapeutic formulations.


In certain embodiments, the tumor or affected area being treated may not, at least initially, be resectable. Treatments with compositions of the invention may increase the resectability of the tumor due to shrinkage at the margins or by elimination of certain particularly invasive portions. Following treatments, resection may be possible. Additional treatments subsequent to resection may serve to eliminate microscopic residual disease at the tumor or targeted site.


Treatments may include various “unit doses.” A unit dose is defined as containing a predetermined quantity of a therapeutic composition(s). The quantity to be administered, and the particular route and formulation, are within the skill of those in the clinical arts. A unit dose need not be administered as a single injection but may comprise continuous infusion over a set period of time. With respect to a viral component of the present invention, a unit dose may conveniently be described in terms of μg or mg of miRNA or miRNA mimetic. Alternatively, the amount specified may be the amount administered as the average daily, average weekly, or average monthly dose.


miRNA can be administered to the patient in a dose or doses of about or of at least about 0.5, 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990, 1000 μg or mg, or more, or any range derivable therein. Alternatively, the amount specified may be the amount administered as the average daily, average weekly, or average monthly dose, or it may be expressed in terms of mg/kg, where kg refers to the weight of the patient and the mg is specified above. In other embodiments, the amount specified is any number discussed above but expressed as mg/m2 (with respect to tumor size or patient surface area).


B. Injectable Compositions and Formulations


In some embodiments, the method for the delivery of a miRNA or an expression construct encoding such or combinations thereof is via systemic administration. However, the pharmaceutical compositions disclosed herein may also be administered parenterally, subcutaneously, directly, intratracheally, intravenously, intradermally, intramuscularly, or even intraperitoneally as described in U.S. Pat. Nos. 5,543,158; 5,641,515 and 5,399,363 (each specifically incorporated herein by reference in its entirety).


Injection of nucleic acids may be delivered by syringe or any other method used for injection of a solution, as long as the nucleic acid and any associated components can pass through the particular gauge of needle required for injection. A syringe system has also been described for use in gene therapy that permits multiple injections of predetermined quantities of a solution precisely at any depth (U.S. Pat. No. 5,846,225).


Solutions of the active compounds as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, mixtures thereof, and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (U.S. Pat. No. 5,466,468, specifically incorporated herein by reference in its entirety). In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils. Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.


In certain formulations, a water-based formulation is employed while in others, it may be lipid-based. In particular embodiments of the invention, a composition comprising a tumor suppressor protein or a nucleic acid encoding the same is in a water-based formulation. In other embodiments, the formulation is lipid based.


For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous, intratumoral, intralesional, and intraperitoneal administration. In this connection, sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, “Remington's Pharmaceutical Sciences” 15th Edition, pages 1035-1038 and 1570-1580). Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. Moreover, for human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biologics standards.


As used herein, a “carrier” includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.


The phrase “pharmaceutically acceptable” refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to a human.


The nucleic acid(s) are administered in a manner compatible with the dosage formulation, and in such amount as will be therapeutically effective. The quantity to be administered depends on the subject to be treated, including, e.g., the aggressiveness of the disease or cancer, the size of any tumor(s) or lesions, the previous or other courses of treatment. Precise amounts of active ingredient required to be administered depend on the judgment of the practitioner. Suitable regimes for initial administration and subsequent administration are also variable, but are typified by an initial administration followed by other administrations. Such administration may be systemic, as a single dose, continuous over a period of time spanning 10, 20, 30, 40, 50, 60 minutes, and/or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or more hours, and/or 1, 2, 3, 4, 5, 6, 7, days or more. Moreover, administration may be through a time release or sustained release mechanism, implemented by formulation and/or mode of administration.


C. Combination Treatments


In certain embodiments, the compositions and methods of the present invention involve a miRNA, or expression construct encoding such. These miRNA compositions can be used in combination with a second therapy to enhance the effect of the miRNA therapy, or increase the therapeutic effect of another therapy being employed. These compositions would be provided in a combined amount effective to achieve the desired effect, such as the killing of a cancer cell and/or the inhibition of cellular hyperproliferation. This process may involve contacting the cells with the miRNA or second therapy at the same or different time. This may be achieved by contacting the cell with one or more compositions or pharmacological formulation that includes or more of the agents, or by contacting the cell with two or more distinct compositions or formulations, wherein one composition provides (1) miRNA; and/or (2) a second therapy. A second composition or method may be administered that includes a chemotherapy, radiotherapy, surgical therapy, immunotherapy or gene therapy.


It is contemplated that one may provide a patient with the miRNA therapy and the second therapy within about 12-24 h of each other and, more preferably, within about 6-12 h of each other. In some situations, it may be desirable to extend the time period for treatment significantly, however, where several days (2, 3, 4, 5, 6 or 7) to several weeks (1, 2, 3, 4, 5, 6, 7 or 8) lapse between the respective administrations.


In certain embodiments, a course of treatment will last 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90 days or more. It is contemplated that one agent may be given on day 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, and/or 90, any combination thereof, and another agent is given on day 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, and/or 90, or any combination thereof. Within a single day (24-hour period), the patient may be given one or multiple administrations of the agent(s). Moreover, after a course of treatment, it is contemplated that there is a period of time at which no treatment is administered. This time period may last 1, 2, 3, 4, 5, 6, 7 days, and/or 1, 2, 3, 4, 5 weeks, and/or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 months or more, depending on the condition of the patient, such as their prognosis, strength, health, etc.


Various combinations may be employed, for example miRNA therapy is “A” and a second therapy is “B”:














A/B/A B/A/B B/B/A A/A/B A/B/B B/A/A A/B/B/B B/A/B/B


B/B/B/A B/B/A/B A/A/B/B A/B/A/B A/B/B/A B/B/A/A


B/A/B/A B/A/A/B A/A/A/B B/A/A/A A/B/A/A A/A/B/A









Administration of any compound or therapy of the present invention to a patient will follow general protocols for the administration of such compounds, taking into account the toxicity, if any, of the vector or any protein or other agent. Therefore, in some embodiments there is a step of monitoring toxicity that is attributable to combination therapy. It is expected that the treatment cycles would be repeated as necessary. It also is contemplated that various standard therapies, as well as surgical intervention, may be applied in combination with the described therapy.


In specific aspects, it is contemplated that a second therapy, such as chemotherapy, radiotherapy, immunotherapy, surgical therapy or other gene therapy, is employed in combination with the miRNA therapy, as described herein.


1. Chemotherapy


A wide variety of chemotherapeutic agents may be used in accordance with the present invention. The term “chemotherapy” refers to the use of drugs to treat cancer. A “chemotherapeutic agent” is used to connote a compound or composition that is administered in the treatment of cancer. These agents or drugs are categorized by their mode of activity within a cell, for example, whether and at what stage they affect the cell cycle. Alternatively, an agent may be characterized based on its ability to directly cross-link DNA, to intercalate into DNA, or to induce chromosomal and mitotic aberrations by affecting nucleic acid synthesis. Most chemotherapeutic agents fall into the following categories: alkylating agents, antimetabolites, antitumor antibiotics, mitotic inhibitors, and nitrosoureas.


a. Alkylating Agents


Alkylating agents are drugs that directly interact with genomic DNA to prevent the cancer cell from proliferating. This category of chemotherapeutic drugs represents agents that affect all phases of the cell cycle, that is, they are not phase-specific. Alkylating agents can be implemented to treat chronic leukemia, non-Hodgkin's lymphoma, Hodgkin's disease, multiple myeloma, and particular cancers of the breast, lung, and ovary. They include: busulfan, chlorambucil, cisplatin, cyclophosphamide (cytoxan), dacarbazine, ifosfamide, mechlorethamine (mustargen), and melphalan. Troglitazaone can be used to treat cancer in combination with any one or more of these alkylating agents.


b. Antimetabolites


Antimetabolites disrupt DNA and RNA synthesis. Unlike alkylating agents, they specifically influence the cell cycle during S phase. They have been used to combat chronic leukemias in addition to tumors of breast, ovary and the gastrointestinal tract. Antimetabolites include 5-fluorouracil (5-FU), cytarabine (Ara-C), fludarabine, gemcitabine, and methotrexate.


5-Fluorouracil (5-FU) has the chemical name of 5-fluoro-2,4(1H,3H)-pyrimidinedione. Its mechanism of action is thought to be by blocking the methylation reaction of deoxyuridylic acid to thymidylic acid. Thus, 5-FU interferes with the synthesis of deoxyribonucleic acid (DNA) and to a lesser extent inhibits the formation of ribonucleic acid (RNA). Since DNA and RNA are essential for cell division and proliferation, it is thought that the effect of 5-FU is to create a thymidine deficiency leading to cell death. Thus, the effect of 5-FU is found in cells that rapidly divide, a characteristic of metastatic cancers.


c. Antitumor Antibiotics


Antitumor antibiotics have both antimicrobial and cytotoxic activity. These drugs also interfere with DNA by chemically inhibiting enzymes and mitosis or altering cellular membranes. These agents are not phase specific so they work in all phases of the cell cycle. Thus, they are widely used for a variety of cancers. Examples of antitumor antibiotics include bleomycin, dactinomycin, daunorubicin, doxorubicin (Adriamycin), and idarubicin, some of which are discussed in more detail below. Widely used in clinical setting for the treatment of neoplasms, these compounds are administered through bolus injections intravenously at doses ranging from 25-75 mg/m2 at 21 day intervals for adriamycin, to 35-100 mg/m2 for etoposide intravenously or orally.


d. Mitotic Inhibitors


Mitotic inhibitors include plant alkaloids and other natural agents that can inhibit either protein synthesis required for cell division or mitosis. They operate during a specific phase during the cell cycle. Mitotic inhibitors comprise docetaxel, etoposide (VP16), paclitaxel, taxol, taxotere, vinblastine, vincristine, and vinorelbine.


e. Nitrosureas


Nitrosureas, like alkylating agents, inhibit DNA repair proteins. They are used to treat non-Hodgkin's lymphomas, multiple myeloma, malignant melanoma, in addition to brain tumors. Examples include carmustine and lomustine.


2. Radiotherapy


Radiotherapy, also called radiation therapy, is the treatment of cancer and other diseases with ionizing radiation. Ionizing radiation deposits energy that injures or destroys cells in the area being treated by damaging their genetic material, making it impossible for these cells to continue to grow. Although radiation damages both cancer cells and normal cells, the latter are able to repair themselves and function properly. Radiotherapy may be used to treat localized solid tumors, such as cancers of the skin, tongue, larynx, brain, breast, or cervix. It can also be used to treat leukemia and lymphoma (cancers of the blood-forming cells and lymphatic system, respectively).


Radiation therapy used according to the present invention may include, but is not limited to, the use of γ-rays, X-rays, and/or the directed delivery of radioisotopes to tumor cells. Other forms of DNA damaging factors are also contemplated such as microwaves, proton beam irradiation (U.S. Pat. Nos. 5,760,395 and 4,870,287) and UV-irradiation. It is most likely that all of these factors effect a broad range of damage on DNA, on the precursors of DNA, on the replication and repair of DNA, and on the assembly and maintenance of chromosomes. Dosage ranges for X-rays range from daily doses of 50 to 200 roentgens for prolonged periods of time (3 to 4 wk), to single doses of 2000 to 6000 roentgens. Dosage ranges for radioisotopes vary widely, and depend on the half-life of the isotope, the strength and type of radiation emitted, and the uptake by the neoplastic cells. Radiotherapy may comprise the use of radiolabeled antibodies to deliver doses of radiation directly to the cancer site (radioimmunotherapy). Once injected into the body, the antibodies actively seek out the cancer cells, which are destroyed by the cell-killing (cytotoxic) action of the radiation. This approach can minimize the risk of radiation damage to healthy cells.


Stereotactic radio-surgery (gamma knife) for brain and other tumors does not use a knife, but very precisely targeted beams of gamma radiotherapy from hundreds of different angles. Only one session of radiotherapy, taking about four to five hours, is needed. For this treatment a specially made metal frame is attached to the head. Then, several scans and x-rays are carried out to find the precise area where the treatment is needed. During the radiotherapy for brain tumors, the patient lies with their head in a large helmet, which has hundreds of holes in it to allow the radiotherapy beams through. Related approaches permit positioning for the treatment of tumors in other areas of the body.


3. Immunotherapy


In the context of cancer treatment, immunotherapeutics, generally, rely on the use of immune effector cells and molecules to target and destroy cancer cells. Trastuzumab (Herceptin™) is such an example. The immune effector may be, for example, an antibody specific for some marker on the surface of a tumor cell. The antibody alone may serve as an effector of therapy or it may recruit other cells to actually affect cell killing. The antibody also may be conjugated to a drug or toxin (chemotherapeutic, radionuclide, ricin A chain, cholera toxin, pertussis toxin, etc.) and serve merely as a targeting agent. Alternatively, the effector may be a lymphocyte carrying a surface molecule that interacts, either directly or indirectly, with a tumor cell target. Various effector cells include cytotoxic T cells and NK cells. The combination of therapeutic modalities, i.e., direct cytotoxic activity and inhibition or reduction of ErbB2 would provide therapeutic benefit in the treatment of ErbB2 overexpressing cancers.


In one aspect of immunotherapy, the tumor or disease cell must bear some marker that is amenable to targeting, i.e., is not present on the majority of other cells. Many tumor markers exist and any of these may be suitable for targeting in the context of the present invention. Common tumor markers include carcinoembryonic antigen, prostate specific antigen, urinary tumor associated antigen, fetal antigen, tyrosinase (p97), gp68, TAG-72, HMFG, Sialyl Lewis Antigen, MucA, MucB, PLAP, estrogen receptor, laminin receptor, erb B and p155. An alternative aspect of immunotherapy is to combine anticancer effects with immune stimulatory effects. Immune stimulating molecules also exist including: cytokines such as IL-2, IL-4, IL-12, GM-CSF, gamma-IFN, chemokines such as MIP-1, MCP-1, IL-8 and growth factors such as FLT3 ligand. Combining immune stimulating molecules, either as proteins or using gene delivery in combination with a tumor suppressor such as MDA-7 has been shown to enhance anti-tumor effects (Ju et al., 2000). Moreover, antibodies against any of these compounds can be used to target the anti-cancer agents discussed herein.


Examples of immunotherapies currently under investigation or in use are immune adjuvants e.g., Mycobacterium bovis, Plasmodium falciparum, dinitrochlorobenzene and aromatic compounds (U.S. Pat. Nos. 5,801,005 and 5,739,169; Hui and Hashimoto, 1998; Christodoulides et al., 1998), cytokine therapy e.g., interferons α, β and γ; IL-1, GM-CSF and TNF (Bukowski et al., 1998; Davidson et al., 1998; Hellstrand et al., 1998) gene therapy e.g., TNF, IL-1, IL-2, p53 (Qin et al., 1998; Austin-Ward and Villaseca, 1998; U.S. Pat. Nos. 5,830,880 and 5,846,945) and monoclonal antibodies e.g., anti-ganglioside GM2, anti-HER-2, anti-p185; Pietras et al., 1998; Hanibuchi et al., 1998; U.S. Pat. No. 5,824,311). Herceptin (trastuzumab) is a chimeric (mouse-human) monoclonal antibody that blocks the HER2-neu receptor. It possesses anti-tumor activity and has been approved for use in the treatment of malignant tumors (Dillman, 1999). A non-limiting list of several known anti-cancer immunotherapeutic agents and their targets includes (Generic Name/Target) Cetuximab/EGFR, Panitumuma/EGFR, Trastuzumab/erbB2 receptor, Bevacizumab/VEGF, Alemtuzumab/CD52, Gemtuzumab ozogamicin/CD33, Rituximab/CD20, Tositumomab/CD20, Matuzumab/EGFR, Ibritumomab tiuxetan/CD20, Tositumomab/CD20, HuPAM4/MUC1, MORAb-009/Mesothelin, G250/carbonic anhydrase IX, mAb 8H9/8H9 antigen, M195/CD33, Ipilimumab/CTLA4, HuLuc63/CS1, Alemtuzumab/CD53, Epratuzumab/CD22, BC8/CD45, HuJ591/Prostate specific membrane antigen, hA20/CD20, Lexatumumab/TRAIL receptor-2, Pertuzumab/HER-2 receptor, Mik-beta-1/IL-2R, RAV12/RAAG12, SGN-30/CD30, AME-133v/CD20, HeFi-1/CD30, BMS-663513/CD137, Volociximab/anti-α5β1 integrin, GC1008/TGFβ, HCD122/CD40, Siplizumab/CD2, MORAb-003/Folate receptor alpha, CNTO 328/IL-6, MDX-060/CD30, Ofatumumab/CD20, and SGN-33/CD33. It is contemplated that one or more of these therapies may be employed with the miRNA therapies described herein.


A number of different approaches for passive immunotherapy of cancer exist. They may be broadly categorized into the following: injection of antibodies alone; injection of antibodies coupled to toxins or chemotherapeutic agents; injection of antibodies coupled to radioactive isotopes; injection of anti-idiotype antibodies; and finally, purging of tumor cells in bone marrow.


4. Gene Therapy


In yet another embodiment, a combination treatment involves gene therapy in which a therapeutic polynucleotide is administered before, after, or at the same time as one or more therapeutic miRNA. Delivery of a therapeutic polypeptide or encoding nucleic acid in conjunction with a miRNA may have a combined therapeutic effect on target tissues. A variety of proteins are encompassed within the invention, some of which are described below. Various genes that may be targeted for gene therapy of some form in combination with the present invention include, but are not limited to inducers of cellular proliferation, inhibitors of cellular proliferation, regulators of programmed cell death, cytokines and other therapeutic nucleic acids or nucleic acid that encode therapeutic proteins.


The tumor suppressor oncogenes function to inhibit excessive cellular proliferation. The inactivation of these genes destroys their inhibitory activity, resulting in unregulated proliferation. The tumor suppressors (e.g., therapeutic polypeptides) p53, FHIT, p16 and C-CAM can be employed.


In addition to p53, another inhibitor of cellular proliferation is p16. The major transitions of the eukaryotic cell cycle are triggered by cyclin-dependent kinases, or CDK's. One CDK, cyclin-dependent kinase 4 (CDK4), regulates progression through the G1. The activity of this enzyme may be to phosphorylate Rb at late G1. The activity of CDK4 is controlled by an activating subunit, D-type cyclin, and by an inhibitory subunit, the p16INK4 has been biochemically characterized as a protein that specifically binds to and inhibits CDK4, and thus may regulate Rb phosphorylation (Serrano et al., 1993; Serrano et al., 1995). Since the p16INK4 protein is a CDK4 inhibitor (Serrano, 1993), deletion of this gene may increase the activity of CDK4, resulting in hyperphosphorylation of the Rb protein. p16 also is known to regulate the function of CDK6.


p16INK4 belongs to a newly described class of CDK-inhibitory proteins that also includes p16B, p19, p21WAF1, and p27KIP1. The p16INK4 gene maps to 9p21, a chromosome region frequently deleted in many tumor types. Homozygous deletions and mutations of the p16INK4 gene are frequent in human tumor cell lines. This evidence suggests that the p16INK4 gene is a tumor suppressor gene. This interpretation has been challenged, however, by the observation that the frequency of the p16INK4 gene alterations is much lower in primary uncultured tumors than in cultured cell lines (Caldas et al., 1994; Cheng et al., 1994; Hussussian et al., 1994; Kamb et al., 1994; Mori et al., 1994; Okamoto et al., 1994; Nobori et al., 1994; Orlow et al., 1994; Arap et al., 1995). Restoration of wild-type p16INK4 function by transfection with a plasmid expression vector reduced colony formation by some human cancer cell lines (Okamoto, 1994; Arap, 1995).


Other genes that may be employed according to the present invention include Rb, APC, DCC, NF-1, NF-2, WT-1, MEN-I, MEN-II, zac1, p73, VHL, MMAC1/PTEN, DBCCR-1, FCC, rsk-3, p27, p27/p16 fusions, p21/p27 fusions, anti-thrombotic genes (e.g., COX-1, TFPI), PGS, Dp, E2F, ras, myc, neu, raf, erb, fms, trk, ret, gsp, hst, abl, E1A, p300, genes involved in angiogenesis (e.g., VEGF, FGF, thrombospondin, BAI-1, GDAIF, or their receptors) and MCC.


5. Surgery


Approximately 60% of persons with cancer will undergo surgery of some type, which includes preventative, diagnostic or staging, curative and palliative surgery. Curative surgery is a cancer treatment that may be used in conjunction with other therapies, such as the treatment of the present invention, chemotherapy, radiotherapy, hormonal therapy, gene therapy, immunotherapy and/or alternative therapies.


Curative surgery includes resection in which all or part of cancerous tissue is physically removed, excised, and/or destroyed. Tumor resection refers to physical removal of at least part of a tumor. In addition to tumor resection, treatment by surgery includes laser surgery, cryosurgery, electrosurgery, and microscopically controlled surgery (Mohs' surgery). It is further contemplated that the present invention may be used in conjunction with removal of superficial cancers, precancers, or incidental amounts of normal tissue.


Upon excision of part of all of cancerous cells, tissue, or tumor, a cavity may be formed in the body. Treatment may be accomplished by perfusion, direct injection or local application of the area with an additional anti-cancer therapy. Such treatment may be repeated, for example, every 1, 2, 3, 4, 5, 6, or 7 days, or every 1, 2, 3, 4, and 5 weeks or every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months. These treatments may be of varying dosages as well.


6. Other Agents


It is contemplated that other agents may be used in combination with the present invention to improve the therapeutic efficacy of treatment. These additional agents include immunomodulatory agents, agents that affect the upregulation of cell surface receptors and GAP junctions, cytostatic and differentiation agents, inhibitors of cell adhesion, agents that increase the sensitivity of the hyperproliferative cells to apoptotic inducers, or other biological agents. Immunomodulatory agents include tumor necrosis factor; interferon alpha, beta, and gamma; IL-2 and other cytokines; F42K and other cytokine analogs; or MIP-1, MIP-1beta, MCP-1, RANTES, and other chemokines. It is further contemplated that the upregulation of cell surface receptors or their ligands such as Fas/Fas ligand, DR4 or DR5/TRAIL (Apo-2 ligand) would potentiate the apoptotic inducing abilities of the present invention by establishment of an autocrine or paracrine effect on hyperproliferative cells. Increases intercellular signaling by elevating the number of GAP junctions would increase the anti-hyperproliferative effects on the neighboring hyperproliferative cell population. In other embodiments, cytostatic or differentiation agents can be used in combination with the present invention to improve the anti-hyperproliferative efficacy of the treatments. Inhibitors of cell adhesion are contemplated to improve the efficacy of the present invention. Examples of cell adhesion inhibitors are focal adhesion kinase (FAKs) inhibitors and Lovastatin. It is further contemplated that other agents that increase the sensitivity of a hyperproliferative cell to apoptosis, such as the antibody c225, could be used in combination with the present invention to improve the treatment efficacy.


Apo2 ligand (Apo2L, also called TRAIL) is a member of the tumor necrosis factor (TNF) cytokine family. TRAIL activates rapid apoptosis in many types of cancer cells, yet is not toxic to normal cells. TRAIL mRNA occurs in a wide variety of tissues. Most normal cells appear to be resistant to TRAIL's cytotoxic action, suggesting the existence of mechanisms that can protect against apoptosis induction by TRAIL. The first receptor described for TRAIL, called death receptor 4 (DR4), contains a cytoplasmic “death domain”; DR4 transmits the apoptosis signal carried by TRAIL. Additional receptors have been identified that bind to TRAIL. One receptor, called DR5, contains a cytoplasmic death domain and signals apoptosis much like DR4. The DR4 and DR5 mRNAs are expressed in many normal tissues and tumor cell lines. Recently, decoy receptors such as DcR1 and DcR2 have been identified that prevent TRAIL from inducing apoptosis through DR4 and DR5. These decoy receptors thus represent a novel mechanism for regulating sensitivity to a pro-apoptotic cytokine directly at the cell's surface. The preferential expression of these inhibitory receptors in normal tissues suggests that TRAIL may be useful as an anticancer agent that induces apoptosis in cancer cells while sparing normal cells. (Marsters et al., 1999).


There have been many advances in the therapy of cancer following the introduction of cytotoxic chemotherapeutic drugs. However, one of the consequences of chemotherapy is the development/acquisition of drug-resistant phenotypes and the development of multiple drug resistance. The development of drug resistance remains a major obstacle in the treatment of such tumors and therefore, there is an obvious need for alternative approaches such as gene therapy.


Another form of therapy for use in conjunction with chemotherapy, radiation therapy or biological therapy includes hyperthermia, which is a procedure in which a patient's tissue is exposed to high temperatures (up to 106° F.). External or internal heating devices may be involved in the application of local, regional, or whole-body hyperthermia. Local hyperthermia involves the application of heat to a small area, such as a tumor. Heat may be generated externally with high-frequency waves targeting a tumor from a device outside the body. Internal heat may involve a sterile probe, including thin, heated wires or hollow tubes filled with warm water, implanted microwave antennae, or radiofrequency electrodes.


A patient's organ or a limb is heated for regional therapy, which is accomplished using devices that produce high energy, such as magnets. Alternatively, some of the patient's blood may be removed and heated before being perfused into an area that will be internally heated. Whole-body heating may also be implemented in cases where cancer has spread throughout the body. Warm-water blankets, hot wax, inductive coils, and thermal chambers may be used for this purpose.


Hormonal therapy may also be used in conjunction with the present invention or in combination with any other cancer therapy previously described. The use of hormones may be employed in the treatment of certain cancers such as breast, prostate, ovarian, or cervical cancer to lower the level or block the effects of certain hormones such as testosterone or estrogen. This treatment is often used in combination with at least one other cancer therapy as a treatment option or to reduce the risk of metastases.


This application incorporates U.S. application Ser. No. 11/349,727 filed on Feb. 8, 2006 claiming priority to U.S. Provisional Application Ser. No. 60/650,807 filed Feb. 8, 2005 herein by references in its entirety.


III. miRNA MOLECULES

MicroRNA molecules (“miRNAs”) are generally 21 to 22 nucleotides in length, though lengths of 19 and up to 23 nucleotides have been reported. The miRNAs are each processed from a longer precursor RNA molecule (“precursor miRNA”). Precursor miRNAs are transcribed from non-protein-encoding genes. The precursor miRNAs have two regions of complementarity that enables them to form a stem-loop- or fold-back-like structure, which is cleaved in animals by a ribonuclease III-like nuclease enzyme called Dicer. The processed miRNA is typically a portion of the stem.


The processed miRNA (also referred to as “mature miRNA”) becomes part of a large complex to down-regulate a particular target gene or its gene product. Examples of animal miRNAs include those that imperfectly basepair with the target, which halts translation (Olsen et al., 1999; Seggerson et al., 2002). siRNA molecules also are processed by Dicer, but from a long, double-stranded RNA molecule. siRNAs are not naturally found in animal cells, but they can direct the sequence-specific cleavage of an mRNA target through a RNA-induced silencing complex (RISC) (Denli et al., 2003).


A. Array Preparation


Certain embodiments of the present invention concerns the preparation and use of mRNA or nucleic acid arrays, miRNA or nucleic acid arrays, and/or miRNA or nucleic acid probe arrays, which are macroarrays or microarrays of nucleic acid molecules (probes) that are fully or nearly complementary (over the length of the prove) or identical (over the length of the prove) to a plurality of nucleic acid, mRNA or miRNA molecules, precursor miRNA molecules, or nucleic acids derived from the various genes and gene pathways modulated by miR-10 miRNAs and that are positioned on a support or support material in a spatially separated organization. Macroarrays are typically sheets of nitrocellulose or nylon upon which probes have been spotted. Microarrays position the nucleic acid probes more densely such that up to 10,000 nucleic acid molecules can be fit into a region typically 1 to 4 square centimeters. Microarrays can be fabricated by spotting nucleic acid molecules, e.g., genes, oligonucleotides, etc., onto substrates or fabricating oligonucleotide sequences in situ on a substrate. Spotted or fabricated nucleic acid molecules can be applied in a high density matrix pattern of up to about 30 non-identical nucleic acid molecules per square centimeter or higher, e.g. up to about 100 or even 1000 per square centimeter. Microarrays typically use coated glass as the solid support, in contrast to the nitrocellulose-based material of filter arrays. By having an ordered array of marker RNA and/or miRNA-complementing nucleic acid samples, the position of each sample can be tracked and linked to the original sample.


A variety of different array devices in which a plurality of distinct nucleic acid probes are stably associated with the surface of a solid support are known to those of skill in the art. Useful substrates for arrays include nylon, glass, metal, plastic, latex, and silicon. Such arrays may vary in a number of different ways, including average probe length, sequence or types of probes, nature of bond between the probe and the array surface, e.g. covalent or non-covalent, and the like. The labeling and screening methods of the present invention and the arrays are not limited in its utility with respect to any parameter except that the probes detect miRNA, or genes or nucleic acid representative of genes; consequently, methods and compositions may be used with a variety of different types of nucleic acid arrays.


Representative methods and apparatus for preparing a microarray have been described, for example, in U.S. Pat. Nos. 5,143,854; 5,202,231; 5,242,974; 5,288,644; 5,324,633; 5,384,261; 5,405,783; 5,412,087; 5,424,186; 5,429,807; 5,432,049; 5,436,327; 5,445,934; 5,468,613; 5,470,710; 5,472,672; 5,492,806; 5,525,464; 5,503,980; 5,510,270; 5,525,464; 5,527,681; 5,529,756; 5,532,128; 5,545,531; 5,547,839; 5,554,501; 5,556,752; 5,561,071; 5,571,639; 5,580,726; 5,580,732; 5,593,839; 5,599,695; 5,599,672; 5,610,287; 5,624,711; 5,631,134; 5,639,603; 5,654,413; 5,658,734; 5,661,028; 5,665,547; 5,667,972; 5,695,940; 5,700,637; 5,744,305; 5,800,992; 5,807,522; 5,830,645; 5,837,196; 5,871,928; 5,847,219; 5,876,932; 5,919,626; 6,004,755; 6,087,102; 6,368,799; 6,383,749; 6,617,112; 6,638,717; 6,720,138, as well as WO 93/17126; WO 95/11995; WO 95/21265; WO 95/21944; WO 95/35505; WO 96/31622; WO 97/10365; WO 97/27317; WO 99/35505; WO 09923256; WO 09936760; WO0138580; WO 0168255; WO 03020898; WO 03040410; WO 03053586; WO 03087297; WO 03091426; WO03100012; WO 04020085; WO 04027093; EP 373 203; EP 785 280; EP 799 897 and UK 8 803 000; the disclosures of which are all herein incorporated by reference.


It is contemplated that the arrays can be high density arrays, such that they contain 2, 20, 25, 50, 80, 100 or more different probes. It is contemplated that they may contain 1000, 16,000, 65,000, 250,000 or 1,000,000 or more different probes. The probes can be directed to mRNA and/or miRNA targets in one or more different organisms or cell types. The oligonucleotide probes range from 5 to 50, 5 to 45, 10 to 40, 9 to 34, or 15 to 40 nucleotides in length in some embodiments. In certain embodiments, the oligonucleotide probes are 5, 10, 15, 20 to 20, 25, 30, 35, 40 nucleotides in length including all integers and ranges there between.


The location and sequence of each different probe sequence in the array are generally known. Moreover, the large number of different probes can occupy a relatively small area providing a high density array having a probe density of generally greater than about 60, 100, 600, 1000, 5,000, 10,000, 40,000, 100,000, or 400,000 different oligonucleotide probes per cm2. The surface area of the array can be about or less than about 1, 1.6, 2, 3, 4, 5, 6, 7, 8, 9, or 10 cm2.


Moreover, a person of ordinary skill in the art could readily analyze data generated using an array. Such protocols are disclosed above, and include information found in WO 9743450; WO 03023058; WO 03022421; WO 03029485; WO 03067217; WO 03066906; WO 03076928; WO 03093810; WO 03100448A1, all of which are specifically incorporated by reference.


B. Sample Preparation


It is contemplated that the RNA and/or miRNA of a wide variety of samples can be analyzed using the arrays, index of probes, or array technology of the invention. While endogenous miRNA is contemplated for use with compositions and methods of the invention, recombinant miRNA—including nucleic acids that are complementary or identical to endogenous miRNA or precursor miRNA—can also be handled and analyzed as described herein. Samples may be biological samples, in which case, they can be from biopsy, fine needle aspirates, exfoliates, blood, tissue, organs, semen, saliva, tears, other bodily fluid, hair follicles, skin, or any sample containing or constituting biological cells, particularly cancer or hyperproliferative cells. In certain embodiments, samples may be, but are not limited to, biopsy, or cells purified or enriched to some extent from a biopsy or other bodily fluids or tissues. Alternatively, the sample may not be a biological sample, but be a chemical mixture, such as a cell-free reaction mixture (which may contain one or more biological enzymes).


C. Hybridization


After an array or a set of probes is prepared and/or the nucleic acid in the sample or probe is labeled, the population of target nucleic acids is contacted with the array or probes under hybridization conditions, where such conditions can be adjusted, as desired, to provide for an optimum level of specificity in view of the particular assay being performed. Suitable hybridization conditions are well known to those of skill in the art and reviewed in Sambrook et al. (2001) and WO 95/21944. Of particular interest in many embodiments is the use of stringent conditions during hybridization. Stringent conditions are known to those of skill in the art.


It is specifically contemplated that a single array or set of probes may be contacted with multiple samples. The samples may be labeled with different labels to distinguish the samples. For example, a single array can be contacted with a tumor tissue sample labeled with Cy3, and normal tissue sample labeled with Cy5. Differences between the samples for particular miRNAs corresponding to probes on the array can be readily ascertained and quantified.


The small surface area of the array permits uniform hybridization conditions, such as temperature regulation and salt content. Moreover, because of the small area occupied by the high density arrays, hybridization may be carried out in extremely small fluid volumes (e.g., about 250 μl or less, including volumes of about or less than about 5, 10, 25, 50, 60, 70, 80, 90, 100 μl, or any range derivable therein). In small volumes, hybridization may proceed very rapidly.


D. Differential Expression Analyses


Arrays of the invention can be used to detect differences between two samples. Specifically contemplated applications include identifying and/or quantifying differences between miRNA or gene expression from a sample that is normal and from a sample that is not normal, between a disease or condition and a cell not exhibiting such a disease or condition, or between two differently treated samples. Also, miRNA or gene expression may be compared between a sample believed to be susceptible to a particular disease or condition and one believed to be not susceptible or resistant to that disease or condition. A sample that is not normal is one exhibiting phenotypic or genotypic trait(s) of a disease or condition, or one believed to be not normal with respect to that disease or condition. It may be compared to a cell that is normal with respect to that disease or condition. Phenotypic traits include symptoms of, or susceptibility to, a disease or condition of which a component is or may or may not be genetic, or caused by a hyperproliferative or neoplastic cell or cells.


An array comprises a solid support with nucleic acid probes attached to the support. Arrays typically comprise a plurality of different nucleic acid probes that are coupled to a surface of a substrate in different, known locations. These arrays, also described as “microarrays” or colloquially “chips” have been generally described in the art, for example, U.S. Pat. Nos. 5,143,854, 5,445,934, 5,744,305, 5,677,195, 6,040,193, 5,424,186 and Fodor et al., (1991), each of which is incorporated by reference in its entirety for all purposes. Techniques for the synthesis of these arrays using mechanical synthesis methods are described in, e.g., U.S. Pat. No. 5,384,261, incorporated herein by reference in its entirety for all purposes. Although a planar array surface is used in certain aspects, the array may be fabricated on a surface of virtually any shape or even a multiplicity of surfaces. Arrays may be nucleic acids on beads, gels, polymeric surfaces, fibers such as fiber optics, glass or any other appropriate substrate, see U.S. Pat. Nos. 5,770,358, 5,789,162, 5,708,153, 6,040,193 and 5,800,992, which are hereby incorporated in their entirety for all purposes. Arrays may be packaged in such a manner as to allow for diagnostics or other manipulation of an all inclusive device, see for example, U.S. Pat. Nos. 5,856,174 and 5,922,591 incorporated in their entirety by reference for all purposes. See also U.S. patent application Ser. No. 09/545,207, filed Apr. 7, 2000 for additional information concerning arrays, their manufacture, and their characteristics, which is incorporated by reference in its entirety for all purposes.


Particularly, arrays can be used to evaluate samples with respect to pathological condition such as cancer and related conditions. It is specifically contemplated that the invention can be used to evaluate differences between stages or sub-classifications of disease, such as between benign, cancerous, and metastatic tissues or tumors.


Phenotypic traits to be assessed include characteristics such as longevity, morbidity, expected survival, susceptibility or receptivity to particular drugs or therapeutic treatments (drug efficacy), and risk of drug toxicity. Samples that differ in these phenotypic traits may also be evaluated using the compositions and methods described.


In certain embodiments, miRNA and/or expression profiles may be generated to evaluate and correlate those profiles with pharmacokinetics or therapies. For example, these profiles may be created and evaluated for patient tumor and blood samples prior to the patient's being treated or during treatment to determine if there are miRNA or genes whose expression correlates with the outcome of the patient's treatment. Identification of differential miRNAs or genes can lead to a diagnostic assay for evaluation of tumor and/or blood samples to determine what drug regimen the patient should be provided. In addition, it can be used to identify or select patients suitable for a particular clinical trial. If an expression profile is determined to be correlated with drug efficacy or drug toxicity, that profile is relevant to whether that patient is an appropriate patient for receiving a drug, for receiving a combination of drugs, or for a particular dosage of the drug.


In addition to the above prognostic assay, samples from patients with a variety of diseases can be evaluated to determine if different diseases can be identified based on miRNA and/or related gene expression levels. A diagnostic assay can be created based on the profiles that doctors can use to identify individuals with a disease or who are at risk to develop a disease. Alternatively, treatments can be designed based on miRNA profiling. Examples of such methods and compositions are described in the U.S. Provisional Patent Application entitled “Methods and Compositions Involving miRNA and miRNA Inhibitor Molecules” filed on May 23, 2005 in the names of David Brown, Lance Ford, Angie Cheng and Rich Jarvis, which is hereby incorporated by reference in its entirety.


E. Other Assays


In addition to the use of arrays and microarrays, it is contemplated that a number of different assays could be employed to analyze miRNAs or related genes, their activities, and their effects. Such assays include, but are not limited to, nucleic acid amplification, polymerase chain reaction, quantitative PCR, RT-PCR, in situ hybridization, Northern hybridization, hybridization protection assay (HPA) (GenProbe), branched DNA (bDNA) assay (Chiron), rolling circle amplification (RCA), single molecule hybridization detection (US Genomics), Invader assay (ThirdWave Technologies), and/or Bridge Litigation Assay (Genaco).


IV. NUCLEIC ACIDS

The present invention concerns nucleic acids, modified or mimetic nucleic acids, miRNAs, mRNAs, genes, and representative fragments thereof that can be labeled, used in array analysis, or employed in diagnostic, therapeutic, or prognostic applications, particularly those related to pathological conditions such as cancer. The molecules may have been endogenously produced by a cell, or been synthesized or produced chemically or recombinantly. They may be isolated and/or purified. Each of the miRNAs described herein and includes the corresponding SEQ ID NO and accession numbers for these miRNA sequences. The name of a miRNA is often abbreviated and referred to without a “hsa-” prefix and will be understood as such, depending on the context. Unless otherwise indicated, miRNAs referred to in the application are human sequences identified as miR-X or let-X, where X is a number and/or letter.


In certain aspects, a miRNA probe designated by a suffix “5P” or “3P” can be used. “5P” indicates that the mature miRNA derives from the 5′ end of the precursor and a corresponding “3P” indicates that it derives from the 3′ end of the precursor, as described on the world wide web at sanger.ac.uk. Moreover, in some embodiments, a miRNA probe is used that does not correspond to a known human miRNA. It is contemplated that these non-human miRNA probes may be used in embodiments of the invention or that there may exist a human miRNA that is homologous to the non-human miRNA. In other embodiments, any mammalian cell, biological sample, or preparation thereof may be employed.


In some embodiments of the invention, methods and compositions involving miRNA may concern miRNA, markers (mRNAs), and/or other nucleic acids. Nucleic acids may be, be at least, or be at most 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990, or 1000 nucleotides, or any range derivable therein, in length. Such lengths cover the lengths of processed miRNA, miRNA probes, precursor miRNA, miRNA containing vectors, mRNA, mRNA probes, control nucleic acids, and other probes and primers.


In many embodiments, miRNA are 19-24 nucleotides in length, while miRNA probes are 19-35 nucleotides in length, depending on the length of the processed miRNA and any flanking regions added. miRNA precursors are generally between 62 and 110 nucleotides in humans.


Nucleic acids of the invention may have regions of identity or complementarity to another nucleic acid. It is contemplated that the region of complementarity or identity can be at least 5 contiguous residues, though it is specifically contemplated that the region is, is at least, or is at most 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 441, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990, or 1000 contiguous nucleotides. It is further understood that the length of complementarity within a precursor miRNA or other nucleic acid or between a miRNA probe and a miRNA or a miRNA gene are such lengths. Moreover, the complementarity may be expressed as a percentage, meaning that the complementarity between a probe and its target is 90% or greater over the length of the probe. In some embodiments, complementarity is or is at least 90%, 95% or 100%. In particular, such lengths may be applied to any nucleic acid comprising a nucleic acid sequence identified in any of SEQ ID NOs described herein, accession number, or any other sequence disclosed herein. Typically, the commonly used name of the miRNA is given (with its identifying source in the prefix, for example, “hsa” for human sequences) and the processed miRNA sequence. Unless otherwise indicated, a miRNA without a prefix will be understood to refer to a human miRNA. Moreover, a lowercase letter in a miRNA name may or may not be lowercase; for example, hsa-mir-130b can also be referred to as miR-130B. The term “miRNA probe” refers to a nucleic acid probe that can identify a particular miRNA or structurally related miRNAs.


It is understood that some nucleic acids are derived from genomic sequences or a gene. In this respect, the term “gene” is used for simplicity to refer to the genomic sequence encoding the precursor nucleic acid or miRNA for a given miRNA or gene. However, embodiments of the invention may involve genomic sequences of a miRNA that are involved in its expression, such as a promoter or other regulatory sequences.


The term “recombinant” may be used and this generally refers to a molecule that has been manipulated in vitro or that is a replicated or expressed product of such a molecule.


The term “nucleic acid” is well known in the art. A “nucleic acid” as used herein will generally refer to a molecule (one or more strands) of DNA, RNA or a derivative or analog thereof, comprising a nucleobase. A nucleobase includes, for example, a naturally occurring purine or pyrimidine base found in DNA (e.g., an adenine “A,” a guanine “G,” a thymine “T” or a cytosine “C”) or RNA (e.g., an A, a G, an uracil “U” or a C). The term “nucleic acid” encompasses the terms “oligonucleotide” and “polynucleotide,” each as a subgenus of the term “nucleic acid.”


The term “miRNA” generally refers to a single-stranded molecule, but in specific embodiments, molecules implemented in the invention will also encompass a region or an additional strand that is partially (between 10 and 50% complementary across length of strand), substantially (greater than 50% but less than 100% complementary across length of strand) or fully complementary to another region of the same single-stranded molecule or to another nucleic acid. Thus, nucleic acids of the invention may encompass a molecule that comprises one or more complementary or self-complementary strand(s) or “complement(s)” of a particular sequence. For example, precursor miRNA may have a self-complementary region, which is up to 100% complementary. miRNA probes or nucleic acids of the invention can include, can be or can be at least 60, 65, 70, 75, 80, 85, 90, 95, 96, 97, 98, 99 or 100% complementary to their target.


It is understood that a “synthetic nucleic acid” of the invention means that the nucleic acid does not have all or part of a chemical structure or sequence of a naturally occurring nucleic acid. Consequently, it will be understood that the term “synthetic miRNA” refers to a “synthetic nucleic acid” that functions in a cell or under physiological conditions as a naturally occurring miRNA.


While embodiments of the invention may involve synthetic miRNAs or synthetic nucleic acids, in some embodiments of the invention, the nucleic acid molecule(s) need not be “synthetic.” In certain embodiments, a non-synthetic nucleic acid or miRNA employed in methods and compositions of the invention may have the entire sequence and structure of a naturally occurring mRNA or miRNA precursor or the mature mRNA or miRNA. For example, non-synthetic miRNAs used in methods and compositions of the invention may not have one or more modified nucleotides or nucleotide analogs. In these embodiments, the non-synthetic miRNA may or may not be recombinantly produced. In particular embodiments, the nucleic acid in methods and/or compositions of the invention is specifically a synthetic miRNA and not a non-synthetic miRNA (that is, not a miRNA that qualifies as “synthetic”); though in other embodiments, the invention specifically involves a non-synthetic miRNA and not a synthetic miRNA. Any embodiments discussed with respect to the use of synthetic miRNAs can be applied with respect to non-synthetic miRNAs, and vice versa.


It will be understood that the term “naturally occurring” refers to something found in an organism without any intervention by a person; it could refer to a naturally-occurring wildtype or mutant molecule. In some embodiments a synthetic miRNA molecule does not have the sequence of a naturally occurring miRNA molecule. In other embodiments, a synthetic miRNA molecule may have the sequence of a naturally occurring miRNA molecule, but the chemical structure of the molecule, particularly in the part unrelated specifically to the precise sequence (non-sequence chemical structure) differs from chemical structure of the naturally occurring miRNA molecule with that sequence. In some cases, the synthetic miRNA has both a sequence and non-sequence chemical structure that are not found in a naturally-occurring miRNA. Moreover, the sequence of the synthetic molecules will identify which miRNA is effectively being provided or inhibited; the endogenous miRNA will be referred to as the “corresponding miRNA.” Corresponding miRNA sequences that can be used in the context of the invention include, but are not limited to, all or a portion of those sequences in the SEQ IDs provided herein, as well as any other miRNA sequence, miRNA precursor sequence, or any sequence complementary thereof. In some embodiments, the sequence is or is derived from or contains all or part of a sequence identified herein to target a particular miRNA (or set of miRNAs) that can be used with that sequence. Any 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260 or any number or range of sequences there between may be selected to the exclusion of all non-selected sequences.


As used herein, “hybridization”, “hybridizes” or “capable of hybridizing” is understood to mean the forming of a double or triple stranded molecule or a molecule with partial double or triple stranded nature. The term “anneal” as used herein is synonymous with “hybridize.” The term “hybridization”, “hybridize(s)” or “capable of hybridizing” encompasses the terms “stringent condition(s)” or “high stringency” and the terms “low stringency” or “low stringency condition(s).”


As used herein “stringent condition(s)” or “high stringency” are those conditions that allow hybridization between or within one or more nucleic acid strand(s) containing complementary sequence(s), but preclude hybridization of random sequences. Stringent conditions tolerate little, if any, mismatch between a nucleic acid and a target strand. Such conditions are well known to those of ordinary skill in the art, and are preferred for applications requiring high selectivity. Non-limiting applications include isolating a nucleic acid, such as a gene or a nucleic acid segment thereof, or detecting at least one specific mRNA transcript or a nucleic acid segment thereof, and the like.


Stringent conditions may comprise low salt and/or high temperature conditions, such as provided by about 0.02 M to about 0.5 M NaCl at temperatures of about 42° C. to about 70° C. It is understood that the temperature and ionic strength of a desired stringency are determined in part by the length of the particular nucleic acid(s), the length and nucleobase content of the target sequence(s), the charge composition of the nucleic acid(s), and to the presence or concentration of formamide, tetramethylammonium chloride or other solvent(s) in a hybridization mixture.


It is also understood that these ranges, compositions and conditions for hybridization are mentioned by way of non-limiting examples only, and that the desired stringency for a particular hybridization reaction is often determined empirically by comparison to one or more positive or negative controls. Depending on the application envisioned it is preferred to employ varying conditions of hybridization to achieve varying degrees of selectivity of a nucleic acid towards a target sequence. In a non-limiting example, identification or isolation of a related target nucleic acid that does not hybridize to a nucleic acid under stringent conditions may be achieved by hybridization at low temperature and/or high ionic strength. Such conditions are termed “low stringency” or “low stringency conditions,” and non-limiting examples of low stringency include hybridization performed at about 0.15 M to about 0.9 M NaCl at a temperature range of about 20° C. to about 50° C. Of course, it is within the skill of one in the art to further modify the low or high stringency conditions to suite a particular application.


A. Nucleobase, Nucleoside, Nucleotide, and Modified Nucleotides


As used herein a “nucleobase” refers to a heterocyclic base, such as for example a naturally occurring nucleobase (i.e., an A, T, G, C or U) found in at least one naturally occurring nucleic acid (i.e., DNA and RNA), and naturally or non-naturally occurring derivative(s) and analogs of such a nucleobase. A nucleobase generally can form one or more hydrogen bonds (“anneal” or “hybridize”) with at least one naturally occurring nucleobase in a manner that may substitute for naturally occurring nucleobase pairing (e.g., the hydrogen bonding between A and T, G and C, and A and U).


“Purine” and/or “pyrimidine” nucleobase(s) encompass naturally occurring purine and/or pyrimidine nucleobases and also derivative(s) and analog(s) thereof, including but not limited to, those a purine or pyrimidine substituted by one or more of an alkyl, carboxyalkyl, amino, hydroxyl, halogen (i.e., fluoro, chloro, bromo, or iodo), thiol or alkylthiol moiety. Preferred alkyl (e.g., alkyl, caboxyalkyl, etc.) moieties comprise of from about 1, about 2, about 3, about 4, about 5, to about 6 carbon atoms. Other non-limiting examples of a purine or pyrimidine include a deazapurine, a 2,6-diaminopurine, a 5-fluorouracil, a xanthine, a hypoxanthine, a 8-bromoguanine, a 8-chloroguanine, a bromothymine, a 8-aminoguanine, a 8-hydroxyguanine, a 8-methylguanine, a 8-thioguanine, an azaguanine, a 2-aminopurine, a 5-ethylcytosine, a 5-methylcyosine, a 5-bromouracil, a 5-ethyluracil, a 5-iodouracil, a 5-chlorouracil, a 5-propyluracil, a thiouracil, a 2-methyladenine, a methylthioadenine, a N,N-diemethyladenine, an azaadenines, a 8-bromoadenine, a 8-hydroxyadenine, a 6-hydroxyaminopurine, a 6-thiopurine, a 4-(6-aminohexyl/cytosine), and the like. Other examples are well known to those of skill in the art.


As used herein, a “nucleoside” refers to an individual chemical unit comprising a nucleobase covalently attached to a nucleobase linker moiety. A non-limiting example of a “nucleobase linker moiety” is a sugar comprising 5-carbon atoms (i.e., a “5-carbon sugar”), including but not limited to a deoxyribose, a ribose, an arabinose, or a derivative or an analog of a 5-carbon sugar. Non-limiting examples of a derivative or an analog of a 5-carbon sugar include a 2′-fluoro-2′-deoxyribose or a carbocyclic sugar where a carbon is substituted for an oxygen atom in the sugar ring. Different types of covalent attachment(s) of a nucleobase to a nucleobase linker moiety are known in the art (Kornberg and Baker, 1992).


As used herein, a “nucleotide” refers to a nucleoside further comprising a “backbone moiety”. A backbone moiety generally covalently attaches a nucleotide to another molecule comprising a nucleotide, or to another nucleotide to form a nucleic acid. The “backbone moiety” in naturally occurring nucleotides typically comprises a phosphorus moiety, which is covalently attached to a 5-carbon sugar. The attachment of the backbone moiety typically occurs at either the 3′- or 5′-position of the 5-carbon sugar. However, other types of attachments are known in the art, particularly when a nucleotide comprises derivatives or analogs of a naturally occurring 5-carbon sugar or phosphorus moiety.


A nucleic acid may comprise, or be composed entirely of, a derivative or analog of a nucleobase, a nucleobase linker moiety and/or backbone moiety that may be present in a naturally occurring nucleic acid. RNA with nucleic acid analogs may also be labeled according to methods of the invention. As used herein a “derivative” refers to a chemically modified or altered form of a naturally occurring molecule, while the terms “mimic” or “analog” refer to a molecule that may or may not structurally resemble a naturally occurring molecule or moiety, but possesses similar functions. As used herein, a “moiety” generally refers to a smaller chemical or molecular component of a larger chemical or molecular structure. Nucleobase, nucleoside and nucleotide analogs or derivatives are well known in the art, and have been described (see for example, Scheit, 1980, incorporated herein by reference).


Additional non-limiting examples of nucleosides, nucleotides or nucleic acids include those in: U.S. Pat. Nos. 5,681,947, 5,652,099 and 5,763,167, 5,614,617, 5,670,663, 5,872,232, 5,859,221, 5,446,137, 5,886,165, 5,714,606, 5,672,697, 5,466,786, 5,792,847, 5,223,618, 5,470,967, 5,378,825, 5,777,092, 5,623,070, 5,610,289, 5,602,240, 5,858,988, 5,214,136, 5,700,922, 5,708,154, 5,728,525, 5,637,683, 6,251,666, 5,480,980, and 5,728,525, each of which is incorporated herein by reference in its entirety.


Labeling methods and kits of the invention specifically contemplate the use of nucleotides that are both modified for attachment of a label and can be incorporated into a miRNA molecule. Such nucleotides include those that can be labeled with a dye, including a fluorescent dye, or with a molecule such as biotin. Labeled nucleotides are readily available; they can be acquired commercially or they can be synthesized by reactions known to those of skill in the art.


Modified nucleotides for use in the invention are not naturally occurring nucleotides, but instead, refer to prepared nucleotides that have a reactive moiety on them. Specific reactive functionalities of interest include: amino, sulfhydryl, sulfoxyl, aminosulfhydryl, azido, epoxide, isothiocyanate, isocyanate, anhydride, monochlorotriazine, dichlorotriazine, mono- or dihalogen substituted pyridine, mono- or disubstituted diazine, maleimide, epoxide, aziridine, sulfonyl halide, acid halide, alkyl halide, aryl halide, alkylsulfonate, N-hydroxysuccinimide ester, imido ester, hydrazine, azidonitrophenyl, azide, 3-(2-pyridyl dithio)-propionamide, glyoxal, aldehyde, iodoacetyl, cyanomethyl ester, p-nitrophenyl ester, o-nitrophenyl ester, hydroxypyridine ester, carbonyl imidazole, and the other such chemical groups. In some embodiments, the reactive functionality may be bonded directly to a nucleotide, or it may be bonded to the nucleotide through a linking group. The functional moiety and any linker cannot substantially impair the ability of the nucleotide to be added to the miRNA or to be labeled. Representative linking groups include carbon containing linking groups, typically ranging from about 2 to 18, usually from about 2 to 8 carbon atoms, where the carbon containing linking groups may or may not include one or more heteroatoms, e.g. S, O, N etc., and may or may not include one or more sites of unsaturation. Of particular interest in many embodiments is alkyl linking groups, typically lower alkyl linking groups of 1 to 16, usually 1 to 4 carbon atoms, where the linking groups may include one or more sites of unsaturation. The functionalized nucleotides (or primers) used in the above methods of functionalized target generation may be fabricated using known protocols or purchased from commercial vendors, e.g., Sigma, Roche, Ambion, Biosearch Technologies and NEN. Functional groups may be prepared according to ways known to those of skill in the art, including the representative information found in U.S. Pat. Nos. 4,404,289; 4,405,711; 4,337,063 and 5,268,486, and U.K. Patent 1,529,202, which are all incorporated by reference.


Amine-modified nucleotides are used in several embodiments of the invention. The amine-modified nucleotide is a nucleotide that has a reactive amine group for attachment of the label. It is contemplated that any ribonucleotide (G, A, U, or C) or deoxyribonucleotide (G, A, T, or C) can be modified for labeling. Examples include, but are not limited to, the following modified ribo- and deoxyribo-nucleotides: 5-(3-aminoallyl)-UTP; 8-[(4-amino)butyl]-amino-ATP and 8-[(6-amino)butyl]-amino-ATP; N6-(4-amino)butyl-ATP, N6-(6-amino)butyl-ATP, N4-[2,2-oxy-bis-(ethylamine)]-CTP; N6-(6-Amino)hexyl-ATP; 8-[(6-Amino)hexyl]-amino-ATP; 5-propargylamino-CTP, 5-propargylamino-UTP; 5-(3-aminoallyl)-dUTP; 8-[(4-amino)butyl]-amino-dATP and 8-[(6-amino)butyl]-amino-dATP; N6-(4-amino)butyl-dATP, N6-(6-amino)butyl-dATP, N4-[2,2-oxy-bis-(ethylamine)]-dCTP; N6-(6-Amino)hexyl-dATP; 8-[(6-Amino)hexyl]-amino-dATP; 5-propargylamino-dCTP, and 5-propargylamino-dUTP. Such nucleotides can be prepared according to methods known to those of skill in the art. Moreover, a person of ordinary skill in the art could prepare other nucleotide entities with the same amine-modification, such as a 5-(3-aminoallyl)-CTP, GTP, ATP, dCTP, dGTP, dTTP, or dUTP in place of a 5-(3-aminoallyl)-UTP.


B. Preparation of Nucleic Acids


A nucleic acid may be made by any technique known to one of ordinary skill in the art, such as for example, chemical synthesis, enzymatic production, or biological production. It is specifically contemplated that miRNA probes of the invention are chemically synthesized.


In some embodiments of the invention, miRNAs are recovered or isolated from a biological sample. The miRNA may be recombinant or it may be natural or endogenous to the cell (produced from the cell's genome). It is contemplated that a biological sample may be treated in a way so as to enhance the recovery of small RNA molecules such as miRNA. U.S. patent application Ser. No. 10/667,126 describes such methods and it is specifically incorporated by reference herein. Generally, methods involve lysing cells with a solution having guanidinium and a detergent.


Alternatively, nucleic acid synthesis is performed according to standard methods. See, for example, Itakura and Riggs (1980) and U.S. Pat. Nos. 4,704,362, 5,221,619, and 5,583,013, each of which is incorporated herein by reference. Non-limiting examples of a synthetic nucleic acid (e.g., a synthetic oligonucleotide), include a nucleic acid made by in vitro chemically synthesis using phosphotriester, phosphite, or phosphoramidite chemistry and solid phase techniques such as described in EP 266,032, incorporated herein by reference, or via deoxynucleoside H-phosphonate intermediates as described by Froehler et al., 1986 and U.S. Pat. No. 5,705,629, each incorporated herein by reference. Various different mechanisms of oligonucleotide synthesis have been disclosed in for example, U.S. Pat. Nos. 4,659,774, 4,816,571, 5,141,813, 5,264,566, 4,959,463, 5,428,148, 5,554,744, 5,574,146, 5,602,244, each of which is incorporated herein by reference.


A non-limiting example of an enzymatically produced nucleic acid include one produced by enzymes in amplification reactions such as PCR™ (see for example, U.S. Pat. Nos. 4,683,202 and 4,682,195, each incorporated herein by reference), or the synthesis of an oligonucleotide described in U.S. Pat. No. 5,645,897, incorporated herein by reference. See also Sambrook et al., 2001, incorporated herein by reference).


Oligonucleotide synthesis is well known to those of skill in the art. Various different mechanisms of oligonucleotide synthesis have been disclosed in for example, U.S. Pat. Nos. 4,659,774, 4,816,571, 5,141,813, 5,264,566, 4,959,463, 5,428,148, 5,554,744, 5,574,146, 5,602,244, each of which is incorporated herein by reference.


Recombinant methods for producing nucleic acids in a cell are well known to those of skill in the art. These include the use of vectors (viral and non-viral), plasmids, cosmids, and other vehicles for delivering a nucleic acid to a cell, which may be the target cell (e.g., a cancer cell) or simply a host cell (to produce large quantities of the desired RNA molecule). Alternatively, such vehicles can be used in the context of a cell free system so long as the reagents for generating the RNA molecule are present. Such methods include those described in Sambrook, 2003, Sambrook, 2001 and Sambrook, 1989, which are hereby incorporated by reference.


C. Isolation of Nucleic Acids


Nucleic acids may be isolated using techniques well known to those of skill in the art, though in particular embodiments, methods for isolating small nucleic acid molecules, and/or isolating RNA molecules can be employed. Chromatography is a process often used to separate or isolate nucleic acids from protein or from other nucleic acids. Such methods can involve electrophoresis with a gel matrix, filter columns, alcohol precipitation, and/or other chromatography. If miRNA from cells is to be used or evaluated, methods generally involve lysing the cells with a chaotropic (e.g., guanidinium isothiocyanate) and/or detergent (e.g., N-lauroyl sarcosine) prior to implementing processes for isolating particular populations of RNA.


In particular methods for separating miRNA from other nucleic acids, a gel matrix is prepared using polyacrylamide, though agarose can also be used. The gels may be graded by concentration or they may be uniform. Plates or tubing can be used to hold the gel matrix for electrophoresis. Usually one-dimensional electrophoresis is employed for the separation of nucleic acids. Plates are used to prepare a slab gel, while the tubing (glass or rubber, typically) can be used to prepare a tube gel. The phrase “tube electrophoresis” refers to the use of a tube or tubing, instead of plates, to form the gel. Materials for implementing tube electrophoresis can be readily prepared by a person of skill in the art or purchased, such as from C.B.S. Scientific Co., Inc. or Scie-Plas.


Methods may involve the use of organic solvents and/or alcohol to isolate nucleic acids, particularly miRNA used in methods and compositions of the invention. Some embodiments are described in U.S. patent application Ser. No. 10/667,126, which is hereby incorporated by reference. Generally, this disclosure provides methods for efficiently isolating small RNA molecules from cells comprising: adding an alcohol solution to a cell lysate and applying the alcohol/lysate mixture to a solid support before eluting the RNA molecules from the solid support. In some embodiments, the amount of alcohol added to a cell lysate achieves an alcohol concentration of about 55% to 60%. While different alcohols can be employed, ethanol works well. A solid support may be any structure, and it includes beads, filters, and columns, which may include a mineral or polymer support with electronegative groups. A glass fiber filter or column has worked particularly well for such isolation procedures.


In specific embodiments, miRNA isolation processes include: a) lysing cells in the sample with a lysing solution comprising guanidinium, wherein a lysate with a concentration of at least about 1 M guanidinium is produced; b) extracting miRNA molecules from the lysate with an extraction solution comprising phenol; c) adding to the lysate an alcohol solution for forming a lysate/alcohol mixture, wherein the concentration of alcohol in the mixture is between about 35% to about 70%; d) applying the lysate/alcohol mixture to a solid support; e) eluting the miRNA molecules from the solid support with an ionic solution; and, f) capturing the miRNA molecules. Typically the sample is dried and resuspended in a liquid and volume appropriate for subsequent manipulation.


V. LABELS AND LABELING TECHNIQUES

In some embodiments, the present invention concerns miRNA that are labeled. It is contemplated that miRNA may first be isolated and/or purified prior to labeling. This may achieve a reaction that more efficiently labels the miRNA, as opposed to other RNA in a sample in which the miRNA is not isolated or purified prior to labeling. In many embodiments of the invention, the label is non-radioactive. Generally, nucleic acids may be labeled by adding labeled nucleotides (one-step process) or adding nucleotides and labeling the added nucleotides (two-step process).


A. Labeling Techniques


In some embodiments, nucleic acids are labeled by catalytically adding to the nucleic acid an already labeled nucleotide or nucleotides. One or more labeled nucleotides can be added to miRNA molecules. See U.S. Pat. No. 6,723,509, which is hereby incorporated by reference.


In other embodiments, an unlabeled nucleotide or nucleotides is catalytically added to a miRNA, and the unlabeled nucleotide is modified with a chemical moiety that enables it to be subsequently labeled. In embodiments of the invention, the chemical moiety is a reactive amine such that the nucleotide is an amine-modified nucleotide. Examples of amine-modified nucleotides are well known to those of skill in the art, many being commercially available such as from Ambion, Sigma, Jena Bioscience, and TriLink.


In contrast to labeling of cDNA during its synthesis, the issue for labeling miRNA is how to label the already existing molecule. The present invention concerns the use of an enzyme capable of using a di- or tri-phosphate ribonucleotide or deoxyribonucleotide as a substrate for its addition to a miRNA. Moreover, in specific embodiments, it involves using a modified di- or tri-phosphate ribonucleotide, which is added to the 3′ end of a miRNA. Enzymes capable of adding such nucleotides include, but are not limited to, poly (A) polymerase, terminal transferase, and polynucleotide phosphorylase. In specific embodiments of the invention, a ligase is contemplated as not being the enzyme used to add the label, and instead, a non-ligase enzyme is employed. Terminal transferase catalyzes the addition of nucleotides to the 3′ terminus of a nucleic acid. Polynucleotide phosphorylase can polymerize nucleotide diphosphates without the need for a primer.


B. Labels


Labels on miRNA or miRNA probes may be colorimetric (includes visible and UV spectrum, including fluorescent), luminescent, enzymatic, or positron emitting (including radioactive). The label may be detected directly or indirectly. Radioactive labels include 125I, 32P, 33P, and 35S. Examples of enzymatic labels include alkaline phosphatase, luciferase, horseradish peroxidase, and β-galactosidase. Labels can also be proteins with luminescent properties, e.g., green fluorescent protein and phycoerythrin.


The colorimetric and fluorescent labels contemplated for use as conjugates include, but are not limited to, Alexa Fluor dyes, BODIPY dyes, such as BODIPY FL; Cascade Blue; Cascade Yellow; coumarin and its derivatives, such as 7-amino-4-methylcoumarin, aminocoumarin and hydroxycoumarin; cyanine dyes, such as Cy3 and Cy5; eosins and erythrosins; fluorescein and its derivatives, such as fluorescein isothiocyanate; macrocyclic chelates of lanthanide ions, such as Quantum Dye™; Marina Blue; Oregon Green; rhodamine dyes, such as rhodamine red, tetramethylrhodamine and rhodamine 6G; Texas Red; fluorescent energy transfer dyes, such as thiazole orange-ethidium heterodimer; and, TOTAB.


Specific examples of dyes include, but are not limited to, those identified above and the following: Alexa Fluor 350, Alexa Fluor 405, Alexa Fluor 430, Alexa Fluor 488, Alexa Fluor 500. Alexa Fluor 514, Alexa Fluor 532, Alexa Fluor 546, Alexa Fluor 555, Alexa Fluor 568, Alexa Fluor 594, Alexa Fluor 610, Alexa Fluor 633, Alexa Fluor 647, Alexa Fluor 660, Alexa Fluor 680, Alexa Fluor 700, and, Alexa Fluor 750; amine-reactive BODIPY dyes, such as BODIPY 493/503, BODIPY 530/550, BODIPY 558/568, BODIPY 564/570, BODIPY 576/589, BODIPY 581/591, BODIPY 630/650, BODIPY 650/655, BODIPY FL, BODIPY R6G, BODIPY TMR, and, BODIPY-TR; Cy3, Cy5, 6-FAM, Fluorescein Isothiocyanate, HEX, 6-JOE, Oregon Green 488, Oregon Green 500, Oregon Green 514, Pacific Blue, REG, Rhodamine Green, Rhodamine Red, Renographin, ROX, SYPRO, TAMRA, 2′,4′,5′,7′-Tetrabromosulfonefluorescein, and TET.


Specific examples of fluorescently labeled ribonucleotides are available from Molecular Probes, and these include, Alexa Fluor 488-5-UTP, Fluorescein-12-UTP, BODIPY FL-14-UTP, BODIPY TMR-14-UTP, Tetramethylrhodamine-6-UTP, Alexa Fluor 546-14-UTP, Texas Red-5-UTP, and BODIPY TR-14-UTP. Other fluorescent ribonucleotides are available from Amersham Biosciences, such as Cy3-UTP and Cy5-UTP.


Examples of fluorescently labeled deoxyribonucleotides include Dinitrophenyl (DNP)-11-dUTP, Cascade Blue-7-dUTP, Alexa Fluor 488-5-dUTP, Fluorescein-12-dUTP, Oregon Green 488-5-dUTP, BODIPY FL-14-dUTP, Rhodamine Green-5-dUTP, Alexa Fluor 532-5-dUTP, BODIPY TMR-14-dUTP, Tetramethylrhodamine-6-dUTP, Alexa Fluor 546-14-dUTP, Alexa Fluor 568-5-dUTP, Texas Red-12-dUTP, Texas Red-5-dUTP, BODIPY TR-14-dUTP, Alexa Fluor 594-5-dUTP, BODIPY 630/650-14-dUTP, BODIPY 650/665-14-dUTP; Alexa Fluor 488-7-OBEA-dCTP, Alexa Fluor 546-16-OBEA-dCTP, Alexa Fluor 594-7-OBEA-dCTP, Alexa Fluor 647-12-OBEA-dCTP.


It is contemplated that nucleic acids may be labeled with two different labels. Furthermore, fluorescence resonance energy transfer (FRET) may be employed in methods of the invention (e.g., Klostermeier et al., 2002; Emptage, 2001; Didenko, 2001, each incorporated by reference).


Alternatively, the label may not be detectable per se, but indirectly detectable or allowing for the isolation or separation of the targeted nucleic acid. For example, the label could be biotin, digoxigenin, polyvalent cations, chelator groups and the other ligands, include ligands for an antibody.


C. Visualization Techniques


A number of techniques for visualizing or detecting labeled nucleic acids are readily available. Such techniques include, microscopy, arrays, Fluorometry, Light cyclers or other real time PCR machines, FACS analysis, scintillation counters, Phosphoimagers, Geiger counters, MRI, CAT, antibody-based detection methods (Westerns, immunofluorescence, immunohistochemistry), histochemical techniques, HPLC (Griffey et al., 1997), spectroscopy, capillary gel electrophoresis (Cummins et al., 1996), spectroscopy; mass spectroscopy; radiological techniques; and mass balance techniques.


When two or more differentially colored labels are employed, fluorescent resonance energy transfer (FRET) techniques may be employed to characterize association of one or more nucleic acid. Furthermore, a person of ordinary skill in the art is well aware of ways of visualizing, identifying, and characterizing labeled nucleic acids, and accordingly, such protocols may be used as part of the invention. Examples of tools that may be used also include fluorescent microscopy, a BioAnalyzer, a plate reader, Storm (Molecular Dynamics), Array Scanner, FACS (fluorescent activated cell sorter), or any instrument that has the ability to excite and detect a fluorescent molecule.


VI. KITS

Any of the compositions described herein may be comprised in a kit. In a non-limiting example, reagents for isolating miRNA, labeling miRNA, and/or evaluating a miRNA population using an array, nucleic acid amplification, and/or hybridization can be included in a kit, as well reagents for preparation of samples from blood samples. The kit may further include reagents for creating or synthesizing miRNA probes. The kits will thus comprise, in suitable container means, an enzyme for labeling the miRNA by incorporating labeled nucleotide or unlabeled nucleotides that are subsequently labeled. In certain aspects, the kit can include amplification reagents. In other aspects, the kit may include various supports, such as glass, nylon, polymeric beads, and the like, and/or reagents for coupling any probes and/or target nucleic acids. It may also include one or more buffers, such as reaction buffer, labeling buffer, washing buffer, or a hybridization buffer, compounds for preparing the miRNA probes, and components for isolating miRNA. Other kits of the invention may include components for making a nucleic acid array comprising miRNA, and thus, may include, for example, a solid support.


Kits for implementing methods of the invention described herein are specifically contemplated. In some embodiments, there are kits for preparing miRNA for multi-labeling and kits for preparing miRNA probes and/or miRNA arrays. In these embodiments, kit comprise, in suitable container means, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more of the following: (1) poly(A) polymerase; (2) unmodified nucleotides (G, A, T, C, and/or U); (3) a modified nucleotide (labeled or unlabeled); (4) poly(A) polymerase buffer; and, (5) at least one microfilter; (6) label that can be attached to a nucleotide; (7) at least one miRNA probe; (8) reaction buffer; (9) a miRNA array or components for making such an array; (10) acetic acid; (11) alcohol; (12) solutions for preparing, isolating, enriching, and purifying miRNAs or miRNA probes or arrays. Other reagents include those generally used for manipulating RNA, such as formamide, loading dye, ribonuclease inhibitors, and DNase.


In specific embodiments, kits of the invention include an array containing miRNA probes, as described in the application. An array may have probes corresponding to all known miRNAs of an organism or a particular tissue or organ in particular conditions, or to a subset of such probes. The subset of probes on arrays of the invention may be or include those identified as relevant to a particular diagnostic, therapeutic, or prognostic application. For example, the array may contain one or more probes that is indicative or suggestive of (1) a disease or condition (acute myeloid leukemia), (2) susceptibility or resistance to a particular drug or treatment; (3) susceptibility to toxicity from a drug or substance; (4) the stage of development or severity of a disease or condition (prognosis); and (5) genetic predisposition to a disease or condition.


For any kit embodiment, including an array, there can be nucleic acid molecules that contain or can be used to amplify a sequence that is a variant of, identical to or complementary to all or part of any of SEQ IDs described herein. In certain embodiments, a kit or array of the invention can contain one or more probes for the miRNAs identified by the SEQ IDs described herein. Any nucleic acid discussed above may be implemented as part of a kit.


The components of the kits may be packaged either in aqueous media or in lyophilized form. The container means of the kits will generally include at least one vial, test tube, flask, bottle, syringe or other container means, into which a component may be placed, and preferably, suitably aliquoted. Where there is more than one component in the kit (labeling reagent and label may be packaged together), the kit also will generally contain a second, third or other additional container into which the additional components may be separately placed. However, various combinations of components may be comprised in a vial. The kits of the present invention also will typically include a means for containing the nucleic acids, and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow molded plastic containers into which the desired vials are retained.


When the components of the kit are provided in one and/or more liquid solutions, the liquid solution is an aqueous solution, with a sterile aqueous solution being particularly preferred.


However, the components of the kit may be provided as dried powder(s). When reagents and/or components are provided as a dry powder, the powder can be reconstituted by the addition of a suitable solvent. It is envisioned that the solvent may also be provided in another container means. In some embodiments, labeling dyes are provided as a dried power. It is contemplated that 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 120, 120, 130, 140, 150, 160, 170, 180, 190, 200, 300, 400, 500, 600, 700, 800, 900, 1000 μg or at least or at most those amounts of dried dye are provided in kits of the invention. The dye may then be resuspended in any suitable solvent, such as DMSO.


Such kits may also include components that facilitate isolation of the labeled miRNA. It may also include components that preserve or maintain the miRNA or that protect against its degradation. Such components may be RNAse-free or protect against RNAses. Such kits generally will comprise, in suitable means, distinct containers for each individual reagent or solution.


A kit will also include instructions for employing the kit components as well the use of any other reagent not included in the kit. Instructions may include variations that can be implemented.


Kits of the invention may also include one or more of the following: Control RNA; nuclease-free water; RNase-free containers, such as 1.5 ml tubes; RNase-free elution tubes; PEG or dextran; ethanol; acetic acid; sodium acetate; ammonium acetate; guanidinium; detergent; nucleic acid size marker; RNase-free tube tips; and RNase or DNase inhibitors.


It is contemplated that such reagents are embodiments of kits of the invention. Such kits, however, are not limited to the particular items identified above and may include any reagent used for the manipulation or characterization of miRNA.


VII. EXAMPLES

The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.


Example 1
Gene Expression Analysis in HL-60 Cells Following Electroporation with Hsa-miR-10a

miRNAs are believed to regulate gene expression by binding to target mRNA transcripts and (1) initiating transcript degradation or (2) altering protein translation from the transcript. Translational regulation leading to an up or down change in protein expression may lead to changes in activity and expression of downstream gene products and genes that are in turn regulated by those proteins. These numerous regulatory effects may be revealed as changes in the global mRNA expression profile. Microarray gene expression analyses were performed to identify genes that are mis-regulated by hsa-miR-10a expression.


Synthetic Pre-miR™-hsa-miR-10a (Ambion, Austin, Tex., USA) or one of two negative control (NC) miRNAs (Pre-miR™ microRNA Precursor Molecule-Negative Control #1, Ambion, cat. no. AM17110 and Pre-miR™ microRNA Precursor Molecule-Negative Control #2, Ambion, cat. no. AM17111) were transfected into quadruplicate samples of HL-60 cells (peripheral blood promyeloblasts; American Type Culture Collection (ATCC), Manassas, Va., USA) using electroporation. Cells (500,000) were electroporated with 5 μg miRNA in a 2-mm cuvette in a total reaction volume of 150 μl. A Gene Pulser Xcell System (Bio-Rad Laboratories, Hercules, Calif., USA) was used to generate a single square wave pulse of 25 milliseconds at 140 V for electroporations. Cells were grown in RPMI1640 medium containing 10% fetal bovine serum and 1× penicillin/streptomycin solution (ATCC) and harvested for RNA isolation 72 hours post electroporation. Total RNA was extracted using RNAqueous-4PCR (Ambion) according to the manufacturer's recommended protocol.


mRNA array analyses were performed by Asuragen Services (Austin, Tex.), according to the company's standard operating procedures. Using the MessageAmp™ II-96 aRNA Amplification Kit (Ambion, cat #1819), 2 μg of total RNA were used for target preparation and labelling with biotin. cRNA yields were quantified using an Agilent Bioanalyzer 2100 capillary electrophoresis protocol. Labelled target was hybridized to Affymetrix mRNA arrays (Human HG-U133A 2.0 arrays) using the manufacturer's recommendations and the following parameters. Hybridizations were carried out at 45° C. for 16 hours in an Affymetrix Model 640 hybridization oven. Arrays were washed and stained on an Affymetrix FS450 Fluidics station, running the wash script Midi_euk2v3450. The arrays were scanned on an Affymetrix GeneChip Scanner 3000. Summaries of the image signal data, group mean values, p-values with significance flags, log ratios and gene annotations for every gene on the array were generated using the Affymetrix Statistical Algorithm MAS 5.0 (GCOS v1.4). Data were normalized for the effect observed by the average of two negative control microRNA sequences and then were averaged together for presentation. Genes altered by treatment were determined by filtering all genes by fold-change relative to the two control transfections. Statistical significance was assessed by a t-test after the omnibus F-test was shown to be significant. A list of genes whose expression levels varied by at least 2-fold from the negative control was assembled. Results of the microarray gene expression analysis are shown in Table 1.









TABLE 1







Genes with increased (positive values) or decreased (negative values)


expression following transfection of HL-60 cells with pre-miR hsa-miR-10a.









Gene




Symbol
RefSeq (Pruitt et al., 2005)
Fold Change












FLJ37549
AK094868, NM_152605
5.90


AL833920,
AL833920, AL834308
4.95


AL834308


C20orf27
BC024036, NM_001039140, NM_017874
4.86


RNH1
NM_002939
4.82


RIPK5
AF068286
4.75


AKAP5
NM_004857
4.69


TAF7L
AK026810, BC043391, NM_024885
4.41


TIGD4
BC037869, NM_145720
4.18


AY358413,
AK027395, AY358413, BC063012, CR593410,
4.16


CR593410,
NM_031476


CRISPLD2


NPM2
BC068078, NM_182795
4.13


JUNB
NM_002229
4.11


XAB2
BC007208, NM_020196
4.07


CCDC57
BC110997, NM_198082
4.04


SOLH
NM_005632
4.03


DVL1,
AK093189, NM_004421, NM_181870, NM_182779,
3.88


DVL1L1
U46461


ZNF585B
NM_152279
3.81


DNAJB5
BC012115, NM_012266
3.71


LPA
NM_005577
3.70


PTHLH
NM_198965
3.70


NR2F2
BC014664, BC106083
3.65


BC104473
BC104473
3.55


LOC153561
BC067830
3.55


PKHD1
AY074797, AY092083, NM_138694, NM_170724
3.52


EPB41L4B
AB032179
3.48


AOC3
NM_003734
3.47


AGER
AB061669, AY755620, AY755621, AY755622,
3.37



AY755623, AY755624, AY755625, AY755627,



NM_001136


C7orf20
NM_015949
3.36


TUBGCP6
NM_020461
3.33


N-PAC
BC032855, NM_032569
3.32


CCL16
NM_004590
3.30


POF1B
AF309774, AK025039, AK026445, NM_024921
3.30


RABIF
NM_002871
3.22


TOLLIP
AK096693, NM_019009
3.22


AYP1
NM_032193
3.21


BC007360
BC007360
3.21


CEECAM1
AF177203, AF218016, NM_016174
3.21


SLC7A13
NM_138817
3.21


BC029609
BC029609
3.18


GRID1
BC039263
3.17


NALP12
AK095460, AY116204, AY116205, NM_144687
3.17


GPR176
NM_007223
3.14


FAM9A
NM_174951
3.09


IGSF4B
NM_021189
3.09


PPP1R15A
NM_014330
3.05


BOLA1
NM_016074
3.04


FLJ13273
NM_001031720
3.01


FAM73B
AK054974, AK075421, BC009114, NM_032809
2.99


PGK2
NM_138733
2.97


SLC19A3
NM_025243
2.97


KRTAP10-
NM_198699
2.94


12


POLR2E
NM_002695
2.94


TNP2
NM_005425
2.93


RAD23A
BC088364, NM_005053
2.92


CD151
NM_004357
2.91


SMOX
AK000753, NM_019025, NM_175840, NM_175842
2.91


MGC35048
NM_153208
2.89


DCN
NM_001920
2.88


SLC2A3
NM_006931
2.88


AK126887
AK126887
2.84


CA3
NM_005181
2.81


SLC4A9
NM_031467
2.79


ATP13A4
AY358110, NM_032279
2.78


MGC13040
NM_032930
2.78


C1orf88,
BC050319, CR609935, NM_181643
2.77


CR609935


C6orf32
AB002384
2.77


ZBTB39
NM_014830
2.74


C9orf52
BC038592, NM_152574
2.72


POLR2C
NM_032940
2.72


CNTN1
NM_001843, NM_175038
2.71


FLJ40365
NM_173482
2.71


RABEPK
AL832249
2.71


URP2
NM_031471, NM_178443
2.71


SHC4
AY358250, NM_203349
2.69


ZNF713
NM_182633
2.66


EYA3
BX648945, NM_001990
2.65


LZTS2
AY029201, NM_032429
2.65


ZNF675
AK093669, NM_138330
2.63


CR612105
CR612105
2.62


PSIP1
BC044568, BC064135, NM_021144
2.62


FRAS1
NM_206841
2.61


AY358160
AY358160
2.58


CCL7
BC092436, NM_006273
2.56


HLA-
NM_033554
2.56


DPA1


FLJ13646
BC066936, NM_024584
2.52


MYRIP
BC109311, BC109312, NM_015460
2.50


PCM1
BC000453
2.49


RAB30
BX641138
2.49


SLC2A12
BC070149, NM_145176
2.49


BC016820
BC016820
2.47


CR936613
CR936613
2.47


LOC124842
NM_207313
2.47


ZC3H5
BC053362
2.46


GIPC2
BC036075, NM_017655
2.45


USP10
CR749515, NM_005153
2.45


COPS2
AB209799, NM_004236
2.44


KLHL26
NM_018316
2.44


LEFTY1
NM_020997
2.44


P8
NM_012385
2.44


IL17RB
NM_018725
2.41


PMCHL2
NM_031888
2.40


PLEKHN1
BC101386, BC101387, NM_032129
2.39


RBM9
AK055213
2.38


SYNGAP1
NM_006772
2.38


VKORC1L1
NM_173517
2.38


ZNF583
NM_152478
2.38


ACOT4
NM_152331
2.37


IFNA4
NM_021068
2.37


C9orf47
AK094842, AK127854, NM_001001938
2.34


CREBL1
NM_004381, U31903
2.34


TSPAN32
AK128812, BC016693, NM_139022
2.34


CWF19L1
AK023984
2.33


LAIR1
AF251510
2.33


BX648310,
BX648310, NM_001031696, NM_012268
2.32


PLD3


CR602282,
BC024007, CR602282
2.29


CTBS


AY732486,
AY732486, CR600380, NM_000062
2.28


SERPING1


DKFZP781I1119
BX537798, CR749246, NM_152622
2.28


GAA
NM_000152
2.28


RAB3A
NM_002866
2.28


ZNF23
BC030658, BC048974, BX537507, NM_145911
2.28


GPT2
BC062555, NM_133443
2.26


IHPK3
NM_054111
2.25


MTCP1
NM_001018025, NM_014221
2.25


JAM3
BC012147, NM_032801
2.24


AK054645,
AK054645, NM_033655
2.23


CNTNAP3


ARMC7
NM_024585
2.23


C17orf66
BC033734, NM_152781
2.23


TMEM105
NM_178520
2.23


MINK1
AL157418
2.22


MLCK
BC109097, NM_182493
2.22


MGC33302
NM_152778
2.19


PRTG
AY630258, NM_173814
2.19


TNFRSF18
NM_004195, NM_148901, NM_148902
2.19


AK001998
AK001998
2.17


AK097976
AK097976
2.17


OPRM1
NM_001008503, NM_001008505
2.16


KRT18
NM_000224
2.15


LOC144363
NM_001001660
2.14


REG3A
NM_138938
2.14


SLC7A1
NM_003045
2.14


CCL23
NM_005064, NM_145898
2.12


DCUN1D2
AK001566, NM_001014283
2.12


TNFSF13
NM_172088
2.12


ELOVL2
NM_017770
2.11


GADD45B
AF087853
2.11


LDHAL6A
NM_144972
2.10


TRIM50B
BC033812
2.10


ATRX
NM_000489, NM_138270, NM_138271, U72936,
2.09



U72937, U72938


DKFZP564O0523
NM_032120
2.06


RIN3
AK026092
2.03


COQ7
AL136647
2.02


STRA13
NM_144998, U95007
2.02


C1QTNF3
NM_030945, NM_181435
2.01


MRPS5
NM_031902
2.01


RAB28
BC018067, NM_001017979
2.01


SF3A2
NM_007165
2.00


ZNF226
NM_001032374
2.00


KIAA0261
AF479418
−2.00


KIF2C
AY026505
−2.00


THBS4
Z19585
−2.00


CEBPE
NM_001805
−2.01


FAM19A4
NM_182522
−2.01


VAT1
NM_006373
−2.01


ERGIC1
BC012766, NM_001031711
−2.02


EWSR1
BC000527
−2.02


LCP1
AK223305, NM_002298
−2.02


IFITM2
NM_006435
−2.03


RHOF
NM_019034
−2.03


HSP90AB1
AF275719, BC012807, NM_007355
−2.04


JOSD1
NM_014876
−2.04


LSM7
NM_016199
−2.04


RAB32
NM_006834
−2.04


CAD
NM_004341
−2.05


MAD2L2
NM_006341
−2.05


MRPL38
NM_032478
−2.05


UROS
NM_000375
−2.05


KATNA1
NM_007044
−2.06


ARF1
NM_001024227
−2.07


MCM7
AF279900, BC013375, NM_005916,
−2.07



NM_182776


MYL4
NM_001002841
−2.08


AK097280,
AK097280, BC017344, NM_024710
−2.10


ISOC2


ARPC5,
BC057237, BC071857, NM_005717
−2.10


BC071857


C18orf24
NM_145060
−2.10


HNRPF
BC016736, NM_004966
−2.10


DNM2
AK097875, AK124881, NM_001005360,
−2.11



NM_001005361, NM_001005362, NM_004945


AK127860,
AK127860, NM_002649
−2.12


PIK3CG


DHX30
NM_138614
−2.12


RASGRP2
AK092882, NM_005825, NM_153819
−2.12


RPRC1
AK095939, BC003083, BC106053, NM_018067
−2.12


THEM4
NM_053055
−2.12


TINF2
NM_012461
−2.12


TMEM102
NM_178518
−2.12


AK127692,
AK127692, NM_175614
−2.13


NDUFA11


FADS1
AK074754, AL512760, NM_013402
−2.13


FLJ12716
BC022185
−2.13


GGT6
BC063111, NM_153338
−2.13


ZNF667
AK126957, NM_022103
−2.13


BC062751,
BC062751, NM_006859, NM_194451
−2.14


LIAS


BRCA2
NM_000059
−2.14


METAP2
AK091730, NM_006838
−2.14


TRPV2
NM_016113
−2.14


IDH2
NM_002168
−2.15


MLSTD2
NM_032228
−2.15


UPF3A
BC023569, NM_023011, NM_080687
−2.15


CANX
AK129990, NM_001746
−2.16


CAPNS1
BC011903, NM_001749
−2.16


CDT1
NM_030928
−2.16


FMNL1
NM_005892
−2.16


PTPN7
AK127214, NM_002832, NM_080588
−2.16


AK124968
AK124968
−2.18


CCL5
NM_002985
−2.18


MYADM
BC013995, NM_001020818
−2.18


PPIE
DQ160195, NM_203456
−2.18


PPP1CA
CR595463, NM_001008709, NM_002708
−2.18


PRPF39
AK001990, NM_017922
−2.18


CLDND1
AF161522
−2.19


KIAA0152
NM_014730
−2.19


MCM3
NM_002388
−2.19


MTA1
BC006177
−2.19


PRDX5
AF124993, NM_012094
−2.19


ARHGDIA
NM_004309
−2.20


C4orf16
NM_018569
−2.20


CYC1
NM_001916
−2.20


AK123945,
AK123945, NM_019023
−2.21


PRMT7


CAPG
NM_001747
−2.21


GNB2
NM_005273
−2.21


PTBP1
NM_002819, NM_031990, NM_031991
−2.21


FIS1
NM_016068
−2.22


POLR1C
NM_004875
−2.22


SCML4
AK093571
−2.23


COPZ1
NM_016057
−2.24


MAP2K3
BC032478, NM_145109
−2.24


VIM
AK093924, NM_003380
−2.25


GANAB
BC065266, NM_198334, NM_198335
−2.26


PRKCD
NM_006254
−2.26


TSTA3
NM_003313
−2.26


FLJ31413
BC068505, NM_152557
−2.27


FNTA
NM_002027
−2.27


MYO1G
NM_033054
−2.27


NR2F1
NM_005654
−2.27


RABAC1
NM_006423
−2.27


AK3
BC013771, NM_016282
−2.28


CORO1A
AB209221, NM_007074
−2.28


ERAL1
NM_005702
−2.28


RABGGTB
NM_004582
−2.28


AB064670
AB064670
−2.29


C6orf108
NM_006443
−2.29


ELK1
AK093966, BC056150
−2.29


EVL
BC023997, NM_016337
−2.29


FLJ11305
BC016614, BC031246
−2.29


PEX26
NM_017929
−2.29


GMDS
AF040260, BC000117, NM_001500
−2.30


DDX11
BC011264, BC050069, NM_030653, U75968
−2.31


AK097197,
AK097197, NM_004712
−2.32


HGS


AK125522,
AK125522, NM_004691
−2.32


ATP6V0D1


COMT
BC000419, NM_000754, NM_007310
−2.32


PCOLN3
BC007527, CR618565
−2.32


CLIC5
BC020923
−2.33


KARS
AF285758
−2.33


MATK
NM_002378, NM_139354, NM_139355
−2.33


SHMT2
BC011911, BC032584, NM_005412
−2.33


VAV1
BC013361, NM_005428
−2.33


TBC1D2B
BC033712, NM_015079
−2.34


TIMELESS
NM_003920
−2.34


TUBGCP2
BC111957, NM_006659
−2.34


AK124363,
AJ419866, AK124363, NM_003730
−2.35


RNASET2


ARRB2
AK097542, BC067368, NM_004313, NM_199004
−2.35


AZU1
NM_001700
−2.35


C1orf160
BC011579, BC090039, NM_032125
−2.35


MASP1
NM_001031849, NM_001879
−2.35


ACOT8
NM_005469, NM_183386
−2.36


C1orf71
AL834246
−2.36


LEMD2
BC039864
−2.36


EIF3S4
NM_003755
−2.37


MBNL1
BC050535, NM_021038, NM_207292, NM_207293,
−2.38



NM_207295, NM_207296, NM_207297


AF242772
AF242772
−2.39


TKT
NM_001064
−2.39


COG3
NM_031431
−2.40


SLC25A1
NM_005984
−2.40


SMG7
AB085674, BC036381, NM_014837, NM_173156,
−2.40



NM_201568, NM_201569


FAM54B
AF173891, AK056721, BC017175, NM_019557
−2.41


FAM73A
NM_198549
−2.41


hCAP-D3
BC098398, NM_015261
−2.41


PDCL3
NM_024065
−2.41


LSM4
NM_012321
−2.42


POLG
NM_002693
−2.42


TFDP1
NM_007111
−2.42


C12orf10
BC051871, NM_021640
−2.43


AP1S1
CR599373, NM_057089
−2.44


COTL1
AK127352, NM_021149
−2.44


CYBA
NM_000101
−2.44


RNF166
AK057201, NM_178841
−2.44


PPP3R1
NM_000945
−2.45


ZNF207
BC008023, CR616570, NM_001032293, NM_003457
−2.46


NUP210
AB020713, NM_024923
−2.47


RFWD3
AK022673, BC002574, NM_018124
−2.47


PSMD8
NM_002812
−2.48


CEBPA
BC063874, NM_004364
−2.49


DCXR
NM_016286
−2.50


IFNGR1
NM_000416
−2.50


PA2G4
BC069786, NM_006191
−2.50


PRMT1
AY775289, NM_001536, NM_198318, NM_198319
−2.50


ALG1
NM_019109
−2.51


AF090928
AF090928
−2.52


BZRP
NM_000714, NM_007311
−2.52


MIB1
AY147849, NM_020774
−2.53


NDUFA10
NM_004544
−2.53


PLEKHB2
NM_017958
−2.53


UQCRC2
NM_003366
−2.53


CDC37
NM_007065
−2.54


EMP3
NM_001425
−2.54


HBS1L
AJ420551, AJ459827, AL137664, NM_006620
−2.54


HCLS1
NM_005335
−2.54


ACTN4
D89980, NM_004924
−2.55


ARHGAP30
AK126163, BC025732, BC053688, BX537846,
−2.55



NM_001025598, NM_181720


ATP5D
NM_001687
−2.55


FUS
CR598388, NM_004960, X71428
−2.55


PSMC3
BC106920, NM_002804
−2.55


PDZD8
NM_173791
−2.56


RBBP4
NM_005610
−2.56


FTL
BC067772
−2.57


FLNA
BC014654
−2.58


PTP4A1
NM_003463
−2.58


SDHC
NM_001035511, NM_001035512, NM_001035513,
−2.58



NM_003001


RCC1
BC069198, NM_001269
−2.59


ALOX5AP
NM_001629
−2.60


P15RS
BC000225, NM_018170
−2.63


ABCF2
NM_005692
−2.64


PAK2
NM_002577
−2.64


CKAP5
BC035554
−2.65


MGC15416
NM_138418
−2.65


NDNL2
NM_138704
−2.65


VASP
BC026019, NM_003370
−2.65


AK128704,
AK128704, NM_006427
−2.67


SIVA


CR603690,
AJ224442, BC001780, CR603690, NM_017528
−2.67


WBSCR22


FASN
NM_004104
−2.67


ITPKB
NM_002221
−2.67


METRN
NM_024042
−2.68


MGC72104
NM_207350
−2.69


PODXL2
BC019330, NM_015720
−2.69


PSENEN
NM_172341
−2.69


BCAP31
NM_005745
−2.70


C14orf104
NM_018139
−2.70


MRPL23
NM_021134
−2.70


PCBP1
NM_006196
−2.70


MGC13114
BC007207
−2.71


RPS9
NM_001013
−2.71


AK125797,
AK125797, D86988, NM_002911
−2.72


RENT1,


UPF1


PSMD3
AK055799, AK094206, NM_002809
−2.72


GRN
AK023348, NM_002087
−2.73


STAT3
NM_003150, NM_139276
−2.73


AK126566,
AK126566, NM_001398
−2.75


ECH1


BCL7C
BC058863, NM_004765
−2.75


NAPRT1
AF258565, AY214327, BC006284, NM_145201
−2.75


C19orf25
BC018441, NM_152482
−2.76


MAGOH
NM_002370
−2.76


PGD
NM_002631
−2.76


SDHB
NM_003000
−2.76


AK124276,
AK124276, CR456537, NM_001003891, NM_015889
−2.77


PCQAP


BTG3
BC011957, CR607894
−2.77


SELO
BC110866, NM_031454
−2.77


IL17R
NM_014339
−2.79


ZNF313
NM_018683
−2.81


GGA1
CR456493, NM_001001560, NM_001001561,
−2.82



NM_013365


MGC33556
NM_001004307
−2.82


AY129014,
AY129014, CR597452, NM_006643
−2.83


CR597452,


SDCCAG3


CD99
U82164
−2.83


IGFBP2
NM_000597
−2.84


IFITM3
NM_021034
−2.85


TXLNA
BC046565, NM_175852
−2.85


WDR18
NM_024100
−2.85


BC071982,
BC071982, NM_006854
−2.86


KDELR2


SLC16A3
NM_004207
−2.86


SSR2
BC000341, NM_003145
−2.86


AP2B1
AY341427, BC006201
−2.87


KIAA2010
AB095930, BC028088, BC038932, BC072409,
−2.88



NM_017936, NM_032560


AP2M1
NM_001025205
−2.89


C1orf37,
CR617405, NM_138391
−2.94


CR617405


ASL
AY203938, CR603567, NM_001024943,
−2.95



NM_001024944, NM_001024946


SNRPA
NM_004596
−3.00


SNX17
NM_014748
−3.00


DGUOK
BC015757, NM_080916, NM_080917, NM_080918,
−3.02



X97386


TCF8
AK091478, NM_030751
−3.02


PSMB8
NM_004159, NM_148919
−3.05


PKN1,
BC040061, BC094766, NM_002741, NM_213560,
−3.06


X80229
X80229


PRKCSH
NM_002743
−3.10


SLC25A6
NM_001636
−3.10


DNASE1
NM_005223
−3.11


ASNA1
NM_004317
−3.12


SLC7A5
NM_003486
−3.14


NUTF2
NM_005796
−3.16


RPUSD1
NM_058192
−3.16


TUBA2
NM_006001, NM_079836
−3.16


PGK1
NM_000291
−3.18


OBFC2B
NM_024068
−3.19


IGLL1
NM_020070
−3.20


UBE2S
NM_014501
−3.20


CALR
NM_004343
−3.24


EEF1D
BC094806, NM_001960, NM_032378
−3.25


RKHD1
AB107353, NM_203304
−3.25


HNRPDL
AB066484
−3.26


EDEM1
NM_014674
−3.27


SRM
NM_003132
−3.29


DDB1
NM_001923
−3.30


FLJ36070
AK131427, BC106047, NM_182574
−3.36


AHCYL1
AF315687, AK131563, NM_006621
−3.37


LOC115098
NM_138442
−3.37


TOR3A
AJ299441
−3.37


CHTF18
BC006437, NM_022092
−3.41


CR614462,
CR614462, NM_020470
−3.41


YIF1A


LARP1
NM_033551
−3.42


STT3A
NM_152713
−3.44


FH
NM_000143
−3.51


C12orf57
NM_138425
−3.56


DDX50
NM_024045
−3.57


DDOST
D29643
−3.61


UBE2M
NM_003969
−3.67


TYMS
AB077208
−3.71


PSMB1
BC020807
−3.78


HSPD1
CR619688, NM_002156
−3.79


MRPL12
AF105278, NM_002949
−3.81


EIF3S2
BC003140, NM_003757
−3.84


AP2S1
NM_004069, NM_021575
−3.86


FKSG30
NM_001017421
−3.91


AHCY
M61831, NM_000687
−3.94


PRMT5,
CR625282, NM_006109
−3.95


SKB1


CR617382,
BC089438, BX247961, CR617382
−3.96


HNRPC


SNRPB
NM_198216
−3.99


SPG7
NM_003119
−4.01


TOMM22
NM_020243
−4.13


GLT25D1
BC108308, NM_024656
−4.14


CTBP1
NM_001012614, NM_001328
−4.15


PRTN3
M29142, NM_002777
−4.20


THOC4
NM_005782
−4.43









Negative fold change values in Table 1 indicate a reduction in mRNA levels for a given gene compared to that observed for the negative controls.


Manipulation of the expression levels of the genes listed in Table 1 represents a potentially useful therapy for cancer and other diseases in which increased or reduced expression of hsa-miR-10a has a role in the disease.


Example 2
Cellular Pathways Affected by Hsa-miR-10a

The mis-regulation of gene expression by hsa-miR-10a (Table 1) affects many cellular pathways that represent potential therapeutic targets for the control of cancer and other diseases and disorders. The inventors determined the identity and nature of the cellular genetic pathways affected by the regulatory cascade induced by hsa-miR-10a expression. Cellular pathway analyses were performed using Ingenuity Pathways Analysis (Version 4.0, Ingenuity Systems; Redwood City, Calif., USA). Alteration of a given pathway was determined by Fisher's Exact test (Fisher, 1922). The most significantly affected pathways following over-expression of hsa-miR-10a in HL-60 cells are shown in Table 2.









TABLE 2







Significantly affected functional cellular pathways following


hsa-miR-10a overexpression in HL-60 cells.








Number



of Genes
Pathway Functions





27
Cellular Movement, Cell-To-Cell Signaling and



Interaction, Hematological System



Development and Function


26
Cellular Growth and Proliferation, Cancer,



Gastrointestinal Disease


24
Molecular Transport, Cellular Assembly and



Organization, Cellular Movement


17
Post-Translational Modification, Gene



Expression, Cell Signaling


17
Cellular Assembly and Organization, Cellular



Function and Maintenance, Lipid



Metabolism


16
Cellular Assembly and Organization, DNA



Replication, Recombination, and Repair,



RNA Post-Transcriptional Modification


16
Cell Cycle, Cancer, Endocrine System Disorders


16
Amino Acid Metabolism, Post-Translational



Modification, Small Molecule



Biochemistry


16
Protein Synthesis, Lipid Metabolism,



Molecular Transport


15
Cell Cycle, Gene Expression, Lipid Metabolism


15
Developmental Disorder, Reproductive System



Disease, Cellular Compromise


15
Cell Cycle, Cancer, Cell Death


14
Cancer, Cellular Function and Maintenance,



Molecular Transport


13
Immune Response, Cell Signaling,



Protein Degradation


12
Cellular Movement, Hematological System



Development and Function, Immune Response


12
Cancer, Reproductive System Disease, Skeletal



and Muscular Disorders


11
Cell Cycle, Cell Morphology, Cell Death









These data demonstrate that hsa-miR-10a directly or indirectly affects the expression of numerous cancer-, cellular proliferation-, cellular development-, cell signaling-, and cell cycle-related genes and thus primarily affects functional pathways related to cancer, cellular growth, development, and proliferation. Those cellular processes all have integral roles in the development and progression of various cancers. Manipulation of the expression levels of genes in the cellular pathways shown in Table 2 represents a potentially useful therapy for cancer and other diseases in which increased or reduced expression of hsa-miR-10a has a role in the disease.


Example 3
Predicted Gene Targets of Hsa-miR-10a

Gene targets for binding of and regulation by hsa-miR-10a were predicted using the proprietary algorithm miRNATarget™ (Asuragen), which is an implementation of the method proposed by Krek (Krek et al., 2005). Predicted target genes are shown in Table 3.









TABLE 3







Predicted target genes of hsa-miR-10a.










RefSeq




Transcript ID


Gene Symbol
(Pruitt et al., 2005)
Description





ABCG1
NM_004915
ATP-binding cassette sub-family G member 1


ABCG1
NM_016818
ATP-binding cassette sub-family G member 1


ABCG1
NM_207174
ATP-binding cassette sub-family G member 1


ABCG1
NM_207627
ATP-binding cassette sub-family G member 1


ABCG1
NM_207628
ATP-binding cassette sub-family G member 1


ABCG1
NM_207629
ATP-binding cassette sub-family G member 1


ABCG1
NM_207630
ATP-binding cassette sub-family G member 1


ACTG1
NM_001614
actin, gamma 1 propeptide


AFF4
NM_014423
ALL1 fused gene from 5q31


AKAP13
NM_006738
A-kinase anchor protein 13 isoform 1


AKAP13
NM_007200
A-kinase anchor protein 13 isoform 2


AKAP13
NM_144767
A-kinase anchor protein 13 isoform 3


ANK3
NM_001149
ankyrin 3 isoform 2


ANK3
NM_020987
ankyrin 3 isoform 1


ANKRD12
NM_015208
ankyrin repeat domain 12


APPBP2
NM_006380
amyloid beta precursor protein-binding protein


ARHGAP12
NM_018287
Rho GTPase activating protein 12


ARIH2
NM_006321
ariadne homolog 2


ARNT
NM_001668
aryl hydrocarbon receptor nuclear translocator


ARNT
NM_178426
aryl hydrocarbon receptor nuclear translocator


ARNT
NM_178427
aryl hydrocarbon receptor nuclear translocator


ARRDC3
NM_020801
arrestin domain containing 3


ARSJ
NM_024590
arylsulfatase J


ASXL1
NM_015338
additional sex combs like 1


BACH2
NM_021813
BTB and CNC homology 1, basic leucine zipper


BAZ1B
NM_023005
bromodomain adjacent to zinc finger domain, 1B


BAZ1B
NM_032408
bromodomain adjacent to zinc finger domain, 1B


BAZ2B
NM_013450
bromodomain adjacent to zinc finger domain, 2B


BCL2L2
NM_004050
BCL2-like 2 protein


BCL6
NM_001706
B-cell lymphoma 6 protein


BCL6
NM_138931
B-cell lymphoma 6 protein


BDNF
NM_001709
brain-derived neurotrophic factor isoform a


BDNF
NM_170731
brain-derived neurotrophic factor isoform b


BDNF
NM_170732
brain-derived neurotrophic factor isoform a


BDNF
NM_170733
brain-derived neurotrophic factor isoform a


BDNF
NM_170734
brain-derived neurotrophic factor isoform c


BDNF
NM_170735
brain-derived neurotrophic factor isoform a


BICD2
NM_001003800
bicaudal D homolog 2 isoform 1


BICD2
NM_015250
bicaudal D homolog 2 isoform 2


BRUNOL6
NM_052840
bruno-like 6, RNA binding protein


BTBD11
NM_001017523
BTB (POZ) domain containing 11 isoform 2


BTBD11
NM_001018072
BTB (POZ) domain containing 11 isoform 3


BTBD11
NM_152322
BTB (POZ) domain containing 11 isoform 1


BTBD14B
NM_052876
transcriptional repressor NAC1


BTRC
NM_003939
beta-transducin repeat containing protein


BTRC
NM_033637
beta-transducin repeat containing protein


C16orf5
NM_013399
cell death inducing protein


CECR6
NM_031890
cat eye syndrome chromosome region, candidate 6


CNNM4
NM_020184
cyclin M4


CNOT6
NM_015455
CCR4-NOT transcription complex, subunit 6


COPS7B
NM_022730
COP9 constitutive photomorphogenic homolog


CRK
NM_005206
v-crk sarcoma virus CT10 oncogene homolog


CRK
NM_016823
v-crk sarcoma virus CT10 oncogene homolog


CSNK1G1
NM_001011664
casein kinase 1, gamma 1 isoform L


CSNK1G1
NM_022048
casein kinase 1, gamma 1 isoform S


CTDSPL
NM_001008392
small CTD phosphatase 3 isoform 1


CTDSPL
NM_005808
small CTD phosphatase 3 isoform 2


CTNNBIP1
NM_001012329
catenin, beta interacting protein 1


CTNNBIP1
NM_020248
catenin, beta interacting protein 1


DLGAP2
NM_004745
discs large-associated protein 2


DOCK11
NM_144658
dedicator of cytokinesis 11


E2F3
NM_001949
E2F transcription factor 3


ELAVL3
NM_001420
ELAV-like protein 3 isoform 1


ELAVL3
NM_032281
ELAV-like protein 3 isoform 2


ELOVL6
NM_024090
ELOVL family member 6, elongation of long chain


EPHA4
NM_004438
ephrin receptor EphA4


EPHA8
NM_020526
EPH receptor A8 isoform 1 precursor


ESRRG
NM_001438
estrogen-related receptor gamma isoform 1


ESRRG
NM_206594
estrogen-related receptor gamma isoform 2


ESRRG
NM_206595
estrogen-related receptor gamma isoform 2


FHL3
NM_004468
four and a half LIM domains 3


FIGN
NM_018086
fidgetin


FLJ10159
NM_018013
hypothetical protein LOC55084


FLJ13576
NM_022484
hypothetical protein LOC64418


FLJ14768
NM_032836
hypothetical protein FLJ14768


FLJ34969
NM_152678
hypothetical protein LOC201627


FLJ36031
NM_175884
hypothetical protein LOC168455


FLJ36874
NM_152716
hypothetical protein LOC219988


FLT1
NM_002019
fms-related tyrosine kinase 1 (vascular


FNBP1L
NM_001024948
formin binding protein 1-like isoform 1


FXR2
NM_004860
fragile X mental retardation syndrome related


GATA6
NM_005257
GATA binding protein 6


GPR3
NM_005281
G protein-coupled receptor 3


GRIN3A
NM_133445
glutamate receptor, ionotropic,


GRM3
NM_000840
glutamate receptor, metabotropic 3 precursor


HAS3
NM_005329
hyaluronan synthase 3 isoform a


HIVEP2
NM_006734
human immunodeficiency virus type I enhancer


HNRPK
NM_031263
heterogeneous nuclear ribonucleoprotein K


HOXA1
NM_005522
homeobox A1 isoform a


HOXA1
NM_153620
homeobox A1 isoform b


HOXA3
NM_030661
homeobox A3 isoform a


HOXA3
NM_153631
homeobox A3 isoform a


HOXA3
NM_153632
homeobox A3 isoform b


HOXD10
NM_002148
homeobox D10


HS6ST2
NM_147175
heparan sulfate 6-O-sulfotransferase 2


ID4
NM_001546
inhibitor of DNA binding 4, dominant negative


IGSF4
NM_014333
immunoglobulin superfamily, member 4


JPH1
NM_020647
junctophilin 1


KCTD16
NM_020768
potassium channel tetramerisation domain


KIAA1434
NM_019593
hypothetical protein LOC56261


KL
NM_153683
klotho isoform b


KL
NM_004795
klotho isoform a


KLF11
NM_003597
Kruppel-like factor 11


KLF13
NM_015995
Kruppel-like factor 13


KLF4
NM_004235
Kruppel-like factor 4 (gut)


LOC440093
NM_001013699
hypothetical protein LOC440093


LPHN1
NM_001008701
latrophilin 1 isoform 1 precursor


LPHN1
NM_014921
latrophilin 1 isoform 2 precursor


LYPLA3
NM_012320
lysophospholipase 3 (lysosomal phospholipase


MAMDC1
NM_182830
MAM domain containing 1


MAPRE1
NM_012325
microtubule-associated protein, RP/EB family,


MLLT6
NM_005937
myeloid/lymphoid or mixed-lineage leukemia


MLR2
NM_032440
ligand-dependent corepressor


MTMR3
NM_021090
myotubularin-related protein 3 isoform c


MTMR3
NM_153050
myotubularin-related protein 3 isoform a


MTMR3
NM_153051
myotubularin-related protein 3 isoform b


NARG1
NM_057175
NMDA receptor regulated 1


NCOA6
NM_014071
nuclear receptor coactivator 6


NCOR2
NM_006312
nuclear receptor co-repressor 2


NFASC
NM_015090
neurofascin precursor


NFAT5
NM_006599
nuclear factor of activated T-cells 5 isoform c


NFAT5
NM_138713
nuclear factor of activated T-cells 5 isoform b


NFAT5
NM_138714
nuclear factor of activated T-cells 5 isoform a


NFAT5
NM_173214
nuclear factor of activated T-cells 5 isoform a


NFIX
NM_002501
nuclear factor I/X (CCAAT-binding transcription


NR4A3
NM_006981
nuclear receptor subfamily 4, group A, member 3


NR4A3
NM_173198
nuclear receptor subfamily 4, group A, member 3


NR4A3
NM_173200
nuclear receptor subfamily 4, group A, member 3


NR5A2
NM_003822
nuclear receptor subfamily 5, group A, member 2


NR5A2
NM_205860
nuclear receptor subfamily 5, group A, member 2


NRP2
NM_003872
neuropilin 2 isoform 2 precursor


NRP2
NM_201266
neuropilin 2 isoform 1 precursor


NRP2
NM_201279
neuropilin 2 isoform 3 precursor


P15RS
NM_018170
hypothetical protein FLJ10656


PHF20L1
NM_024878
PHD finger protein 20-like 1 isoform 3


PHF20L1
NM_198513
PHD finger protein 20-like 1 isoform 2


POMT2
NM_013382
putative protein O-mannosyltransferase


PPARA
NM_001001928
peroxisome proliferative activated receptor,


PPARA
NM_001001929
peroxisome proliferative activated receptor,


PPARA
NM_001001930
peroxisome proliferative activated receptor,


PPARA
NM_005036
peroxisome proliferative activated receptor,


PRRT3
NM_207351
hypothetical protein LOC285368


PUM2
NM_015317
pumilio homolog 2


PURB
NM_033224
purine-rich element binding protein B


PURG
NM_001015508
purine-rich element binding protein G isoform B


RAP2A
NM_021033
RAP2A, member of RAS oncogene family


RYBP
NM_012234
RING1 and YY1 binding protein


SCARB2
NM_005506
scavenger receptor class B, member 2


SCN3A
NM_006922
sodium channel, voltage-gated, type III, alpha


SDC1
NM_001006946
syndecan 1 precursor


SDC1
NM_002997
syndecan 1 precursor


SFRS1
NM_006924
splicing factor, arginine/serine-rich 1


SFRS10
NM_004593
splicing factor, arginine/serine-rich 10


SGCD
NM_000337
delta-sarcoglycan isoform 1


SHC1
NM_003029
SHC (Src homology 2 domain containing)


SIX4
NM_017420
sine oculis homeobox homolog 4


SLC25A1
NM_005984
solute carrier family 25 (mitochondrial carrier;


SMAP1
NM_021940
stromal membrane-associated protein


SNX4
NM_003794
sorting nexin 4


SORCS1
NM_052918
SORCS receptor 1 isoform a


SORCS1
NM_001013031
SORCS receptor 1 isoform b


SPTY2D1
NM_194285
hypothetical protein LOC144108


SVOP
NM_018711
SV2 related protein


TBX5
NM_000192
T-box 5 isoform 1


TBX5
NM_080717
T-box 5 isoform 3


TBX5
NM_181486
T-box 5 isoform 1


TFAP2C
NM_003222
transcription factor AP-2 gamma


TNRC6B
NM_001024843
trinucleotide repeat containing 6B isoform 2


TNRC6B
NM_015088
trinucleotide repeat containing 6B isoform 1


TRIM2
NM_015271
tripartite motif-containing 2


USP46
NM_022832
ubiquitin specific protease 46


WDR26
NM_025160
WD repeat domain 26


WNK3
NM_001002838
WNK lysine deficient protein kinase 3 isoform 2


WNK3
NM_020922
WNK lysine deficient protein kinase 3 isoform 1


XRN1
NM_019001
5′-3′ exoribonuclease 1


YOD1
NM_018566
hypothetical protein LOC55432


ZMYND11
NM_006624
zinc finger, MYND domain containing 11 isoform


ZNF367
NM_153695
zinc finger protein 367


ZNF608
NM_020747
zinc finger protein 608









The predicted gene targets that exhibited altered mRNA expression levels in HL-60 cells, following transfection with Pre-miR hsa-miR-10a, are shown in Table 4.









TABLE 4







Predicted hsa-miR-10a targets that exhibited altered


mRNA expression levels in HL-60


cells after transfection with Pre-miR hsa-miR-10a.










RefSeq



Gene
Transcript ID


Symbol
(Pruitt et al., 2005)
Description





P15RS
NM_018170
hypothetical protein FLJ10656


SLC25A1
NM_005984
solute carrier family




25 (mitochondrial carrier;









The predicted gene targets of hsa-miR-10a whose mRNA expression levels are affected by hsa-miR-10a represent particularly useful candidates for cancer therapy and therapy of other diseases through manipulation of their expression levels.


Example 4
Cancer Related Gene Expression Altered by Hsa-miR-10a

Cell proliferation and growth pathways are commonly altered in tumors. The inventors have shown that hsa-miR-10a directly or indirectly regulates the transcripts of proteins that are critical in the regulation of these pathways. Many of these targets have inherent oncogenic or tumor suppressor activity. Hsa-miR-10a targets that are associated with various cancer types are shown in Table 5.


Many genes that are affected by hsa-miR-10a have been associated with blood borne malignancies, such as various forms of leukemia (acute myeloid leukemia, acute lymphoblastic leukemia, chronic myeloid leukemia, chronic lymphoblastic leukemia, and T-cell leukemia), Ewings Sarcoma or acquired α-thalassemia (Table 5). This observation may be explained by the fact that HL-60 cells, a cell line derived from acute myeloid leukemia, were used for this analysis. However, transfection of hsa-miR-10a also affected target genes that are critical in the development of many solid tumors. Therefore, hsa-miR-10a or hsa-miR-10a inhibitors may have broad therapeutic utility in the treatment of human cancer.


Hsa-miR-10a controls genes and their products that function in the regulation of adhesion, migration, signal transduction, cell cycle progression, transcription, replication, chromosomal stability, apoptosis, as well as metabolisms of fatty acids, sugars and nucleotides. For instance, hsa-miR-10a targets that function in intracellular signaling pathways include STAT3, TNFSF13, IGFBP2 and PRKCD. STAT3 (signal transducer and activator of transcription 3) is a member of the STAT transcription factor protein family that consists of seven members (Hodge et al., 2005). Among the STAT proteins, STAT3 shows a strong association with many different types of cancer and is the one most frequently hyperactivated in many tumors (Hodge et al., 2005). STAT3 is directly regulated by the Janus kinase (JAK) in cytokine signaling and also lies downstream of many oncogenic pathways: STAT3 is activated in response to Src, VEGF (vascular endothelial growth factor), the ABL tyrosine kinase and IL6 (interleukin 6) (Yu et al., 2007). STAT3 itself is inherently oncogenic and—when constitutively active—induces cellular transformation of fibroblasts in culture. STAT3 is anti-apoptotic and promotes oncogenesis as well as immune evasion. Therefore, STAT3 is an attractive cancer drug target for small molecules and RNA interference (Hodge et al., 2005; Yu et al., 2007). TNFSF13 (tumor necrosis factor ligand superfamily member 13), also known as TALL or APRIL (a proliferation-inducing ligand) binds to TNF receptors and controls various intracellular pathways in immune cells and other tissues. TNFSF13 is abundantly expressed in a variety of human malignancies and promotes tumor-cell proliferation (Dillon et al., 2006). TNFSF13-antagonists, such as neutralizing antibodies, inhibit the growth of human cancer xenografts in vivo and are currently being tested in clinical trials (Dillon et al., 2006). IGFBP2 (insulin-like growth factor-binding protein 2) is a member of the IGFBP gene family that comprises six members, coding for membrane-bound or secretory proteins that modulate the activity of class I and II insulin-like growth factors (Hoeflich et al., 2001). Increased serum IGFBP2 levels were found in patients with carcinomas of the lung, colon, adrenal glands, ovary, prostate and the central nervous system, as well as in patients with lymphoid tumors, non-islet cell tumor hypoglycemia and Wilms' tumor (Hoeflich et al., 2001; Firth and Baxter, 2002). Since IGFBP2 levels are low in well-differentiated hepatoblastoma or normal livers and are high in poorly differentiated hepatoblastoma, IGFBP2 might serve as a marker and/or causative component for tumor differentiation. PRKCD (protein kinase C delta, PKC delta) belongs to a family of serine-threonine kinases that are activated in response to signaling induced by receptor tyrosine kinases. Functional studies have suggested that PKCs play a role in carcinogenesis and maintenance of the malignant phenotype (Koivunen et al., 2006). The human PRKCD gene is located on chromosome 3p in a region that is frequently subject to loss of heterozygosity in a wide range of tumors. Since PRKCD promotes cell survival and chemotherapeutic resistance in NSCLC cells, but also promotes apoptosis in other cell types, PRKCD may have both tumor suppressor and proliferation capabilities (Jackson and Foster, 2004; Koivunen et al., 2006). There is evidence that suggests that downregulation of PRKCD enhances breast cancer progression and may also contribute to the proliferation in renal cell carcinoma (Jackson and Foster, 2004).


Other cancer-associated targets of interest are MTA1, ATRX, FUS, EWSR1 and CEBPA, all of which have been implicated in regulating transcriptional processes. MTA1 (metastasis-associated gene 1) is involved in the transcriptional silencing machinery of mammalian cells (Hofer et al., 2004). Elevated levels of MTA1 are frequently detected in many cancer types and correlate with tumor invasiveness, metastasis, tumor progression and angiogenesis (Toh et al., 1994; Toh et al., 1997; Toh et al., 1999; Sasaki et al., 2002; Yi et al., 2003; Hofer et al., 2004; Jang et al., 2006). Inhibition of MTA1 protein expression results in growth inhibition of cancer cell lines, marking this protein as a therapeutic target (Nawa et al., 2000). ATRX is a chromatin-associated protein that may act as a transcriptional cofactor and influence the epigenetic control of gene expression (Steensma et al., 2005). Inactivating somatic mutations of the trans-acting chromatin-associated factor ATRX are causatively linked to acquired α-thalassemia, the best characterized of the acquired red blood cell disorders in patients with hematological malignancy. The EWSR1 gene (Ewing's Sarcoma breakpoint region 1) codes for the EWS protein, an RNA binding molecule that also contains a transactivation domain (Janknecht, 2005). EWSR1 is involved in several translocation events, giving rise to fusion proteins with oncogenic properties. In most cases, the EWSR1 gene recombines with sequences specific for transcription factors, such as ETS (mammalian homolog of the v-Ets oncoprotein originally isolated from the transforming erythroblastosis virus E26) or WT1 (Wilms' Tumor 1 gene) (Gerald and Haber, 2005). These oncoproteins act as aberrant transcription factors due to the fusion of an ETS DNA binding domain to a highly potent EWS transactivation domain. Next to Ewing's Sarcoma, various EWS fusion proteins have been found in myxoid liposarcoma, chondrosarcoma and acute leukemia. The FUS gene (also known as TLS, translocated in liposarcoma) is located on the same genomic locus as EWSR1 and therefore also subject to chromosomal translocations and fusion proteins (Janknecht, 2005). Similar to EWS fusion proteins, FUS chimeras have oncogenic properties. CEBPA, CCATT/enhancer-binding protein alpha, is a key transcription factor involved in late differentiation events of several cell types (Schuster and Porse, 2006). It is also a well-characterized inhibitor of mitotic growth in most cell lines. CEBPA acts as a tumor suppressor in the hematopoietic system: inactivating mutations are found in AML and myelodysplastic syndrome (MDS), and mutated CEBPA contributes to tumorigenesis (Nerlov, 2004; Zhang et al., 2004; Schuster and Porse, 2006). CEBPA is also expressed at reduced levels in lung and breast carcinomas, suggesting that CEBPA is a potential contributor to the malignant phenotype. Introduction of CEBPA into cells derived from lung cancer, breast cancer or leukemia leads to robust growth arrest, suggesting a tumor suppressor function for CEBPA (Schuster and Porse, 2006).


Hsa-miR-10a also regulates vimentin (VIM) and fatty acid synthase (FASN). Vimentin is the major intermediate filament protein of mesenchymal cells and governs proteins that are critical in attachment, migration and cell signaling. Vimentin has key roles in adhesion by regulating integrin functions. Vimentin is often expressed at elevated levels in human malignancies which correlates with invasiveness and poor survival (Caselitz et al., 1983; Churg, 1985; Upton et al., 1986; Sommers et al., 1992; Gilles et al., 1996; Islam et al., 2000; Singh et al., 2003; Ngan et al., 2007). Downregulation of vimentin expression by RNA interference inhibits carcinoma cell migration and adhesion (McInroy and Maatta, 2007). FASN, often referred to as FAS, is the sole protein in the human genome capable of the reductive de novo synthesis of long-chain fatty acids from acetyl-CoA, malonyl-CoA, and nicotinamide adenine dinucleotide phosphate (NADPH) (Kuhajda, 2006). FASN is elevated and active in various cancer cells. Since fatty acid synthesis expends energy, FASN expression might confer some survival or growth advantage to human cancer cells. FASN expression correlates with poor prognosis of patients with carcinomas of the lung, breast, prostate, skin and soft-tissue sarcomas and is predictive for recurrence of prostate cancer (Kuhajda, 2006).


In summary, hsa-miR-10a governs the activity of proteins that are critical regulators of cell proliferation and tumor development. Hsa-miR-10a controls genes with inherent oncogenic properties, including FASN, VIM, MTA1, EWSR1, FUS, STAT3 or IGFBP2, and genes with tumor suppressor activity, including TNFSF13 and ATRX. Based on the review of these genes, their related pathways and how they are regulated by hsa-miR-10a, introduction of hsa-miR-10a or hsa-miR-10a inhibitors into a variety of cancer cell types would likely result in a therapeutic response.









TABLE 5







Disease associated mRNAs altered by hsa-miR-10a having prognostic or therapeutic value for the treatment of various


diseases or malignancies.











Gene

Cellular




Symbol
Gene Title
Process
Disease
Reference





SIVA
CD27 binding
apoptosis
BC
(Chu et al., 2005)


ATRX
ATR-X
transcription
AML, alpha thalassemia
(Lacayo et al., 2004; Steensma et al., 2005; Serrano et






al., 2006)


BRCA2
BRCA-2
chromosomal
BC, OC
(Wooster and Weber, 2003; Gottardo et al., 2007)




stability


BTG3
B-cell
cell cycle
ALL
(Gottardo et al., 2007)



translocation



gene 3


BZRP
Benzodiazepine
apoptosis
L, BC, G, CRC, AC, PC,
(Hardwick et al., 1999; Sutter et al., 2002; Han et al.,



receptor,

FS, OepC
2003; Kletsas et al., 2004; Furre et al., 2005; Maaser



peripheral


et al., 2005; Pretner et al., 2006; Vlodavsky and



type


Soustiel, 2007)


CCL23
Chemokine
signal
AML
(Steinbach et al., 2006; Bruserud et al., 2007)



ligand 23
transduction


CCL5
RANTES
signal
TCL, PC, MCL, BC,
(Luboshits et al., 1999; Niwa et al., 2001; Moran et




transduction
NSCLC, BC, CeC, AML
al., 2002; Mori et al., 2004; Ek et al., 2006; Olsnes et






al., 2006; Vaday et al., 2006; Yaal-Hahoshen et al.,






2006)


CCL7
MCP3
signal
CeC, CRC
(Wetzel et al., 2001; Hu et al., 2002)




transduction


CD151
SFA-1
signal
NSCLC, CRC, GB, FS,
(Tokuhara et al., 2001; Kohno et al., 2002; Ang et al.,




transduction
PC
2004)


CD99
MIC2
signal
EWS, OC, PaC, LC, OS,
(Sohn et al., 1998; Choi et al., 2000; Scotlandi et al.,




transduction,
CeC, BC, M, GC
2000; Goto et al., 2004; Yoo et al., 2005; Byun et al.,




cell adhesion

2006; Manara et al., 2006; Wilkerson et al., 2006;






Zhou et al., 2006; Lee et al., 2007)


CDC37
cell division
cell cycle
PC, AML, MM, HCC,
(Stepanova et al., 2000; Casas et al., 2003; Katayama



cycle 37


et al., 2004; Pascale et al., 2005; Pearl, 2005)


CDT1
double
chromosomal
NSCLC
(Pabst et al., 2001; Karakaidos et al., 2004; Schuster



parked, DUP
stability

and Porse, 2006)


CEBPA
CCAAT/enhancer-
transcription
AML, LC, HCC, BC
(Pabst et al., 2001; Schuster and Porse, 2006)



binding



protein alpha


COMT
catechol-O-
metabolism,
BC, schizophrenia,
(Amin and Ismail, 1983; Huang et al., 1999; Rivest et



methyltransferase
neural
Alzheimer disease,
al., 1999; Egan et al., 2001; Fan et al., 2005; Borroni




function
Parkinson disease
et al., 2007)


ELK1
Elk-1
transcription
BC, OepC
(Shao et al., 1998; Chai et al., 2001; Chen et al., 2006)


EWSR1
EWS
RNA-
EWS, LS, CS, ALL,
(Martini et al., 2002; Gerald and Haber, 2005;




binding,
AML, MT
Janknecht, 2005)




transcription


FASN
Fatty acid
fat
OC, BC, BldC, CeC, PC,
(Ye et al., 2000; Camassei et al., 2003; Menendez et



synthase
metabolism
RB, CRC
al., 2004; Kuhajda, 2006)


FH
fumarase
sugar
RCC, LM
(Eng et al., 2003)




metabolism


FUS
TLS
RNA-
LS, EWS, AML
(Martini et al., 2002; Janknecht, 2005; Perez-Mancera




binding,

and Sanchez-Garcia, 2005)




transcription


GADD45B
MYD118
apoptosis
L, HCC, NHL, OepC,
(Selvakumaran et al., 1994; Qiu et al., 2003; Ying et





NPC, LC
al., 2005)


IGFBP2
IGFBP-2
signal
G, OC, CRC, LC, PC,
(Hoeflich et al., 2001; Firth and Baxter, 2002; Dunlap




transduction
WT, ALL, HB
et al., 2007)


JUNB
Jun B
transcription
L, CML, HCC, TCL, HL,
(Bossy-Wetzel et al., 1992; Mathas et al., 2002; Mao





FS
et al., 2003; Yang et al., 2003; Passegue et al., 2004;






Chang et al., 2005; Liu et al., 2006; Ott et al., 2007)


MATK
CTK, CHK
signal
BC, PaC, AC, GB, NB
(Zrihan-Licht et al., 1998; Kim et al., 2004; Fu et al.,




transduction

2006)


MTA1
metastasis-
transcription
BC, GC, OepC, PaC, LC,
(Toh et al., 1994; Toh et al., 1997; Toh et al., 1999;



associated

HCC, OC, PC, EC
Iguchi et al., 2000; Sasaki et al., 2002; Hamatsu et al.,



gene 1


2003; Yi et al., 2003; Hofer et al., 2004; Balasenthil et






al., 2006; Jang et al., 2006)


P8
P8
transcription
BC, TC, PaC
(Ree et al., 1999; Su et al., 2001; Ito et al., 2005)


PRKCD
protein kinase
apoptosis
BldC, HCC, RCC, BC,
15907369, 15054085(Jackson and Foster, 2004;



C delta

NSCLC, MM
Koivunen et al., 2006)


STAT3
Stat-3
signal
MM, CLL, AML, LC,
(Hodge et al., 2005; Yu et al., 2007)




transduction
BC, CRC, RCC, PC,





PaC, M, GC, CeC, OC,





HCC, SCCHN


TFDP1
E2F
cell cycle
M, HCC, NHL
(Halaban et al., 2000; Wang et al., 2001; Chan et al.,



dimerization


2002; Yasui et al., 2002)



partner


TINF2
TIN2
replication
TCL, HCC, GC
(Yamada et al., 2002; Oh et al., 2005; Bellon et al.,






2006)


TNFSF13
APRIL,
signal
HCC, GB, MM, LC,
(Dillon et al., 2006)



TALL2
transduction
CRC


TYMS
thymidylate
nucleotide
GBM, GC, L, TC, CRC
(Kass and Munster, 1980; Sakamoto et al., 1991;



synthetase
synthesis

Libra et al., 2004; Toriumi et al., 2004; Grunda et al.,






2006)


VAV1
Vav-1
signal
PaC, NB, CML, AML
(Luger et al., 1996; Turhan et al., 1998; Hornstein et




transduction

al., 2003; Bertagnolo et al., 2005; Fernandez-Zapico






et al., 2005; Opalinska et al., 2005; Prieto-Sanchez et






al., 2006)


VIM
vimentin
adhesion and
HCC, M, L, BC, PC,
(Caselitz et al., 1983; Stark et al., 1984; Ben-Ze'ev




migration
CeC, CRC, RCC,
and Raz, 1985; Churg, 1985; Upton et al., 1986;





SCCHN, AC, CLL, MT,
Ferrari et al., 1990; Sommers et al., 1992; Gilles et al.,





LC
1996; Rutka et al., 1999; Islam et al., 2000; Khoury et






al., 2002; Singh et al., 2003; Hu et al., 2004; McInroy






and Maatta, 2007; Ngan et al., 2007)





Abbreviations:


AC, astrocytoma;


ALL, acute lymphoblastic leukemia;


AML, acute myeloid leukemia;


BC, breast carcinoma;


BldC, bladder carcinoma;


CeC, cervical carcinoma;


CLL, chronic lymphoblastic leukemia;


CML, chronic myeloid leukemia;


CRC, colorectal carcinoma;


CS, chondrosarcoma;


EC, endometrial carcinoma;


EWS, Ewing's sarcoma;


FS, fibrosarcoma;


G, glioma;


GB, glioblastoma;


GBM, glioblastoma multiforme;


GC, gastric carcinoma;


HB, hepatoblastoma;


HCC, hepatocellular carcinoma;


HL, Hodgkin lymphoma;


L, leukemia;


LC, lung carcinoma;


LM, leiomyoma;


LS, liposarcoma;


M, melanoma;


MCL, mantle cell lymphoma;


MM, multiple myeloma;


MT, mesothelioma;


NB, neuroblastoma;


NHL, non-Hodgkin lymphoma;


NPC, nasopharyngeal carcinoma;


NSCLC, non-small cell lung carcinoma;


OC, ovarian carcinoma;


OepC, oesophageal carcinoma;


OS, osteosarcoma;


PaC, pancreatic carcinoma;


PC, prostate carcinoma;


RB, retinoblastoma;


RCC, renal cell carcinoma;


SCCHN, squamous cell carcinoma of the head and neck;


TC, thyroid carcinoma;


TCL, T-cell leukemia;


WT, Wilm's tumor






REFERENCES

The following references, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated herein by reference.

  • U.S. Pat. No. 4,337,063
  • U.S. Pat. No. 4,404,289
  • U.S. Pat. No. 4,405,711
  • U.S. Pat. No. 4,659,774
  • U.S. Pat. No. 4,659,774
  • U.S. Pat. No. 4,682,195
  • U.S. Pat. No. 4,683,202
  • U.S. Pat. No. 4,704,362
  • U.S. Pat. No. 4,816,571
  • U.S. Pat. No. 4,816,571
  • U.S. Pat. No. 4,870,287
  • U.S. Pat. No. 4,959,463
  • U.S. Pat. No. 4,959,463
  • U.S. Pat. No. 5,141,813
  • U.S. Pat. No. 5,141,813
  • U.S. Pat. No. 5,143,854
  • U.S. Pat. No. 5,143,854
  • U.S. Pat. No. 5,202,231
  • U.S. Pat. No. 5,214,136
  • U.S. Pat. No. 5,221,619
  • U.S. Pat. No. 5,223,618
  • U.S. Pat. No. 5,242,974
  • U.S. Pat. No. 5,264,566
  • U.S. Pat. No. 5,264,566
  • U.S. Pat. No. 5,268,486
  • U.S. Pat. No. 5,288,644
  • U.S. Pat. No. 5,324,633
  • U.S. Pat. No. 5,378,825
  • U.S. Pat. No. 5,384,261
  • U.S. Pat. No. 5,384,261
  • U.S. Pat. No. 5,399,363
  • U.S. Pat. No. 5,405,783
  • U.S. Pat. No. 5,412,087
  • U.S. Pat. No. 5,424,186
  • U.S. Pat. No. 5,424,186
  • U.S. Pat. No. 5,428,148
  • U.S. Pat. No. 5,428,148
  • U.S. Pat. No. 5,429,807
  • U.S. Pat. No. 5,432,049
  • U.S. Pat. No. 5,436,327
  • U.S. Pat. No. 5,445,934
  • U.S. Pat. No. 5,445,934
  • U.S. Pat. No. 5,446,137
  • U.S. Pat. No. 5,466,468
  • U.S. Pat. No. 5,466,786
  • U.S. Pat. No. 5,468,613
  • U.S. Pat. No. 5,470,710
  • U.S. Pat. No. 5,470,967
  • U.S. Pat. No. 5,472,672
  • U.S. Pat. No. 5,480,980
  • U.S. Pat. No. 5,492,806
  • U.S. Pat. No. 5,503,980
  • U.S. Pat. No. 5,510,270
  • U.S. Pat. No. 5,525,464
  • U.S. Pat. No. 5,525,464
  • U.S. Pat. No. 5,527,681
  • U.S. Pat. No. 5,529,756
  • U.S. Pat. No. 5,532,128
  • U.S. Pat. No. 5,543,158
  • U.S. Pat. No. 5,545,531
  • U.S. Pat. No. 5,547,839
  • U.S. Pat. No. 5,554,501
  • U.S. Pat. No. 5,554,744
  • U.S. Pat. No. 5,554,744
  • U.S. Pat. No. 5,556,752
  • U.S. Pat. No. 5,561,071
  • U.S. Pat. No. 5,571,639
  • U.S. Pat. No. 5,574,146
  • U.S. Pat. No. 5,574,146
  • U.S. Pat. No. 5,580,726
  • U.S. Pat. No. 5,580,732
  • U.S. Pat. No. 5,593,839
  • U.S. Pat. No. 5,599,672
  • U.S. Pat. No. 5,599,695
  • U.S. Pat. No. 5,602,240
  • U.S. Pat. No. 5,602,244
  • U.S. Pat. No. 5,602,244
  • U.S. Pat. No. 5,610,289
  • U.S. Pat. No. 5,610,287
  • U.S. Pat. No. 5,614,617
  • U.S. Pat. No. 5,623,070
  • U.S. Pat. No. 5,624,711
  • U.S. Pat. No. 5,631,134
  • U.S. Pat. No. 5,637,683
  • U.S. Pat. No. 5,639,603
  • U.S. Pat. No. 5,641,515
  • U.S. Pat. No. 5,645,897
  • U.S. Pat. No. 5,652,099
  • U.S. Pat. No. 5,654,413
  • U.S. Pat. No. 5,658,734
  • U.S. Pat. No. 5,661,028
  • U.S. Pat. No. 5,665,547
  • U.S. Pat. No. 5,667,972
  • U.S. Pat. No. 5,670,663
  • U.S. Pat. No. 5,672,697
  • U.S. Pat. No. 5,677,195
  • U.S. Pat. No. 5,681,947
  • U.S. Pat. No. 5,695,940
  • U.S. Pat. No. 5,700,637
  • U.S. Pat. No. 5,700,922
  • U.S. Pat. No. 5,705,629
  • U.S. Pat. No. 5,708,153
  • U.S. Pat. No. 5,708,154
  • U.S. Pat. No. 5,714,606
  • U.S. Pat. No. 5,728,525
  • U.S. Pat. No. 5,728,525
  • U.S. Pat. No. 5,739,169
  • U.S. Pat. No. 5,744,305
  • U.S. Pat. No. 5,744,305
  • U.S. Pat. No. 5,760,395
  • U.S. Pat. No. 5,763,167
  • U.S. Pat. No. 5,770,358
  • U.S. Pat. No. 5,777,092
  • U.S. Pat. No. 5,789,162
  • U.S. Pat. No. 5,792,847
  • U.S. Pat. No. 5,800,992
  • U.S. Pat. No. 5,800,992
  • U.S. Pat. No. 5,801,005
  • U.S. Pat. No. 5,807,522
  • U.S. Pat. No. 5,824,311
  • U.S. Pat. No. 5,830,645
  • U.S. Pat. No. 5,830,880
  • U.S. Pat. No. 5,837,196
  • U.S. Pat. No. 5,846,225
  • U.S. Pat. No. 5,846,945
  • U.S. Pat. No. 5,847,219
  • U.S. Pat. No. 5,856,174
  • U.S. Pat. No. 5,858,988
  • U.S. Pat. No. 5,859,221
  • U.S. Pat. No. 5,871,928
  • U.S. Pat. No. 5,872,232
  • U.S. Pat. No. 5,876,932
  • U.S. Pat. No. 5,886,165
  • U.S. Pat. No. 5,919,626
  • U.S. Pat. No. 5,922,591
  • U.S. Pat. No. 6,004,755
  • U.S. Pat. No. 6,040,193
  • U.S. Pat. No. 6,040,193
  • U.S. Pat. No. 6,087,102
  • U.S. Pat. No. 6,251,666
  • U.S. Pat. No. 6,368,799
  • U.S. Pat. No. 6,383,749
  • U.S. Pat. No. 6,617,112
  • U.S. Pat. No. 6,638,717
  • U.S. Pat. No. 6,720,138
  • U.S. Pat. No. 6,723,509
  • U.S. patent Ser. No. 09/545,207
  • U.S. patent Ser. No. 11/141,707
  • U.S. patent Ser. No. 11/273,640
  • U.S. patent Ser. No. 11/349,727
  • U.S. patent Ser. No. 10/667,126
  • U.S. patent Ser. No. 10/667,126
  • U.S. Patent Ser. 60/575,743
  • U.S. Patent Ser. 60/649,584
  • U.S. Patent Ser. 60/650,807
  • Adelaide et al., Genes Chromosomes Cancer, 37(4):333-345, 2003.
  • Akiba et al., Int J Oncol, 18(2):257-264, 2001.
  • Akino et al., Gastroenterology, 129(1):156-169, 2005.
  • Alipov et al., Histopathology, 46(2):202-208, 2005.
  • Amin and Ismail, Gegenbaurs Morphol Jahrb, 129(1):125-128, 1983.
  • Ando et al., J Biol Chem, 279(24):25549-25561, 2004.
  • Ang et al., Cancer Epidemiol Biomarkers Prev, 13(11 Pt 1):1717-1721, 2004.
  • Aoki et al., Proc Natl Acad Sci USA, 101(37):13613-13617, 2004.
  • Arap et al., Cancer Res., 55(6):1351-1354, 1995.
  • Ariyanayagam-Baksh et al., Mod Pathol, 16(10):992-995, 2003.
  • Aspland et al., Oncogene, 20(40):5708-5717, 2001.
  • Austin and Cook, J Biol Chem, 280(39):33280-33288, 2005.
  • Austin-Ward and Villaseca, Revista Medica de Chile, 126(7):838-845, 1998.
  • Bader and Vogt, Oncogene, 23(18):3145-3150, 2004.
  • Bader et al., Nat Rev Cancer, 5(12):921-929, 2005.
  • Bae et al., J Biol Chem, 275(33):25255-25261, 2000.
  • Bagga et al., Cell, 122(4):553-563, 2005.
  • Bai et al., Mol Pain, 3:15, 2007.
  • Balasenthil et al., Hum Pathol, 37(6):656-661, 2006.
  • Bandyopadhyay et al., Oncogene, 21(22):3541-3551, 2002.
  • Barr, Oncogene, 20(40):5736-5746, 2001.
  • Bar-Shira et al., Cancer Res, 62(23):6803-6807, 2002.
  • Bartel et al., Cancer Cell, 2(1):9-15, 2002.
  • Beeram et al., J Clin Oncol, 23(27):6771-6790, 2005.
  • Behrens et al., J Pathol, 194(1):43-50, 2001.
  • Beisner et al., Cancer Res, 66(15):7554-7561, 2006.
  • Bell and Dutta, Annu Rev Biochem, 71:333-374, 2002.
  • Bellon et al., Int J Cancer, 119(9):2090-2097, 2006.
  • Ben-Ze'ev and Raz, Cancer Res, 45(6):2632-2641, 1985.
  • Bertagnolo et al., Exp Cell Res, 306(1):56-63, 2005.
  • Biswas et al., Cancer Res, 64(14):4687-4692, 2004.
  • Blobe et al., J Biol Chem, 276(27):24627-24637, 2001.
  • Borczuk et al., Am J Pathol, 163(5):1949-1960, 2003.
  • Borroni et al., Neurobiol Aging, 28(8):1231-1238, 2007.
  • Bossy-Wetzel et al., Genes Dev, 6(12A):2340-2351, 1992.
  • Boultwood et al., Br J Haematol, 126(4):508-511, 2004.
  • Bravou et al., Int J Oncol, 27(6):1511-1518, 2005.
  • Bruserud et al., Haematologica, 92(3):332-341, 2007.
  • Buggy et al., Br J Cancer, 91(7):1308-1315, 2004.
  • Bukowski et al., Clinical Cancer Res., 4(10):2337-2347, 1998.
  • Byun et al., J Biol Chem, 281(46):34833-34847, 2006.
  • Cahill et al., Nature, 392(6673):300-303, 1998.
  • Caldas et al., Cancer Res., 54:3568-3573, 1994.
  • Calin and Croce, Nat Rev Cancer, 6(11):857-866, 2006.
  • Camassei et al., Invest Opthalmol Vis Sci, 44(6):2399-2403, 2003.
  • Cao et al., Cell, 107(6):763-775, 2001.
  • Cao et al., Genes Dev, 21(5):531-536, 2007.
  • Carrano et al., Nat Cell Biol, 1(4):193-199, 1999.
  • Carrington and Ambros, Science, 301(5631):336-338, 2003.
  • Carter and Brunet, Curr Biol, 17(4):R113-114, 2007.
  • Casas et al., Cancer Genet Cytogenet, 146(2):89-101, 2003.
  • Caselitz et al., Virchows Arch A Pathol Anat Histopathol, 400(1):43-51, 1983.
  • Chai et al., Oncogene, 20(11):1357-1367, 2001.
  • Chan et al., Appl Immunohistochem Mol Morphol, 10(4):322-326, 2002.
  • Chandler et al., Int J Cancer, 81(3):451-458, 1999.
  • Chang et al., Int J Cancer, 114(6):942-949, 2005.
  • Chang et al., Oncol Rep, 13(3):433-438, 2005.
  • Chen et al., World J Gastroenterol, 12(48):7859-7863, 2006.
  • Chendrimada et al., Nature, 447(7146):823-828, 2007.
  • Cheng et al., Cancer Res., 54(21):5547-5551, 1994.
  • Chieffi et al., Prostate, 66(3):326-333, 2006.
  • Choi et al., Arch Pathol Lab Med, 124(4):563-569, 2000.
  • Choi et al., Oncogene, 23(42):7095-7103, 2004.
  • Cho-Vega et al., Hum Pathol, 35(9):1095-1100, 2004.
  • Christodoulides et al., Microbiology, 144(Pt 11):3027-3037, 1998.
  • Chu et al., Cancer Res, 65(12):5301-5309, 2005.
  • Churg, Am J Surg Pathol, 9(5):360-365, 1985.
  • Cipriano and Chen, Oncogene, 17(12):1549-1556, 1998.
  • Claudio et al., Clin Cancer Res, 8(6):1808-1815, 2002.
  • Cohen et al., Oncogene, 20(2):141-146, 2001.
  • Coll et al., Embo J, 2(12):2189-2194, 1983.
  • Costello et al., Cancer Res, 57(7):1250-1254, 1997.
  • Crossen and Morrison, Hum Genet, 91(4):380-382, 1993.
  • Crossen et al., Cancer Genet Cytogenet, 79(1):70-73, 1995.
  • Cully et al., Cancer Res, 65(22):10363-10370, 2005.
  • Cummins et al., In: IRT: Nucleosides and nucleosides, La Jolla Calif., 72, 1996.
  • Davidson et al., Clin Cancer Res, 7(3):551-557, 2001.
  • Davidson et al., J. Immunother., 21(5):389-398, 1998.
  • Denli et al., Trends Biochem. Sci., 28:196, 2003.
  • Didenko, Biotechniques, 31(5):1106-1116, 1118, 1120-1121, 2001.
  • Dillman, Cancer Biother. Radiopharm., 14(1):5-10, 1999.
  • Dillon et al., Nat Rev Drug Discov, 5(3):235-246, 2006.
  • Dittmer, Mol Cancer, 2:29, 2003.
  • Dong et al., Mol Endocrinol, 20(10):2315-2325, 2006.
  • Dreyfus et al., Nouv Rev Fr Hematol, 31(3):217-221, 1989.
  • Dunlap et al., Proc Natl Acad Sci USA, 104(28):11736-11741, 2007.
  • Dyer and Bremner, Nat Rev Cancer, 5(2):91-101, 2005.
  • Egan et al., Proc Natl Acad Sci USA, 98(12):6917-6922, 2001.
  • Egilmez et al., J Exp Clin Cancer Res, 20(4):549-552, 2001.
  • Egle et al., Proc Natl Acad Sci USA, 101(16):6164-6169, 2004.
  • Egloff et al., Cancer Res, 66(1):6-9, 2006.
  • Einama et al., Pancreas, 32(4):376-381, 2006.
  • Ek et al., Int J Cancer, 118(8):2092-2097, 2006.
  • Emptage et al., Neuron, 29(1):197-208, 2001.
  • Endoh et al., Br J Cancer, 93(12):1395-1399, 2005.
  • Eng et al., Nat Rev Cancer, 3(3):193-202, 2003.
  • EP 266,032
  • EP 373 203
  • EP 785 280
  • EP 799 897
  • Esquela-Kerscher and Slack, Nat Rev Cancer, 6(4):259-269, 2006.
  • Fakharzadeh et al., Embo J, 10(6):1565-1569, 1991.
  • Fan et al., Biol Psychiatry, 57(2):139-144, 2005.
  • Fernandez-Zapico et al., Cancer Cell, 7(1):39-49, 2005.
  • Ferrari et al., Cancer Res, 50(7):1988-1991, 1990.
  • Firth and Baxter, Endocr Rev, 23(6):824-854, 2002.
  • Fisher, J Royal Statistical Soc, 85(1):87-94, 1922.
  • Fleischer et al., Int J Oncol, 28(1):25-32, 2006.
  • Fodor et al., Biochemistry, 30(33):8102-8108, 1991.
  • Froehler et al., Nucleic Acids Res., 14:5399-5407, 1986.
  • Fu et al., Int J Oncol, 29(6):1453-1458, 2006.
  • Fujimoto et al., Ann Oncol, 13(10): 1598-1604, 2002a.
  • Fujimoto et al., Ann Oncol, 13(10):1605-1611, 2002b.
  • Fujiwara et al., Oncogene, 10(5):891-895, 1995.
  • Furre et al., Cancer Res, 65(23):11051-11060, 2005.
  • Garzon et al., Proc Natl Acad Sci USA, 103(13):5078-5083, 2006.
  • Gerald and Haber, Semin Cancer Biol, 15(3):197-205, 2005.
  • Gilles et al., J Pathol, 180(2):175-180, 1996.
  • Ginestier et al., Clin Cancer Res, 12(15):4533-4544, 2006.
  • Golay et al., Blood, 87(5):1900-1911, 1996.
  • Gomez-Bougie et al., Eur J Immunol, 34(11):3156-3164, 2004.
  • Gonzalez et al., Nature, 440(7084):702-706, 2006.
  • Goto et al., Histopathology, 45(4):384-392, 2004.
  • Goto et al., Oncogene, 2007.
  • Gottardo et al., Br J Haematol, 137(4):319-328, 2007.
  • Goyns et al., Br J Cancer, 56(5):611-613, 1987.
  • Grabsch et al., J Pathol, 200(1):16-22, 2003.
  • Gratas et al., Cancer Res, 58(10):2057-2062, 1998.
  • Griffey et al., J. Mass Spectrom, 32(3):305-13, 1997.
  • Grunda et al., J Neurooncol, 80(3):261-274, 2006.
  • Gstaiger et al., Proc Natl Acad Sci USA, 98(9):5043-5048, 2001.
  • Halaban et al., J Exp Med, 191(6):1005-1016, 2000.
  • Hamatsu et al., Oncol Rep, 10(3):599-604, 2003.
  • Han et al., J Recept Signal Transduct Res, 23(2-3):225-238, 2003.
  • Hanahan and Weinberg, Cell, 100(1):57-70, 2000.
  • Hanibuchi et al., Int. J. Cancer, 78(4):480-485, 1998.
  • Hansel et al., Am J Surg Pathol, 29(3):390-399, 2005.
  • Hardwick et al., Cancer Res, 59(4):831-842, 1999.
  • Hayette et al., Blood, 102(4):1549-1550, 2003.
  • Hellstrand et al., Acta Oncologica, 37(4):347-353, 1998.
  • Hishikawa et al., J Biol Chem, 274(52):37461-37466, 1999.
  • Hodge et al., Eur J Cancer, 41(16):2502-2512, 2005.
  • Hoeflich et al., Cancer Res, 61(24):8601-8610, 2001.
  • Hofer et al., Cancer Res, 64(3):825-829, 2004.
  • Hornstein et al., J Pathol, 199(4):526-533, 2003.
  • Houston and O'Connell, Curr Opin Pharmacol, 4(4):321-326, 2004.
  • Hsu et al., Mol Endocrinol, 12(9):1432-1440, 1998.
  • Hu et al., Oncogene, 23(1):298-302, 2004.
  • Hu et al., World J Gastroenterol, 8(6):1067-1072, 2002.
  • Huang et al., Cancer Res, 59(19):4870-4875, 1999.
  • Huang et al., Clin Cancer Res, 12(2):487-498, 2006.
  • Hui and Hashimoto, Infection Immun., 66(11):5329-5336, 1998.
  • Hussussian et al., Nat. Genet., 8(1):15-21, 1994.
  • Iguchi et al., Int J Oncol, 16(6):1211-1214, 2000.
  • Inui et al., Biochem Biophys Res Commun, 303(3):978-984, 2003.
  • Iolascon et al., Hepatology, 27(4):989-995, 1998.
  • Iorio et al., Cancer Res, 65(16):7065-7070, 2005.
  • Islam et al., J Cell Biochem, 78(1):141-150, 2000.
  • Itakura and Riggs, Science, 209:1401-1405, 1980.
  • Ito et al., Am J Clin Pathol, 114(5):719-725, 2000.
  • Ito et al., Anticancer Res, 23(5A):3819-3824, 2003.
  • Ito et al., Anticancer Res, 25(5):3419-3423, 2005.
  • Ito et al., Mod Pathol, 11(2):209-215, 1998.
  • Jackson and Foster, Faseb J, 18(6):627-636, 2004.
  • Jang et al., Cancer Sci, 97(5):374-379, 2006.
  • Janknecht, Gene, 363:1-14, 2005.
  • Jansen et al., Embo J, 2(11):1969-1975, 1983.
  • Jiang et al., Cancer Res, 64(16):5787-5794, 2004.
  • Jonson et al., Int J Oncol, 19(1):71-81, 2001.
  • Jung et al., Pathol Int, 56(9):503-509, 2006.
  • Kalin et al., Cancer Res, 66(3):1712-1720, 2006.
  • Kalinichenko et al., Genes Dev, 18(7):830-850, 2004.
  • Kamata et al., J Cancer Res Clin Oncol, 131(9):591-596, 2005.
  • Kamb et al., Nat. Genet., 8(1):23-26, 1994.
  • Kamb et al., Science, 2674:436-440, 1994.
  • Kapsimali et al., Genome Biol, 8(8):R173 [Epub ahead of print] 2007.
  • Karakaidos et al., Am J Pathol, 165(4):1351-1365, 2004.
  • Karakaidos et al., Am J Pathol, 165(4):1351-1365, 2004.
  • Karin et al., Nat Rev Cancer, 2(4):301-310, 2002.
  • Kass and Munster, Haematologica, 65(2):196-203, 1980.
  • Kastan and Lim, Nat Rev Mol Cell Biol, 1(3):179-186, 2000.
  • Katayama et al., Int J Oncol, 25(3):579-595, 2004.
  • Kaufmann et al., Blood, 91(3):991-1000, 1998.
  • Keen and Taylor, Nat Rev Cancer, 4(12):927-936, 2004.
  • Kerckaert et al., Leukemia, 4(1):16-19, 1990.
  • Khoury et al., Ann Diagn Pathol, 6(3):154-158, 2002.
  • Kim et al., Cancer Res, 66(4):2153-2161, 2006.
  • Kim et al., Cancer, 101(5):1018-1027, 2004.
  • Kim et al., Clin Cancer Res, 11(2 Pt 1):473-482, 2005.
  • Kiriakidou et al., Cell, 129(6):1141-1151, 2007.
  • Kirikoshi et al., Int J Oncol, 19(1):111-115, 2001.
  • Kitada et al., Blood, 91(9):3379-3389, 1998.
  • Kitange et al., Cancer, 89(11):2292-2300, 2000.
  • Kitange et al., Mod Pathol, 12(6):618-626, 1999.
  • Kletsas et al., Biochem Pharmacol, 67(10):1927-1932, 2004.
  • Klostermeier and Millar, Biopolymers, 61(3):159-79, 2001-2002.
  • Kohno et al., Int J Cancer, 97(3):336-343, 2002.
  • Koivunen et al., Cancer Lett, 235(1):1-10, 2006.
  • Koivunen et al., Cancer Res, 64(16):5693-5701, 2004.
  • Komiya et al., Jpn J Cancer Res, 88(4):389-393, 1997.
  • Kops et al., Nat Rev Cancer, 5(10):773-785. 2005.
  • Krajewska et al., Am J Pathol, 148(5):1567-1576, 1996.
  • Krasagakis et al., Br J Cancer, 77(9): 1492-1494, 1998.
  • Krek et al., Nat Genet, 37(5):495-500, 2005.
  • Kristjansdottir and Rudolph, Chem Biol, 11(8):1043-1051, 2004.
  • Kuhajda, Cancer Res, 66(12):5977-5980, 2006.
  • Kulkarni et al., Leukemia, 16(1):127-134, 2002.
  • Lacayo et al., Blood, 104(9):2646-2654, 2004.
  • Lagos-Quintana et al., Rna, 9(2):175-179, 2003.
  • Lahn and Sundell, Melanoma Res, 14(2):85-89, 2004.
  • Lam et al., Br J Neurosurg, 14(1):28-32, 2000.
  • Lambros et al., J Pathol, 205(1):29-40, 2005.
  • Lau et al., Science, 294(5543):858-862, 2001.
  • Lauffart et al., BMC Womens Health, 5:8, 2005.
  • Lee and Ambros, Science, 294(5543):862-864, 2001.
  • Lee et al., Clin Cancer Res, 13(9):2584-2591, 2007.
  • Leprince et al., Nature, 306(5941):395-397, 1983.
  • L'Hote and Knowles, Exp Cell Res, 304(2):417-431, 2005.
  • Li et al., Cell Death Differ, 12(3):292-303, 2005.
  • Li et al., Oncogene, 22(10):1501-1510, 2003.
  • Libra et al., BMC Cancer, 4:11, 2004.
  • Lim et al., Nature, 433(7027):769-773, 2005.
  • Lim et al., Nature, 433(7027):769-773, 2005.
  • Lin and Gelman, Cancer Res, 57(11):2304-2312, 1997.
  • Lin et al., Gastroenterology, 128(1):9-23, 2005.
  • Liu and Erikson, Proc Natl Acad Sci USA, 100(10):5789-5794, 2003.
  • Liu et al., Cancer Res, 66(7):3593-3602, 2006.
  • Liu et al., Int J Hematol, 84(5):425-431, 2006.
  • Lo Vasco et al., Leukemia, 18(6):1122-1126, 2004.
  • Luboshits et al., Cancer Res, 59(18):4681-4687, 1999.
  • Lucke et al., Cancer Res, 61(2):482-485, 2001.
  • Luger et al., Blood, 87(4):1326-1334, 1996.
  • Lujambio et al., Cancer Res, 67(4):1424-1429, 2007.
  • Lukiw, Neuroreport, 18(3):297-300, 2007.
  • Ma et al., Nature, 449(7163):682-688, 2007.
  • Maaser et al., Clin Cancer Res, 11(5):1751-1756, 2005.
  • Makeyev et al., Mol Cell, 27(3):435-448, 2007.
  • Malumbres and Barbacid, Nat Rev Cancer, 1(3):222-231, 2001.
  • Manara et al., Mol Biol Cell, 17(4):1910-1921, 2006.
  • Mao et al., Blood, 101(4):1513-1519, 2003.
  • Markowitz et al., Science, 268(5215):1336-1338, 1995.
  • Markowitz, Biochim Biophys Acta, 1470(1):M13-20, 2000.
  • Marone et al., Int J Cancer, 75(1):34-39, 1998.
  • Marsters et al., Recent Prog. Horm. Res., 54:225-234, 1999.
  • Martinez-Lorenzo et al., Int J Cancer, 75(3):473-481, 1998.
  • Martini et al., Cancer Res, 62(19):5408-5412, 2002.
  • Massague et al., Cell, 103(2):295-309, 2000.
  • Mathas et al., Embo J, 21(15):4104-4113, 2002.
  • McGary et al., Cancer Biol Ther, 1(5):459-465, 2002.
  • McInroy and Maatta, Biochem Biophys Res Commun, 360(1):109-114, 2007.
  • Mendrzyk et al., J Clin Oncol, 23(34):8853-8862, 2005.
  • Menendez et al., Proc Natl Acad Sci USA, 101(29):10715-10720, 2004.
  • Merle et al., Gastroenterology, 127(4):1110-1122, 2004.
  • Mishima et al., Brain Res, 1131(1):37-43, Epub Dec. 19, 2006. 2007.
  • Moller et al., Int J Cancer, 57(3):371-377, 1994.
  • Momand et al., Nucleic Acids Res, 26(15):3453-3459, 1998.
  • Moran et al., Clin Cancer Res, 8(12):3803-3812, 2002.
  • Mori et al., Cancer Res., 54(13):3396-3397, 1994.
  • Mori et al., Gastroenterology, 131(3):797-808, 2006.
  • Mori et al., Int J Cancer, 111(4):548-557, 2004.
  • Morishita et al., Hepatology, 40(3):677-686, 2004.
  • Mundt et al., Biochem Biophys Res Commun, 239(2):377-385, 1997.
  • Murphy et al., J Clin Pathol, 58(5):525-534, 2005.
  • Nakagawa et al., Oncogene, 23(44):7366-7377, 2004.
  • Nakayama et al., Am J Pathol, 149(6):1931-1939, 1996.
  • Nakayama et al., Cancer, 92(12):3037-3044, 2001.
  • Nakayama et al., Mod Pathol, 12(1):61-68, 1999.
  • Nauert et al., Curr Biol, 7(1):52-62, 1997.
  • Nawa et al., J Cell Biochem, 79(2):202-212, 2000.
  • Nerlov, Nat Rev Cancer, 4(5):394-400, 2004.
  • Ngan et al., Br J Cancer, 96(6):986-992, 2007.
  • Niwa et al., Clin Cancer Res, 7(2):285-289, 2001.
  • Nobori et al., Nature, 368(6473):753-756, 1994.
  • O'Connor et al., Embo J, 17(2):384-395, 1998.
  • Oh et al., Am J Pathol, 166(1):73-80, 2005.
  • Ohsaki et al., Cancer Res, 52(13):3534-3538, 1992.
  • Okamoto et al., Proc. Natl. Acad. Sci. USA, 91(23):11045-11049, 1994.
  • Olsen et al., Dev. Biol., 216:671, 1999.
  • Olsnes et al., Cancer Immunol Immunother, 55(7):830-840, 2006.
  • Opalinska et al., Clin Cancer Res, 11(13):4948-4954, 2005.
  • Orlow et al., Cancer Res, 54(11):2848-2851, 1994.
  • Ott et al., Oncogene, 26(33):4863-4871, 2007.
  • Ovcharenko et al., Rna, 11(6):985-993, 2005.
  • Ozaki et al., Cancer Res, 60(22):6519-6525, 2000.
  • Pabst et al., Nat Genet, 27(3):263-270, 2001.
  • Paik et al., Cell, 128(2):309-323, 2007.
  • Parekh et al., Biochem Pharmacol, 63(6):1149-1158, 2002.
  • Pascale et al., Hepatology, 42(6):1310-1319, 2005.
  • Passegue et al., Cell, 119(3):431-443, 2004.
  • Payton and Coats, Int J Biochem Cell Biol, 34(4):315-320, 2002.
  • Payton et al., Oncogene, 21(55):8529-8534, 2002.
  • PCT Appln. WO 0168255
  • PCT Appln. WO 03020898
  • PCT Appln. WO 03022421
  • PCT Appln. WO 03023058
  • PCT Appln. WO 03029485
  • PCT Appln. WO 03040410
  • PCT Appln. WO 03053586
  • PCT Appln. WO 03066906
  • PCT Appln. WO 03067217
  • PCT Appln. WO 03076928
  • PCT Appln. WO 03087297
  • PCT Appln. WO 03091426
  • PCT Appln. WO 03093810
  • PCT Appln. WO 03100448A1
  • PCT Appln. WO 04020085
  • PCT Appln. WO 04027093
  • PCT Appln. WO 09923256
  • PCT Appln. WO 09936760
  • PCT Appln. WO 93/17126
  • PCT Appln. WO 95/11995
  • PCT Appln. WO 95/21265
  • PCT Appln. WO 95/21944
  • PCT Appln. WO 95/21944
  • PCT Appln. WO 95/35505
  • PCT Appln. WO 96/31622
  • PCT Appln. WO 97/10365
  • PCT Appln. WO 97/27317
  • PCT Appln. WO 9743450
  • PCT Appln. WO 99/35505
  • PCT Appln. WO0138580
  • PCT Appln. WO03100012
  • Pearl, Curr Opin Genet Dev, 15(1):55-61, 2005.
  • Perez-Mancera and Sanchez-Garcia, Semin Cancer Biol, 15(3):206-214, 2005.
  • Petit et al., Genomics, 57(3):438-441, 1999.
  • Pietras et al., Cancer Cell, 3(5):439-443, 2003.
  • Pietras et al., Oncogene, 17(17):2235-2249, 1998.
  • Prentice et al., Oncogene, 24(49):7281-7289, 2005.
  • Pretner et al., Anticancer Res, 26(1A):9-22, 2006.
  • Prieto-Sanchez et al., Br J Haematol, 133(6):642-645, 2006.
  • Prochownik et al., Genes Chromosomes Cancer, 22(4):295-304, 1998.
  • Pruitt et al., Nucleic Acids Res, 33(Database issue):D501-504, 2005.
  • Qian et al., Proc Natl Acad Sci USA, 99(23):14925-14930, 2002.
  • Qin et al., Proc. Natl. Acad. Sci. USA, 95(24):14411-14416, 1998.
  • Qiu et al., Am J Pathol, 162(6):1961-1974, 2003.
  • Rapp et al., Proc Natl Acad Sci USA, 80(14):4218-4222, 1983.
  • Ree et al., Cancer Res, 59(18):4675-4680, 1999.
  • Reiter et al., Clin Cancer Res, 12(17):5136-5141, 2006.
  • Remington's Pharmaceutical Sciences” 15th Edition, pages 1035-1038 and 1570-1580, 1990.
  • Rincon-Arano et al., Cancer, 97(3):575-585, 2003.
  • Ritch et al., J Biol Chem, 278(23):20971-20978, 2003.
  • Rivest et al., Can J Neurol Sci, 26 Suppl 2:S34-38, 1999.
  • Robles et al., J Biol Chem., 277(28):25431-25438, 2002.
  • Romieu-Mourez et al., Cancer Res, 61(9):3810-3818, 2001.
  • Rosenkilde and Schwartz, Apmis, 112(7-8):481-495, 2004.
  • Rothhammer et al., Cell Mol Life Sci, 61(1):118-128, 2004.
  • Rous, J Exp Med, 13:397-411, 1911.
  • Ru et al., Oncogene, 21(30):4673-4679, 2002.
  • Rust et al., J Clin Pathol, 58(5):520-524, 2005.
  • Rutka et al., Int J Dev Neurosci, 17(5-6):503-515, 1999.
  • Sacchi et al., Leukemia, 2(1):12-18, 1988.
  • Sacchi et al., Science, 231(4736):379-382, 1986.
  • Saeki et al., Cancer, 89(8):1670-1676, 2000.
  • Saigusa et al., Cancer Sci, 96(10):676-683, 2005.
  • Sakamoto et al., Thyroid, 1(4):347-351, 1991.
  • Sambrook and Russell, Molecular Cloning: A Laboratory Manual 3rd Ed., Cold Spring Harbor Laboratory Press, 2001.
  • Sambrook et al., In: DNA microaarays: a molecular cloning manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 2003.
  • Sambrook et al., In: Molecular cloning: a laboratory manual, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989.
  • Sanchez-Aguilera et al., Blood, 103(6):2351-2357, 2004.
  • Sano et al., Histopathology, 46(5):532-539, 2005.
  • Sasaki et al., J Surg Res, 101(2):242-247, 2001.
  • Sasaki et al., Lung Cancer, 35(2):149-154, 2002.
  • Schulze-Bergkamen et al., BMC Cancer, 6:232, 2006.
  • Schuster and Porse, Biochim Biophys Acta, 1766(1):88-103, 2006.
  • Scotlandi et al., Cancer Res, 60(18):5134-5142, 2000.
  • Seggerson et al., Dev. Biol., 243:215, 2002.
  • Seike et al., Lung Cancer, 38(3):229-234, 2002.
  • Selvakumaran et al., Mol Cell Biol, 14(4):2352-2360, 1994.
  • Semple and Duncker, Biotechnol Adv, 22(8):621-631, 2004.
  • Serrano et al., Acta Haematol, 116(2):77-89, 2006.
  • Serrano et al., Nature, 366:704-707, 1993.
  • Serrano et al., Science, 267(5195):249-252, 1995.
  • Shao et al., Oncogene, 17(4):527-532, 1998.
  • Shapira et al., Cancer, 103(7):1336-1346, 2005.
  • Sherr and McCormick, Cancer Cell, 2(2):103-112, 2002.
  • Sherr and Roberts, Genes Dev, 13(12):1501-1512, 1999.
  • Shetty et al., Br J Cancer, 93(11):1295-1300, 2005.
  • Shibahara et al., Anticancer Res, 25(3B): 1881-1888, 2005.
  • Shigeishi et al., Oncol Rep, 15(4):933-938, 2006.
  • Shigemasa et al., Jpn J Cancer Res, 93(5):542-550, 2002.
  • Shinoura et al., Cancer Gene Ther, 7(2):224-232, 2000.
  • Sieghart et al., J Hepatol, 44(1):151-157, 2006.
  • Singh et al., Cancer Res, 63(9):2306-2311, 2003.
  • Slotky et al., Breast Cancer Res, 7(5):R737-744, 2005.
  • Smirnova et al., Eur J Neurosci, 21(6):1469-1477, 2005.
  • Smith et al., Biochem Biophys Res Commun, 234(2):397-405, 1997.
  • Smith et al., Br J Cancer, 93(6):719-729, 2005.
  • Sohn et al., Am J Pathol, 153(6):1937-1945, 1998.
  • Sommers et al., Cancer Res, 52(19):5190-5197, 1992.
  • Soufla et al., Cancer Lett, 221(1):105-118, 2005.
  • Span et al., Oncogene, 21(55):8506-8509, 2002.
  • Sparmann and Bar-Sagi, Cancer Cell, 6(5):447-458, 2004.
  • Stark et al., Blood, 63(2):415-420, 1984.
  • Steensma et al., Blood, 105(2):443-452, 2005.
  • Stehelin et al., Nature, 260(5547):170-173, 1976.
  • Steinbach et al., Clin Cancer Res, 12(8):2434-2441, 2006.
  • Stepanova et al., Oncogene, 19(18):2186-2193, 2000.
  • Strebhardt and Ullrich, Nat Rev Cancer, 6(4):321-330, 2006.
  • Su et al., Clin Cancer Res, 7(5):1320-1324, 2001.
  • Sui et al., Oncol Rep, 15(4):765-771, 2006.
  • Sutter et al., Int J Cancer, 102(4):318-327, 2002.
  • Tagawa et al., Oncogene, 24(8):1348-1358, 2005.
  • Takanami, Oncol Rep, 13(4):727-731, 2005.
  • Takimoto et al., Biochem Biophys Res Commun, 251(1):264-268, 1998.
  • Tan et al., Leuk Res, 27(2):125-131, 2003.
  • Tanaka et al., Proc Natl Acad Sci USA, 95(17):10164-10169, 1998.
  • Tanner et al., Clin Cancer Res, 6(5):1833-1839, 2000.
  • Thome, Nat Rev Immunol, 4(5):348-359, 2004.
  • Toh et al., Br J Cancer, 79(11-12): 1723-1726, 1999.
  • Toh et al., Int J Cancer, 74(4):459-463, 1997.
  • Toh et al., J Biol Chem, 269(37):22958-22963, 1994.
  • Tokuhara et al., Clin Cancer Res, 7(12):4109-4114, 2001.
  • Tokuhara et al., Int Surg, 88(1):25-33, 2003.
  • Toriumi et al., Anticancer Res, 24(4):2455-2463, 2004.
  • Traub et al., Breast Cancer Res Treat, 99(2):185-191, 2006.
  • Tsai et al., J Natl Cancer Inst, 85(11):897-901, 1993.
  • Turhan et al., Hematol Cell Ther, 40(5):217-221, 1998.
  • Turner et al., Nat Rev Cancer, 4(10):814-819, 2004.
  • Tuveson et al., Cancer Cell, 4(2):95-98, 2003.
  • U.K. Patent 1,529,202
  • UK 8 803 000
  • Ulisse et al., Int J Cancer, 119(2):275-282, 2006.
  • Upton et al., Am J Surg Pathol, 10(8):560-567, 1986.
  • Vaday et al., Prostate, 66(2):124-134, 2006.
  • Vanhaesebroeck et al., Trends Biochem Sci, 22(7):267-272, 1997.
  • Venkatasubbarao et al., Anticancer Res, 20(1A):43-51, 2000.
  • Viard-Leveugle et al., J Pathol, 201(2):268-277, 2003.
  • Visvanathan et al., Genes Dev, 21(7):744-749, 2007.
  • Vlodavsky and Soustiel, J Neurooncol, 81(1):1-7, 2007.
  • Vogt et al., Cell Cycle, 4(7):908-913, 2005.
  • Vos et al., J Biol Chem, 278(30):28045-28051, 2003.
  • Wang et al., Mol Carcinog, 31(2):90-100, 2001.
  • Wang et al., Oncogene, 22(10):1486-1490, 2003.
  • Wang et al., Proc Natl Acad Sci USA, 98(20):11468-11473, 2001.
  • Wechsler et al., Cancer Res, 57(21):4905-4912, 1997.
  • Weichert et al., Int J Oncol, 23(3):633-639, 2003.
  • Weil et al., Biotechniques, 33(6):1244-1248, 2002.
  • Wetzel et al., J Gene Med, 3(4):326-337, 2001.
  • Wiemer, Eur J Cancer, 43(10):1529-1544, 2007.
  • Wiemer, Eur J Cancer, 43(10):1529-1544, 2007.
  • Wikman et al., Oncogene, 21(37):5804-5813, 2002.
  • Wilkerson et al., J Cutan Pathol, 33(10):663-666, 2006.
  • Wohlschlegel et al., Am J Pathol, 161(1):267-273, 2002.
  • Wolowiec et al., Leuk Lymphoma, 35(1-2):147-157, 1999.
  • Wooster and Weber, N Engl J Med, 348(23):2339-2347, 2003.
  • Wooster and Weber, N Engl J Med, 348(23):2339-2347, 2003.
  • Woszczyk et al., Med Sci Monit, 10(1):CR33-37, 2004.
  • Wu et al., Eur J Cancer, 38(14):1838-1848, 2002.
  • Wuilleme-Toumi et al., Leukemia, 19(7):1248-1252, 2005.
  • Xi et al., Clin Cancer Res, 12(8):2484-2491, 2006a.
  • Xi et al., Clin Chem, 52(3):520-523, 2006b.
  • Xia et al., Cancer Res, 61(14):5644-5651, 2001.
  • Yaal-Hahoshen et al., Clin Cancer Res, 12(15):4474-4480, 2006.
  • Yamada et al., Anticancer Res, 22(6A):3303-3307, 2002.
  • Yamamoto et al., Int J Oncol, 13(2):233-239, 1998.
  • Yang et al., Blood, 101(8):3205-3211, 2003.
  • Yasui et al., Hepatology, 35(6):1476-1484, 2002.
  • Ye et al., Biochim Biophys Acta, 1493(3):373-377, 2000.
  • Yeatman, Nat Rev Cancer, 4(6):470-480, 2004.
  • Yi et al., Chin Med Sci J, 18(2):87-92, 2003.
  • Ying et al., Clin Cancer Res, 11 (18):6442-6449, 2005.
  • Yoo et al., J Korean Med Sci, 20(1):50-55, 2005.
  • Yu and Feig, Oncogene, 21(49):7557-7568, 2002.
  • Yu et al., Nat Rev Immunol, 7(1):41-51, 2007.
  • Zangemeister-Wittke and Huwiler, Cancer Biol Ther, 5(10):1355-1356, 2006.
  • Zhang et al., Immunity, 21(6):853-863, 2004.
  • Zhou et al., Life Sci, 78(22):2643-2649, 2006.
  • Zrihan-Licht et al., J Biol Chem, 273(7):4065-4072, 1998.

Claims
  • 1. A method of modulating gene expression in a cell comprising administering to the cell an amount of an isolated nucleic acid comprising a miR-10 nucleic acid sequence or a miR-10 inhibitor in an amount sufficient to modulate the expression of one or more genes identified in Table 1, 3, 4, or 5.
  • 2. The method of claim 1, wherein the cell is in a subject having, suspected of having, or at risk of developing a metabolic, an immunologic, an infectious, a cardiovascular, a digestive, an endocrine, an ocular, a genitourinary, a blood, a musculoskeletal, a nervous system, a congenital, a respiratory, a skin, or a cancerous condition.
  • 3. (canceled)
  • 4. The method of claim 2, wherein the cancerous condition is astrocytoma, acute lymphoblastic leukemia, acute myeloid leukemia, breast carcinoma, bladder carcinoma, cervical carcinoma, chronic lymphoblastic leukemia, chronic myeloid leukemia, colorectal carcinoma, chondrosarcoma, endometrial carcinoma, Ewing's sarcoma, fibrosarcoma, glioma, glioblastoma, glioblastoma multiforme, gastric carcinoma, hepatoblastoma, hepatocellular carcinoma, Hodgkin lymphoma, leukemia, lung carcinoma, leiomyoma, liposarcoma, melanoma, mantle cell lymphoma, multiple myeloma, mesothelioma, neuroblastoma, non-Hodgkin lymphoma, nasopharyngeal carcinoma, non-small cell lung carcinoma, ovarian carcinoma, oesophageal carcinoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, retinoblastoma, renal cell carcinoma, squamous cell carcinoma of the head and neck, thyroid carcinoma, T-cell leukemia, or Wilm's tumor, wherein the modulation of one or more gene is sufficient for a therapeutic response.
  • 5. The method of claim 2, wherein the cancerous condition is acute myeloid leukemia.
  • 6. The method of claim 1, wherein the expression of a gene is down-regulated.
  • 7. The method of claim 1, wherein the expression of a gene is up-regulated.
  • 8. (canceled)
  • 9. (canceled)
  • 10. The method of claim 1, wherein the cell is a cancer cell.
  • 11. The method of claim 10, wherein the cancer cell is an epithelial, an endothelial, a mesothelial, a stromal, a mucosal, a brain, a glial, a neuronal, a blood, a leukemic, an endometrial, an esophageal, a lung, a cardiovascular, a liver, a lymphoid, a breast, a bone, a connective tissue, a fat, a retinal, a thyroid, a glandular, a salivary gland, an adrenal, a pancreatic, a stomach, an intestinal, a kidney, a bladder, a colon, a colorectal, a prostate, a, an ovarian, a cervical, a, a splenic, a skin, a smooth muscle, a cardiac muscle, or a striated muscle cell.
  • 12. The method of claim 1, wherein the isolated miR-10 nucleic acid or miR-10 inhibitor is a recombinant nucleic acid.
  • 13. (canceled)
  • 14. The method of claim 12, wherein the recombinant nucleic acid is DNA.
  • 15.-17. (canceled)
  • 18. The method of claim 1, wherein the miR-10 nucleic acid or miR-10 inhibitor is a synthetic nucleic acid.
  • 19. (canceled)
  • 20. The method of claim 1, wherein the miR-10 or miR-10 inhibitor is a hsa-miR-10 or has-miR-10 inhibitor.
  • 21. The method of claim 1, wherein the nucleic acid is administered enterally or parenterally.
  • 22. (canceled)
  • 23. The method of claim 21, wherein parenteral administration is intravascular, intracranial, intrapleural, intratumoral, intraperitoneal, intravascular, intralymphatic, intraglandular, subcutaneous, topical, intrabronchial, intratracheal, intranasal, inhaled, or instilled.
  • 24. The method of claim 1, wherein the nucleic acid is comprised in a pharmaceutical formulation.
  • 25. The method of claim 24, wherein the pharmaceutical formulation is a lipid composition.
  • 26. The method claim 24, wherein the pharmaceutical formulation is a nanoparticle composition.
  • 27. The method of claim 24, wherein the pharmaceutical formulation consists of biocompatible and biodegradable molecules.
  • 28.-43. (canceled)
  • 44. A method of treating a patient diagnosed with or suspected of having or suspected of developing a pathological condition or disease related to a gene modulated by a miRNA comprising the steps of: (a) administering to the patient an amount of an isolated nucleic acid comprising a miR-10 nucleic acid sequence or miR-10 inhibitor in an amount sufficient to modulate a cellular pathway or a physiologic pathway associated with one or more genes identified in Table 1, 3, 4, or 5; and(b) administering a second therapy, wherein the modulation of the cellular pathway or physiologic pathway sensitizes the patient to the second therapy.
  • 45.-49. (canceled)
  • 50. A method of assessing a cell, tissue, or subject comprising assessing expression of miR-10 in combination with assessing expression of one or more gene from Table 1, 3, 4, or 5 in at least one sample.
  • 51. (canceled)
Parent Case Info

This application claims priority to U.S. Provisional Patent Application Ser. No. 61/016,264 filed Dec. 21, 2007, which is incorporated herein by reference in its entirety.

Provisional Applications (1)
Number Date Country
61016264 Dec 2007 US