WO2005014854A1 - Materials and methods for colorectal cancer screening, diagnosis, and therapy - Google Patents

Materials and methods for colorectal cancer screening, diagnosis, and therapy Download PDF

Info

Publication number
WO2005014854A1
WO2005014854A1 PCT/EP2004/008819 EP2004008819W WO2005014854A1 WO 2005014854 A1 WO2005014854 A1 WO 2005014854A1 EP 2004008819 W EP2004008819 W EP 2004008819W WO 2005014854 A1 WO2005014854 A1 WO 2005014854A1
Authority
WO
WIPO (PCT)
Prior art keywords
prox
expression
protein
cells
molecule
Prior art date
Application number
PCT/EP2004/008819
Other languages
French (fr)
Inventor
Kari Alitalo
Tatiana Petrova
Antti Nykanen
Original Assignee
Licentia, Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Licentia, Ltd. filed Critical Licentia, Ltd.
Priority to EP04763854A priority Critical patent/EP1654383A1/en
Priority to US10/567,630 priority patent/US8029984B2/en
Publication of WO2005014854A1 publication Critical patent/WO2005014854A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57419Specifically defined cancers of colon
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/154Methylation markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • the present invention relates generally to methods and materials for altering colorectal cancer progression.
  • the present invention also relates to techniques for screening for colon cancer and/or premalignancies.
  • the transcription factor Prox-1 is expressed in a number of tissues during embryonic development, including lens fiber cells, subpbpulation of neurons in brains and neural tube, skeletal muscle, heart, liver, pancreas and lymphatic endothelial cells. Targeted inactivation of Prox-1 results in the defects of eye development because of the failure of lens fiber cells to elongate (Wigle et al., Nat. Genet. 21: 318-22, 1999). Prox-1 is also necessary for the migration of hepatocytes during liver development (Sosa-Pineda et al., Nat. Genet. 25: 254-5, 2000).
  • Prox-1 deficient embryos lack lymphatic vasculature, while the blood vessel development is not affected (Wigle et ab, Cell 98: 769-778, 1999). Recently, others and we have demonstrated the essential role of Prox-1 in the regulation of the lymphatic endothelial phenotype.
  • Notch is a transmembrane protein that acts as a receptor in a cell-cell signaling mechanism, and in combination with other cellular factors, influences differentiation, proliferation and apoptotic events at all stages of development (Artavanis-Tsakonas, Science 284: 770-776, 1999). In animal models, mutations in the Notch receptor have resulted in developmental abnormalities (Joutel et al., Nature 383: 707, 1996; Lb et al., Nature Genet. 16:243, 1997). Cancer treatments generally promote tumor regression by inhibiting tumor cell proliferation, inhibiting angiogenesis (growth of new blood vessels that is necessary to support tumor growth) and/or prohibiting metastasis by reducing tumor cell motility or invasiveness.
  • Colon cancers are a very common malignancy and colon cancers are typically adenocarcinomas, or sometimes carcinoid tumors. Treatment is primarily surgical resection of the colon, although chemotherapy has been found to be beneficial in some cases. These treatment options for colon cancer are of unpredictable and sometimes limited value, especially if the cancer has not been identified and removed at early stages. There continues to exist a need for novel therapies and diagnostic methods for cancer conditions.
  • the present invention addresses one or more ongoing needs by providing materials and methods for screening for and treating cancerous and precancerous conditions, especially colorectal in nature.
  • the invention provides materials and methods to screen a mammalian subject for a cancerous or precancerous condition based on analysis of Prox-1 expression in cells from the mammalian subject.
  • materials and methods are provided for screening colon tissue for signs of cancerous or precancerous pathology.
  • the method includes a method of screening colon tissue for a pathological condition, said method comprising: measuring Prox-1 expression in a biological sample that comprises colon tissue from a mammalian subject, wherein elevated Prox-1 expression in the colon tissue correlates with a pathological phenotype.
  • the determination of elevated Prox-1 expression is generally made by way of a comparison, e.g., to a measurement of Prox-1 expression in healthy colon tissue (from the same subject or others of the same species, preferably matched for sex, age, race, or other characteristics); or to a measurement of Prox-1 expression in diseased (especially neoplastic) colon tissue.
  • a comparison e.g., to a measurement of Prox-1 expression in healthy colon tissue (from the same subject or others of the same species, preferably matched for sex, age, race, or other characteristics); or to a measurement of Prox-1 expression in diseased (especially neoplastic) colon tissue.
  • an increased (e.g., elevated) Prox-1 expression in the colon tissue from the mammalian subject correlates with a pathological phenotype.
  • comparable levels of expression in the tissue from the subject co ⁇ elates with a pathological phenotype.
  • the invention includes a method of screening colon tissue for a pathological condition, the method
  • the invention includes a method of screening colon tissue for a pathological condition, the method comprising steps of: (a) obtaining a biological sample comprising colon tissue from a mammalian subject; (b) measuring Prox-1 expression in the colon tissue; and (c) comparing Prox-1 expression in the colon tissue to Prox-1 expression in healthy colon tissue, wherein increased Prox-1 expression in the colon tissue corcelates with a pathological phenotype.
  • pathological condition is intended to include any abnormality or evidence of disease that warrants medical treatment or monitoring due to concern of developing disease.
  • the method can be characterized as a screen for colon cancer or colorectal cancers, and increased Prox-1 expression in the colon tissue is scored as being indicative of a cancerous or precancerous condition.
  • the method can be combined with any other molecular, cellular, pathological, or patient symptom criteria to assist a medical practitioner in early diagnosis and therapeutic or prophylactic therapy.
  • the method further comprises measuring expression of at least one gene or protein selected from the group consisting of CD44, Encl, and ID2 in the colon tissue, wherein elevated Prox-1 expression and elevated expression of the at least one gene/protein in the colon tissue conelate. with a pathological phenotype.
  • the method further comprising measuring activation of -catemn/TCF pathway in the colon tissue, wherein activation of the -catemn/TCF pathway and elevated Prox-1 expression in the colon tissue correlate with a pathological phenotype.
  • Activation of the -catemn/TCF pathway can be measured by a variety of indicators, including mutations in an APC gene; mutations in a -catenin gene; and nuclear localization of -catenin.
  • the biological sample is any tissue or fluid sample obtained in any way from a mammalian subject that includes cells from the large intestine. Biopsies or other surgically removed specimens are preferred. Stool or feces may contain sufficient colon tissue for some embodiments of the assay.
  • the assay may be performed on any mammalian subject, including laboratory animals used in cancer research, livestock, and domestic pets. Humans are most prefened. Any available technique can be used for measuring Prox-1 expression, including direct and indirect techniques.
  • the measuring comprises measuring Prox-1 protein in the biological sample.
  • Prefened techniques for measuring amounts or concentrations of Prox-1 protein in a sample are immunological techniques that involve use of a polyclonal or monoclonal antibody that specifically binds Prox-1, or use of a Prox-1 -binding fragment of such an antibody.
  • the measuring comprises contacting the colon tissue with a Prox-1 antibody or antigen-binding fragment thereof.
  • Quantification of the amount of bound antibody provides a measurement of Prox-1 protein expressed in the sample
  • frnmunoassays such as radioimmunoassay, immunoradiometric assay (labeled antibody), or an enzyme- linked immunosorbent assay (ELISA) are contemplated.
  • the measuring comprises measuring Prox-1 mRNA in the colon tissue. Elevated levels of Prox-1 mRNA in the sample are scored as elevated Prox-1 expression. Any available assay for measuring specific oligonucleotides is suitable. Prefened materials for such measurements are oligonucleotide probes complementary to all or a portion of the Prox-1 mRNA sequence.
  • Probes of at least 14 and more preferably 18 nucleotides are prefened to assure specificity.
  • One technique for measuring Prox-1 mRNA comprises in situ hybridization to measure Prox-1 mRNA in the colon sample. Other techniques involve steps of isolating mRNA from the colon tissue and measuring Prox-1 mRNA in the isolated mRNA, for example, by Northern hybridization procedures.
  • quantitative reverse transcriptase polymerase chain reaction (PCR), real-time PCR, or other PCR techniques are employed to quantitatively amplify Prox- 1 mRNA (relative to control samples) to provide a quantitative measurement of Prox- 1 mRNA in the colon tissue.
  • Prox-1 expression is measured indirectly by measuring a functional property of Prox-1, such as measuring Prox-1 binding to DNA or downstream Prox-1 transcription factor effects.
  • the screening method further includes a comparing step whereby
  • Prox-1 expression in the colon tissue is compared to Prox-1 expression in healthy colon tissue, wherein increased Prox-1 expression in the colon tissue coreelates with a pathological phenotype.
  • Prox-1 expression is elevated in a statistically significant manner in pathological specimens studied, compared to healthy colon tissue samples.
  • the comparison is performed by taking simultaneous or sequential measurements of a test sample and a sample of colon tissue that is known to be taken from healthy tissue.
  • data is accumulated on the quantity of Prox-1 mRNA or protein in healthy tissues, and the amount that is measured in the colon tissue from the biological sample is compared to this predetermined amount.
  • comparing Prox-1 measurements from a test sample to measurements from a cancerous or precancerous condition can provide an equivalent indication of the presence or absence of the pathological condition, wherein a test sample with Prox-1 expression comparable to the elevated level observed in a cancer correlates with a pathological phenotype.
  • a database of Prox-1 measurements from colon tissues can be developed, preferably containing information about healthiness or disease of the tissue; age, sex, race/ethnicity of the donor, and location from which the sample was taken.
  • comparisons can be analyzed using statistical analysis to determine the statistical significance of a measurement's deviation from a mean, optionally selecting entries from the database by selecting for the patient's age, sex, ethnicity, and other factors to best match the patient (mammalian subject) being tested.
  • Such statistical analysis permits establishment of one or more "cutoff" values for the Prox-1 measurement that are conelated with a likelihood of having, or developing, a cancerous condition. If elevated Prox-1 is detected, then in a prefened embodiment, the method further comprises a step of administering to a human subject identified as having a pathological condition characterized by increased Prox-1 expression in colon tissue a composition comprising a Prox-1 inhibitor. .
  • the invention provides a method of inliibiting the growth of colon cancer cells, such as colon carcinoma cells, colon adenoma cells, or colon adenocarcinoma cells in a mammalian subject comprising a step of: administering to the subject a composition comprising a molecule that suppresses expression of Prox-1, thereby inhibiting the growth of colon carcinoma cells.
  • colon cancer cells such as colon carcinoma cells, colon adenoma cells, or colon adenocarcinoma cells in a mammalian subject
  • administering to the subject a composition comprising a molecule that suppresses expression of Prox-1, thereby inhibiting the growth of colon carcinoma cells.
  • the method further comprises a step of identifying a mammalian subject with a colon cancer characterized by increased Prox-1 expression.
  • the composition is administered to such a patient after the identifying step, because cancers characterized by the elevated expression are expected to be the cancers most likely to respond to the inhibitors.
  • Exemplary cancers (neoplasms) in which Prox-1 elevation has been observed include colorectal adenomas and colorectal carcinomas, as described below in greater detail.
  • the composition to be administered preferably includes, in addition to the Prox-1 inhibitor, a pharmaceutically acceptable diluent, adjuvant, or carrier medium.
  • the composition optionally includes additional antineoplastic agents.
  • inhibitor molecules include antisense oligonucleotides that inhibit Prox-1 expression; micro-RNA that inhibits Prox-1 expression; small (short) interfering RNA (siRNA) that inhibit Prox-1 expression (e.g., siRNA that comprise at least one nucleotide sequence set forth in SEQ ID NOS: 4, 5, 6, and 7); zinc finger proteins that inhibit Prox-1 expression; polypeptides that act as dominant negative form of Prox-1 protein, such as Prox-1 forms that have a disrupted DNA binding domain or transactivation domain(s); polynucleotides that encode dominant-negative Prox-1 proteins; Prox-1 antibodies and fragments thereof; polynucleotides that encode Prox-1 antibodies or encode " polypeptides that comprise Prox-1 binding domains; small molecules discovered and- designed through screening based on the teachings herein, and so
  • U.S. Patent Application Publication No. 2003/0224516 discloses exemplary molecules for inhibiting Prox-1 expression and is incorporated herein by reference.
  • the inhibitor is preferably administered in an amount and in a regimen that halts or inhibits neoplastic growth of the affected colorectal tissue.
  • the tissue itself preferably reverts to a non-transformed, more healthy looking phenotype.
  • one apparent benchmark of beneficial administration is an increase in Notch-1 signaling.
  • the composition is administered in an amount effective to suppress Prox-1 expression and increase Notch 1 signaling.
  • Other indications of efficacy relate to modulation of prostaglandin synthesis.
  • the composition is administered in an amount effective to increase 15-PDGH activity or decrease prostaglandin D2 synthase activity.
  • colorectal cancers also are often characterized by increases in the -catenin TCF signaling pathway, relative to what is observable in healthy colorectal tissue.
  • the regimen further comprises administering to the subject an inhibitor of the -catenin/TCF signaling pathway.
  • the patient's diseased tissue is first pre-screened for elevated expression/signaling of this pathway.
  • the categories of inhibitors described above for Prox-1 are specifically contemplated for the -catenin/TCF pathway as well.
  • the inhibitor of the -catenin/TCF signaling pathway is dominant negative form of TCF-4.
  • the inhibitor optionally targets (inhibits) TCF-4, ⁇ -catenin, or c-myc expression or activity.
  • administration of the Prox-1 inhibitor is combined with administration of a COX-2 inhibitor, such as any of the increasing class of non-steroidal anti-inflammatory agents.
  • administration of the Prox-1 inhibitor is combined with administration of a Notch signaling pathway agonist, such as a Notch . ligand or expression vector to cause expression of a Notch ligand.
  • exemplary Notch ligands include Jaggedl, Jagged2, Deltal, Delta3, Delta4, or Senate.
  • a molecule that comprises an inhibitor of DNA methyltransferases Such inhibitors are themselves contemplated as efficacious for inhibiting Prox-1 expression, and can be combined with any other Prox-1 inhibitor described herein for combination therapy.
  • An exemplary methyltransferase inhibitor is 5-aza-2'-deoxycytidine.
  • the Prox-1 inhibitor composition is admimstered in combination with any known antineoplastic agent that is used in cancer therapy.
  • the Prox-1 inhibitor and/or Cox-2 inhibitor are combined (in a medicament or as a combination therapy) with an agent that induces differentiation in colorectal cancer cell lines.
  • Exemplary agents include 1,25- dihydroxyvitamin D3 and analogs thereof; butyrate; and retinoids.
  • the Prox-1 inhibitor composition can be administered simultaneously with the other active agents, which may be in admixture with the Prox-1 inhibitor, or may be in a separate composition.
  • Each composition preferably includes a pharmaceutically acceptable diluent, adjuvant, or carrier. When the agents are separately administered, they may be administered in any order.
  • the invention includes a method of inhibiting Prox-1 function in a mammalian subject having a disease characterized by of Prox-1 over-expression in cells, comprising the step of administering to said mammalian subject a composition, said composition comprising a compound effective to inhibit Prox-1 function in cells.
  • the invention includes the use of a Prox-1 inhibitor in the manufacture of a medicament for the treatment of a disease characterized by Prox-1 over-expression in cells, especially cancerous or precancerous cells of colorectal origin.
  • the medicament optionally includes the additional agents described above, either in admixture with the Prox-1 inhibitor or separated, yet packaged together (preferably with instructions for treating the disease).
  • the invention provides a method of screening for Prox-1 modulators comprising the steps of: (a) contacting a test molecule with Prox-1 protein, or a nucleic acid comprising a nucleotide sequence that encodes Prox-1 protein, under conditions which permit the interaction of the test molecule with the Prox-1 protein or nucleic acid; and (b) measuring the interaction between the test molecule and Prox-1 protein or the nucleic acid, wherein a test molecule that binds the Prox-1 protein or nucleic acid is identified as a Prox-1 modulator.
  • Test molecule refers to the molecule that is under evaluation for the ability to modulate (i.e., increase or decrease) the activity of Prox-1 protein.
  • test molecule that is a Prox-1 modulator will interact directly with Prox- 1.
  • the screens described herein can identify test molecules that modulate Prox-1 protein activity indirectly, such as by affecting Prox-1 gene expression.
  • the screens work with essentially any test molecule, and the invention is not Umited in this manner.
  • the test molecule is a protein, a carbohydrate, a lipid, or a nucleic acid.
  • Molecules which regulate Prox-1 expression include nucleic acids which are complementary to nucleic acids encoding a Prox-1 protein, or are complementary to nucleic acid sequences which direct or control the expression of Prox-1 protein, and which act as anti-sense regulators of expression.
  • the test molecule may be a member of a chemical library, such as libraries commonly maintained in large pharmaceutical companies or libraries generated combinatorially.
  • the test molecule interacts with Prox-1 by binding to the Prox-1 DNA binding domain, thereby effecting Prox-1 activity.
  • the assays set forth herein can be readily modified by adding such additional test compounds either simultaneous with, or subsequent to, or prior to, the first test compound.
  • the measurement of the interaction of test molecules with Prox-1 may is carried out using solution-phase assays or immunoassays.
  • the invention provides a method of screening for modulators of binding between a DNA and Prox-1 protein comprising steps of: (a) contacting a DNA with a Prox-1 protein in the presence and in the absence of a putative modulator compound; (b) detecting binding between the DNA and the Prox-1 protein in the presence and absence of the putative modulator compound; and (c) identifying a modulator compound based on a decrease or increase in binding between the DNA and the Prox-1 protein in the presence of the putative modulator compound, as compared to binding in the absence of the putative modulator compound.
  • molecules that modulate binding between DNA and Prox-1 are formulated into a composition or a growth media for contacting a cell from a colorectal cancer or colorectal cancer cell Une, and a modulator that inhibits growth of the cell is selected as a prefened modulator for development as a therapeutic.
  • the invention provides a method of screening for modulators of binding between a DNA and Prox-1 protein comprising steps of: (a) contacting a DNA with a Prox-1 protein in the presence and in the absence of a putative modulator compound; (b) detecting binding between the DNA and the Prox-1 protein in the presence and absence of the putative modulator compound; and (c) identifying a modulator compound based on a decrease or increase in differentiation in the presence of the putative modulator compound, as compared to differentiation in the absence of the putative modulator compound.
  • In vivo screening also is contemplated, either in addition to or in place of in vitro screening.
  • the test compound preferably is formulated into a pharmaceutically acceptable diluent, adjuvant, or earner.
  • this formulation is administered to a mammal with pathological (e.g., cancerous) Prox-1 expressing colon tissue, and the efficacy of the formulation at inhibiting disease progression is monitored.
  • pathological e.g., cancerous
  • a method described above optionally further comprises steps of formulating a composition comprising the selected Prox-1 modulator and a pharmaceutically acceptable carrier; administering the composition to a mammalian subject having a colorectal cancer; and monitoring the m-immalian subject for growth, metastasis, shrinkage, or disappearance of the colorectal cancer.
  • “Putative modulator compounds” are analogous to the "test molecules” described above in that they are alleged to have an effect on Prox-1 protein activity " and are being identified as such using the methods described herein.
  • detecting DNA binding to Prox-1 protein and identifying an increase or decrease of DNA binding to Prox-1 protein employs immuno-based assays or various other assays that measure biological activity.
  • embodied by the invention are methods wherein identifying a modulator compound the use of proliferation and/or differentiation assays.
  • siRNA short interfering RNA
  • the siRNA molecule can be adapted for use to treat colorectal cancer and any other indications that respond to the level of Prox-1.
  • the siRNA molecule comprises a sense region and an antisense region.
  • the antisense region comprises sequence complementary to an RNA sequence encoding Prox-1, or a fragment thereof, and the sense region comprise sequence complementary to the antisense region.
  • the siRNA molecule can comprise two nucleic acid fragments, wherein one fragment comprises the sense region and the second fragment comprises the antisense region of said siRNA molecule.
  • a siRNA molecule of the invention can comprise any contiguous Prox-1 sequence.
  • the siRNA constructs are between 18 and 100 nucleotides in length.
  • the siRNA constructs are 21 nucleotides in length.
  • the sense region of a siRNA molecule of the invention comprises a 3 '-terminal overhang and the antisense region comprises a 3'- terminal overhang.
  • the 3 '-terminal overhangs each are preferably from 1 to 5 nucleo tides. More preferably, the 3 '-terminal overhangs are 2 nucleotides.
  • the antisense region of the 3 '-terminal nucleotide overhang is complementary to RNA encoding Prox-1.
  • the antisense region of Prox-1 siRNA constructs can comprise a sequence complementary to sequence having any of SEQ ID NOs. 4 and 6. Further, the antisense region of Prox-1 siRNA constructs can comprise a having any of SEQ ID NOs. 5 and 7. - In yet an additional embodiment of the invention, compounds, particularly antisense oligonucleotides, which are targeted to a nucleic acid encoding Prox-1, and which modulate the expression of Prox-1 are provided.
  • the antisense oligonucleotides of the invention are preferably complementary to (at least a segment of) the genomic Prox-1 sequence set forth as SEQ ID NO:l.
  • the antisense oligonucleotides of the invention comprise a region complementary to a promoter or other control region, an exon, an intron, or an exon- intron boundary. Also embodied by the present invention are antisense oligonucleotides that are complementary to a region within 20-200 bases of an exon- intron splice junction. As detailed herein, pharmaceutical compositions comprising antisense oligonucleotides are also provided. The foregoing paragraphs are not intended to define every aspect of the invention, and additional aspects are described in other sections, such as the Detailed Description.
  • Figures IA, IB, and IC depict the elevated Prox-1 mRNA levels in colorectal tumors.
  • a cancer RNA profiling array was hybridized to probes for Prox-1 (Fig. IA) and the lymphatic endothelial marker LYVE-l (Fig. IB).
  • Fig. IC illustrates the quantification of dot blot in Fig. 1 A, the asterisk indicating tumor samples in which Prox-1 expression is significantly different from that of the normal tissue (P ⁇ 0.005).
  • Figures 2A-2I depict Prox-1 expression patterns in colon cancer and normal colonic epithelium. Frozen sections of colon adenomas (Fig.
  • FIG. 2A-C depicts the efficacy of Prox-1 suppression for inhibiting SW480R cell growth in soft agar.
  • SW480R cells were transfected with GFP siPRNA, Prox-1 siRNA A16 or Prox-1 siRNA A25 or left untreated, and seeded in soft agar in triplicate. The number of colonies was scored after two weeks of growth. DETAILED DESCRIPTION Demonstrated herein for the first time is the importance of Prox-1 in cancer.
  • Prox-1 gene and protein is overexpressed in colorectal cancers, as compared to healthy colon tissue and other cancer tissues.
  • Prox-1 was overexpressed in 68% of colorectal carcinomas and in 80% of premalignant lesions that were examined, indicating that Prox-1 is important for tumorigenesis, and therefore a useful marker for screening and a useful target for intervention.
  • Prox-1 expression was restricted to two cell types, neuroendocrine cells and non-proliferating cells at the very base of the colonic crypts, a location that conesponds to the stem cell compartment.
  • Contemplated and provided for in the present invention are polynucleotides and polypeptides for screening and diagnosis of colorectal cancer and/or premalignancies.
  • Intervention to suppress Prox-1 expression in colorectal cells resulted in increased activation of Notch signal transduction.
  • ablation of Prox-1 resulted in cell growth arrest and increased expression of epithelial markers.
  • This was accompanied by an upregulation of the cell cycle inhibitor p21cipl which has been shown to be important for the differentiation of intestinal epithelia (Quaroni et al., Am. J. Physiol. Cell Physiol. 279: C1045-57, 2000; Yang et ab, Cancer Res. 61, 565- 9, 2001), and by an increased expression of components of the Notch signaling pathway.
  • Prox-1 may be involved in the maintenance of an undifferentiated state of colonic intestinal stem cells, and overexpression of Prox-1 in cancer cells and resulting inhibition of the Notch signaling pathway may lead to the de-differentiation frequently observed upon malignant transformation.
  • the suppression of Prox-1 expression also negatively regulates prostaglandin activity in the tumor cell lines studied. It is, therefore, contemplated that suppression of Prox-1 or activation of Notch signaling in tumor cells can provide a differentiation therapy for colon carcinoma.
  • the present invention provides compositions and methods for suppressing Prox-1 expression.
  • A. Inhibitory Nucleic Acid Constructs for the Suppression of Prox-1 Expression As discussed herein, Prox-1 is overexpressed in colorectal cancer cells and suppression of Prox-1 expression results in increased Notch signal transduction and modified expression of enzymes of the prostaglandin biosynthetic pathway. This data provides an indication to disrupt the expression or activity of Prox-1 as a method of alleviating the symptoms of and/or inhibiting the growth or metastasis of colon cancer. Such disruption is achieved using any materials or methods available to - - inhibit Prox-1 mRNA or protein expression, or inhibit Prox-1 binding, and any Prox-1 activity.
  • the present section discusses nucleic acid-based methods of disrupting the expression of Prox-1.
  • Polynucleotide products which are useful in this endeavor include antisense polynucleotides, ribozymes, small interfering RNAs, natural or designed microRNAs, triple helix polynucleotides, and novel transcription factors that modulate the expression of Prox-1 protein.
  • Techniques for making and delivering antisense polynucleotides and ribozymes are well known to those in the art and have been extensively described in scientific, patent, and trade literature.
  • Kuneck et ⁇ b (Nucleic Acids Res. ;30:1911-8. 2002)
  • Anti-sense RNA and DNA molecules act to directly block the translation of mRNA by binding to targeted mRNA and preventing protein translation.
  • An example of an antisense polynucleotide is an oligodeoxyribonucleotide derived from the translation initiation site, e.g., between -10 and +10 regions of the relevant nucleotide sequence.
  • Antisense oligonucleotides of 8- 200 nucleotides in length that include at least a portion of this region of the Prox-1 cDNA or genomic sequences set forth as SEQ ID NOs: 1 and 2 (or are complementary to) are prefened Prox-1 inhibitors of the invention.
  • Antisense polynucleotides are typically generated within the cell by expression from antisense constructs that contain the antisense nucleic acid strand as the transcribed strand. Antisense methodology takes advantage of the fact that nucleic acids tend to pair with "complementary" sequences. By complementary, it is meant that polynucleotides are those which are capable of base-pairing according to the standard Watson-Crick complementarity rules.
  • the larger purines will base pair with the smaller pyrimidines to form combinations of guanine paired with cytosine (G:C) and adenine paired with either thymine (A:T) in the case of DNA, or adenine paired with uracil (A:U) in the case of RNA.
  • G:C cytosine
  • A:T thymine
  • A:U uracil
  • Inclusion of less common bases such as inosine, 5-methylcytosine, 6-methyladenine, hypoxanthine and others in hybridizing sequences does not interfere with pairing.
  • ds double-stranded
  • Antisense polynucleotides when introduced into a target cell, specifically bind to their target polynucleotide and interfere with transcription, RNA processing, transport, translation and/or stability.
  • Antisense RNA constructs, or DNA encoding such antisense RNA's may be employed to inhibit gene transcription or translation or both within a host cell, either in vitro or in vivo, such as within a host animal, including a human subject.
  • Antisense constructs may be designed to bind to the promoter and other control regions, exons, introns or even exon-intron boundaries of a gene. Highly effective antisense constructs include regions complementary to intron/exon splice junctions.
  • a prefened embodiment includes an antisense construct with complementarity to regions within 50-200 bases of an intron-exon splice junction. It has been observed that some exon sequences can be included in the construct without seriously affecting the target selectivity thereof. The amount of exonic material included will vary depending on the particular exon and intron sequences used. One can readily test whether too much exon DNA is included simply by testing the constructs in vitro to determine whether normal cellular function is affected or whether the expression of related genes having complementary sequences is affected. For purposes of making antisense oligonucleotides, polynucleotide sequences that are substantially complementary over their entire length and have zero or very few base mismatches are prefened.
  • sequences of fifteen bases in length preferably have complementary nucleotides at thirteen or fourteen or fifteen positions.
  • sequences which are completely complementary will be sequences which are entirely complementary throughout their entire length and have no base mismatches.
  • Other sequences with lower degrees of homology also are contemplated.
  • an antisense construct which has limited regions of high homology, but also contains anon-homologous region (e.g., ribozymes) could be designed. These molecules, though having less than 50% homology, would bind to target sequences under appropriate conditions.
  • Antisense nucleic acids are introduced into cells (e.g., by a viral vector or colloidal dispersion system such as a liposome).
  • Genbank Database is accessible on the world wide web at ncbi.nlm.nih.gov.
  • Related Prox-1 protein and/or nucleic acid sequences from other sources may be identified using probes directed at these sequences. Such additional sequences may be useful in certain aspects of the present invention.
  • antisense sequences may be full length genomic or cDNA copies, they also may be shorter fragments or oligonucleotides e.g., polynucleotides of 100 or less bases. Although shorter oligomers (8-20) are easier to make and more easily permeable in vivo, other factors also are involved in determining the specificity of base pairing. For example, the binding affinity and sequence specificity of an oligonucleotide to its complementary target increases with increasing length. It is contemplated that oligonucleotides of 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, or more base pairs will be used.
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • the cleavage event renders the mRNA unstable and prevents protein expression.
  • the mechanism of ribozyme action involves sequence specific interaction of the ribozyme molecule to complementary target RNA, followed by an endonucleolytic cleavage.
  • engineered hammerhead for which the substrate sequence requirements are minimal, or other motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of RNA sequences encoding protein complex components. Design of the hammerhead ribozyme and the therapeutic uses of ribozymes are disclosed in Usman et al., Cunent Opin.
  • Ribozymes can also be prepared and used as described in Long et al., FASEB J. (1993) 7:25; Symons, Ann. Rev. Biochem. (1992) 61 :641 ; Pe ⁇ otta et al., Biochem. (1992) 31:16-17; Ojwang et al., Proc. Natl. Acad. Sci. (USA) (1992) 89:10802-10806; and U.S. Pat. No. 5,254,678. Methods of cleaving RNA using ribozymes is described in U.S. Pat. No.
  • RNA target Specific ribozyme cleavage sites within any potential * RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites which include the following sequences, GUA, GUU, and GUC. Once identified, short RNA sequences of between 15 and 20 ribonucleotides conesponding to the region of the target gene containing the cleavage site may be evaluated for predicted structural features, such as secondary structure, that may render the oligonucleotide sequence unsuitable. The suitability of candidate targets may also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using ribonuclease protection assays (Draper PCT WO 93/23569; and U.S. Pat. No.
  • ribozymes can be designed to specifically bind and cut the corresponding mRNA species. Ribozymes, . therefore, provide a means to inhibit the expression Prox-1.
  • endogenous gene expression can be reduced by inactivating or "knocking out" the gene or its promoter using targeted homologous recombination. (E.g., see Smithies et al., 1985, Nature 317:230-234; Thomas & Capecchi, 1987, Cell 51:503-512; Thompson et ab, 1989 Cell 5:313-321).
  • a mutant, non-functional gene flanked by DNA homologous to the endogenous gene (either the coding regions or regulatory regions of the gene) can be used to transfect cells that express that gene in vivo. Insertion of the DNA construct, via targeted homologous recombination, results in inactivation of the gene. Gene expression can also be reduced by targeting deoxyribonucleoti.de sequences complementary to the regulatory region of the target gene (i.e., the gene promoter and/or enhancers) to form triple helical structures that prevent transcription of the gene in target cells in the body. (See generally, Helene, C.
  • Nucleic acid molecules used in triple helix formation for the inhibition of transcription are generally single stranded deoxyribonucleotides.
  • the base composition must be designed to promote triple helix formation via Hoogsteen base pairing rules, which generally require sizeable stretches of either purines or pyrimidines to be present on one strand of a duplex.
  • Nucleotide sequences may be pyrimidine-based, which will result in TAT and CGC+ triplets across the three associated strands of the resulting triple helix.
  • the pyrimidine-rich molecules provide base complementarity to a purine-rich region of a single strand of the duplex in a parallel orientation to that strand.
  • nucleic acid molecules may be chosen that are purine-rich, for example, containing a stretch of G residues. These molecules will form a triple helix with a DNA duplex that is rich in GC pairs, in which the majority of the purine residues are located on a single strand of the targeted duplex, resulting in GGC triplets across the three strands in the triplex.
  • the potential sequences that can be targeted for triple helix formation may be increased by creating a so called “switchback" nucleic acid molecule.
  • Switchback molecules are synthesized in an alternating 5'-3', 3'-5' manner, such that they base pair with first one strand of a duplex and then the other, eliminating the necessity for a sizeable stretch of either purines or pyrimidines to be present on one strand of a duplex.
  • Another technique for inhibiting the expression of a gene involves the use of RNA for induction of RNA interference (RNAi), using double stranded (dsRNA) (Fire et al, Nature 391: 806-811.
  • RNAi RNA interference
  • dsRNA double stranded
  • RNAi small interfering RNA sequences
  • RNAi small interfering RNA sequences
  • RNAi small interfering RNA sequences
  • RNAi is the process by which dsRNA induces homology-dependent degradation of complimentary mRNA.
  • the presence of dsRNA in cells triggers the RNAi response though a mechanism that has yet to be fully characterized.
  • a synthetic antisense nucleic acid molecule is hybridized by complementary base pairing with a "sense" ribonucleic acid to form a double stranded RNA.
  • the presence of long dsRNAs in cells stimulates the activity of a ribonuclease III enzyme.
  • the dsRNA antisense and sense nucleic acid molecules are provided that conespond to at least about 20, 25, 50, 100, 250 or 500 nucleotides or an entire Prox-1 coding strand, or to only a portion thereof.
  • the siRNAs are 30 nucleotides or less in length, and more preferably 21- to 23-nucleotides, with characteristic 2- to 3- nucleotide 3 '-overhanging ends, which are generated by ribonuclease in cleavage from longer dsRNAs. (See e.g. Tuschl T. Nat Biotechnol. 20:446-48.
  • RNA molecules have been also shown to function as siRNAs (i.e., enter the RNAi pathway and specifically target mRNA for degradation in mammalian cells (Martinez et ab, Cell 110, 563-574, 2002). Cleavage of the target RNA takes place in the middle of the region complementary to the antisense strand of the siRNA duplex (Elbashir et ab, 2001, Genes Dev., 15, 188). Intracellular transcription of small RNA molecules can be achieved by cloning the siRNA templates into RNA polymerase in (Pol DI) transcription units, which normally encode the small nuclear RNA (snRNA) U6 or the human RNAse P RNA HI.
  • snRNA small nuclear RNA
  • siRNAs are expressed as stem-loop hairpin RNA structures that give rise to siRNAs after intracellular processing (Brummelkamp et al. Science 296:550-553. 2002, herein incorporated by reference).
  • a stem loop hairpin can be expressed within an umelated Pol IT transcribed mRNA transcript.
  • a stem-loop hairpin designed to contain the siRNA sequence also contains conserved microRNA sequences within the loop and stem regions, thus resembling a natural precursor mRNA structure. Subsequently, the precursor can be processed by the cellular RNAi components to yield mature, functional siRNA/miRNA.
  • RNAi has been studied in a variety of systems.
  • RNAi may be carried out by administering sense and antisense nucleic acids of the invention in the same solution without annealing prior to administration, and may even be performed by administering the nucleic acids in separate vehicles within a very close timeframe.
  • Genetic control can also be achieved through the design of novel transcription factors for modulating expression of the gene of interest in native cells and animals.
  • the Cys2-His2 zinc finger proteins which bind DNA via their zinc finger domains, have been shown to be amenable to structural changes that lead to the recognition of different target sequences.
  • These artificial zinc finger proteins recognize specific target sites with high affinity and low dissociation constants, and are able to act as gene switches to modulate gene expression.
  • a zinc finger protein consisting of 6 tandem repeats of zinc fingers would be expected to ensure specificity for a particular sequence (Segal et al., (1999) Proc Natl Acad Sci USA 96:2758-2763).
  • the artificial zinc finger repeats designed based on target sequences, are fused to activation or repression domains to promote or suppress gene expression (Liu et ab, (1997) Proc Natl Acad Sci USA 94:5525-30).
  • the zinc finger domains can be fused to the TATA box-binding factor (TBP) with varying lengths of linker region between the zinc finger peptide and the TBP to create either transcriptional activators or repressors (Kim et al., (1997) Proc Natl Acad Sci USA 94:3616-3620).
  • TBP TATA box-binding factor
  • Such proteins, and polynucleotides that encode them, have utility for modulating expression in vivo in both native cells, animals and humans.
  • the novel transcription factor can be delivered to the target cells by transfecting constructs that express the transcription factor (gene therapy), or by introducing the protein.
  • Engineered zinc finger proteins can also be designed to bind RNA sequences for use in therapeutics as alternatives to antisense or catalytic RNA methods (McColl et al, (1999) Proc Natl Acad Sci USA 96:9521-6; Wu et ab, (1995) Proc Natl Acad Sci USA 92:344-348).
  • Inactivation of Prox-1 function can also be accomplished using an overexpressed dominant negative form of Prox-1.
  • a "dominant negative protein” is a mutant form of a protein which has the property of inhibiting the function of the endogenous, wild type form of the protein which conesponds to the mutant protein.
  • dominant negative proteins have amino acid substitutions or are truncated forms of the wild type protein.
  • the mutation may be in a substrate-binding domain (or DNA binding domain), a catalytic domain, or a cellular localization domain.
  • a dominant negative form of Prox-1 may include a mutant truncated with respect to the DNA binding domain or transactivation domain.
  • Disruption of the DNA binding domain entails truncation of the protein to exclude amino acids 572-634 of SEQ ID NO. 3, based on homology to Prospero (Drosophila).
  • Disruption of the transactivation domain entails the deletion of amino acids 635-737.
  • dominant negatives may include truncated forms of Prox-1 lacking the last 60 amino acids or the first 575 amino acids.
  • the mutant polypeptide will be overproduced. Point mutations can be made that have such an effect.
  • fusion of different polypeptides of various lengths to the terminus of a protein can yield dominant negative mutants.
  • General strategies for making dominant negative mutants are described in Herskowitz, Nature (1987) 329:219-222.
  • Anti-sense RNA and DNA molecules, ribozymes, RNAi, triple helix polynucleotides, and novel transcription factors can be prepared by any method known in the art for the synthesis of DNA and RNA molecules.
  • RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding the antisense RNA molecule. Such DNA sequences may be incorporated into a wide variety of vectors which incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters.
  • RNA polymerase promoters such as the T7 or SP6 polymerase promoters.
  • antisense cDNA constructs that synthesize antisense RNA constitutively or inducibly, depending on the promoter used, can be introduced stably or transiently into cells.
  • Gene Therapy As described n detail in the preceding section, a variety of genetic manipulations to achieve modulation of Prox-1 protein expression or activity are contemplated. Additionally, where administration of proteins is contemplated, such as zinc finger proteins targeted to Prox-1, administration of a gene therapy vector to cause the protein of interest to be produced in vivo also is contemplated. Where inhibition of proteins is contemplated (e.g., through use of antibodies or small molecule inhibitors), inhibition of protein expression in vivo by genetic techniques, such as knock-out techniques or anti-sense therapy, is contemplated. It is now widely recognized that DNA may be introduced into a cell using a variety of viral vectors.
  • Exemplary vectors that have been described in the literature include replication-deficient retroviral vectors, including but not limited to lentivirus vectors (Kim et al., J. Virol., 72(1): 811-816 (1998); Kingsman & Johnson, Scrip Magazine, October, 1998, pp. 43-46.); adenoviral (see, for example, U.S. Patent No. 5,824,544; U.S. Patent No. 5,707,618; U.S. Patent No. 5,792,453; U.S. Patent No. 5,693,509; U.S. Patent No. 5,670,488; U.S. Patent No. 5,585,362; Quantin et al., Proc. Natl. Acad. Sci.
  • prefened polynucleotides include a suitable promoter and polyadenylation sequence to promote expression in the target tissue of interest.
  • suitable promoters/enhancers for mammalian cell expression include, e.g., cytomegalovirus promoter/enhancer (Lehner et al., J. Clin. Microbiob, 29:2494-2502 (1991); Boshart et al., Cell, 41:521-530 (1985)); Rous sarcoma virus promoter (Davis et al., Hum.
  • simian virus 40 promoter long terminal repeat (LTR) of retroviruses, keratin 14 promoter, and myosin heavy chain promoter.
  • LTR long terminal repeat
  • retroviruses keratin 14 promoter
  • myosin heavy chain promoter myosin heavy chain promoter.
  • non-viral dehvery is contemplated. These include calcium phosphate precipitation (Graham and Van Der Eb, Virology, 52:456- 467 (1973); Chen and Okayama, Mol. Cell Biol, 7:2745-2752, (1987); Rippe, et al, Mol. Cell Biol, 10:689-695 (1990)), DEAE-dextran (Gopal, Mol.
  • the expression construct (or indeed the peptides discussed above) may be entrapped in a liposome.
  • Liposomes are vesicular structures characterized by a phospholipid bilayer membrane and an inner aqueous medium. Multilamellar liposomes have multiple lipid layers separated by aqueous medium. They form spontaneously when phospholipids are suspended in an excess of aqueous solution.
  • the lipid components undergo self-reanangement before the formation of closed structures and entrap water and dissolved solutes between the lipid bilayers (Ghosh and Bachhawat, "In Liver Diseases, Targeted Diagnosis And Therapy Using Specific Receptors And Ligands," Wu, G., Wu, C, ed., New York: Marcel Dekker, pp. 87-104 (1991)).
  • the addition of DNA to cationic liposomes causes a topological transition from liposomes to optically birefringent Uquid-crystalline condensed globules (Radler, et al, Science, 275(5301):810-4, (1997)).
  • DNA-lipid complexes are potential non-viral vectors for use in gene therapy and delivery. Liposome-mediated nucleic acid delivery and expression of foreign DNA in vitro has been very successful. Also contemplated in the present invention are various commercial approaches involving "lipofection" technology.
  • the liposome may be complexed with a hemagglutinating virus (HVJ). This has been shown to facilitate fusion with the cell membrane and promote cell entry of liposome-encapsulated DNA (Kaneda, et al, Science, 243:375-378 (1989)).
  • HVJ hemagglutinating virus
  • the liposome may be complexed or employed in conjunction with nuclear nonhistone chromosomal proteins (HMG-1) (Kato, et al, J. Biol. Chem., 266:3361-3364 (1991)).
  • HMG-1 nuclear nonhistone chromosomal proteins
  • the liposome may be complexed or employed in conjunction with both HVJ and HMG-1.
  • expression constructs have been successfully employed in transfer and expression of nucleic acid in vitro and in vivo, then they are applicable for the present invention.
  • Other vector delivery systems that can be employed to deliver a nucleic acid encoding a therapeutic gene into cells include receptor-mediated delivery vehicles.
  • Receptor-mediated gene targeting vehicles generally consist of two components: a cell receptor-specific ligand and a DNA-binding agent.
  • ligands have been used for receptor-mediated gene transfer. The most extensively characterized ligands are asialoorosomucoid (ASOR) (Wu and Wu (1987), supra) and transferrin (Wagner, et al, Proc. Natl Acad Sci. USA, 87(9):3410-3414 (1990)).
  • the dehvery vehicle may comprise a ligand and a liposome.
  • a nucleic acid encoding a therapeutic gene also may be specifically delivered into a particular cell type by any number of receptor-ligand systems with or without liposomes.
  • the expression construct may simply consist of naked recombinant DNA or plasmids. Transfer of the construct may be performed by any of the methods mentioned above that physically or chemically permeabilize the cell membrane.
  • the tumor may be directly injected with the expression vector or protein.
  • a tumor bed may be treated prior to, during or after resection. Following resection, one generally will deliver the vector by a catheter left in place following surgery.
  • One may utilize the tumor vasculature to introduce the vector into the tumor by injecting a supporting vein or artery.
  • a more distal blood supply route also may be utilized.
  • cells from the patient are removed and maintained outside the body for at least some period of time. During this period, a therapy is delivered, after which the cells are reintroduced into the patient; preferably, any tumor cells in the sample have been killed.
  • the present invention contemplates an antibody that is immunoreactive with a Prox-1 protein molecule of the present invention, or any portion thereof.
  • Such antibodies include, but are not limited to, polyclonal, monoclonal, chimeric, single chain, Fab fragments and fragments produced by a Fab expression library, bifunctional/bispecific antibodies, humanized antibodies, CDR grafted antibodies, human antibodies and antibodies which include portions of CDR sequences specific for Prox-1 protein.
  • the antibodies are useful as diagnostic reagents for measuring Prox-1 expression in a biological sample (e.g., a biopsy of colon tissue), and are useful for binding to Prox-1 protein to inhibit Prox-1 activity where the antibodies are delivered into cells.
  • Neutralizing antibodies i.e., those which may suppress Prox-1 expression, are especially prefened for therapeutic embodiments.
  • an antibody is a monoclonal antibody.
  • the invention provides for a pharmaceutical composition comprising a therapeutically effective amount of an antibody directed against Prox-1 protein.
  • the antibody may bind to and neutralize the apoptotic effects of the Prox-1 protein.
  • the antibody may be formulated with a pharmaceutically acceptable adjuvant.
  • Means for preparing and characterizing antibodies are well known in the art (see, e.g., Harlow and Lane, ANTIBODIES: A LABORATORY MANUAL, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1988).
  • a polyclonal antibody is prepared by immunizing an animal with an immunogen comprising a polypeptide of the present invention and collecting antisera from that immunized animal.
  • an immunogen comprising a polypeptide of the present invention
  • a wide range of animal species can be used for the production of antisera.
  • an animal used for production of anti-antisera is a non-human animal including rabbits, mice, rats, hamsters, goat, sheep, pigs or horses. Because of the relatively large blood volume of rabbits, a rabbit is a prefened choice for production of polyclonal antibodies.
  • various adjuvants may be used to increase immunological response.
  • adjuvants include but are not limited to Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol.
  • BCG Bacilli Calmette-Guerin
  • Corynebacterium parvum are potentially useful human adjuvants.
  • Antibodies, both polyclonal and monoclonal, specific for isoforms of antigen may be prepared using conventional immunization techniques, as will be generally known to those of skill in the art.
  • the term "specific for” is intended to mean that the variable regions of the antibodies recognize and bind Prox-1 protein and are capable of distinguishing Prox-1 protein from other antigens, for example other secreted proapoptotic factors.
  • a composition containing antigenic epitopes of the compounds of the present invention can be used to immunize one or more experimental animals, such as a rabbit or mouse, which will then proceed to produce specific antibodies against the compounds of the present invention.
  • Polyclonal antisera may be obtained, after allowing time for antibody generation, simply by bleeding the animal and preparing serum samples from the whole blood.
  • Monoclonal antibodies to Prox-1 protein may be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture.
  • Such cell lines suited for use in hybridoma-producing fusion procedures preferably are non-antibody-producing, have high fusion efficiency, and enzyme deficiencies that render them incapable of growing in certain selective media which support the growth of only the desired fused cells (hybridomas).
  • the immunized animal is a mouse
  • rats one may use R210.RCY3, Y3-Ag 1.2.3, IR983F and 4B210
  • U-266, GM1500-GRG2, LICR-LON-HMy2 and UC729-6 are all useful in connection with cell fusions.
  • hybridomas and cell lines produced by such techniques for producing the monoclonal antibodies are contemplated to be novel compositions of the present invention.
  • An exemplary method for producing monoclonal antibodies against Prox-1 is provided in Example 1. Those of skill in the art will appreciate that such a method may be modified using techniques well known to those of skill in the art and still produce antibodies within the scope of the present invention.
  • Antibodies may also be produced by inducing in vivo production in the lymphocyte population or by screening recombinant immunoglobulin libraries or panels of highly specific binding reagents as disclosed in Orlandi et al (Proc Natl Acad Sci 86: 3833-3837; 1989), and Winter G and Milstein C (Nature 349: 293-299, 1991).
  • Fully human antibodies relate to antibody molecules in which essentially the entire sequences of both the light chain and the heavy chain, including the CDRs, arise from human genes. Such antibodies are termed "human antibodies/' or "fully human antibodies” herein.
  • Human monoclonal antibodies can be prepared by the trioma technique; the human B-cell hybridoma technique (see Kozbor, et al, Immunol Today 4: 72 (1983)) and the EBV hybridoma technique to produce human monoclonal antibodies (see Cole, et al, 1985 In: MONOCLONAL ANTIBODIES AND
  • Human monoclonal antibodies may be utilized in the practice of the present invention and may be produced by using human hybridomas (see Cote, et al., Proc Natl Acad Sci USA 80: 2026-2030 (1983)) or by transforming human B-cells with Epstein Ban Virus in vitro (see Cole, et al., 1985 In: MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96).
  • human antibodies can also be produced using additional techniques, including phage display libraries (Hoogenboom and Winter, J. Mol.
  • human antibodies can be made by introducing human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, which closely resembles that seen in humans in all respects, including gene rea ⁇ angement, assembly, and antibody repertoire. This approach is described, for example, in U.S. Patent Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; 5,661,016, and in Marks et al.
  • Human antibodies may additionally be produced using transgenic nonhuman animals which are modified so as to produce fully human antibodies rather than the animal's endogenous antibodies in response to challenge by an antigen. (See PCT publication WO94/02602).
  • the endogenous genes encoding the heavy and tight immunoglobulin chains in the nonhuman host have been incapacitated, and active loci encoding human heavy and tight chain immunoglobuhns are inserted into the host's genome.
  • the human genes are incorporated, for example, using yeast artificial chromosomes containing the requisite human DNA segments.
  • An animal which provides all the desired modifications is then obtained as progeny by crossbreeding intermediate transgenic animals containing fewer than the full complement of the modifications.
  • the prefened embodiment of such a nonhuman animal is a mouse, and is termed the XenomouseTM as disclosed in PCT publications WO 96/33735 and WO 96/34096. This animal produces B cells which secrete fully human immunoglobuhns.
  • the antibodies can be obtained directly from the animal after immunization with an immunogen of interest, as, for example, a preparation of a polyclonal antibody, or alternatively from immortalized B cells derived from the animal, such as hybridomas producing monoclonal antibodies. Additionally, the genes encoding the immunoglobuhns with human variable regions can be recovered and expressed to obtain the antibodies directly, or can be further modified to obtain analogs of antibodies such as, for example, single chain Fv molecules.
  • An example of a method of producing a nonhuman host, exemplified as a mouse, lacking expression of an endogenous immunoglobulin heavy chain is disclosed in U.S. Patent No. 5,939,598.
  • Antibodies as described herein are useful in standard immunochemical procedures, such as ELISA, radioimmuno assays, and Western blot methods and in immunohistochemical procedures such as tissue staining, as well as in other procedures which may utihze antibodies specific to Prox-1 protein -related antigen epitopes. Additionally, it is proposed that monoclonal antibodies specific to the particular Prox-1 protein of different species may be utilized in other useful applications. In general, both polyclonal and monoclonal antibodies against Prox-1 protein may be used in a variety of embodiments. In certain aspects, the antibodies may be employed for therapeutic purposes in which the inhibition of Prox-1 protein activity is desired (e.g., to reduce apoptosis in neuronal cells).
  • Antibodies may be used to block Prox-1 protein action. Antibodies of the present invention also may prove useful in diagnostic purposes in order, for example, to detect increases or decreases in Prox-1 protein in tissue samples including samples for sites of inflammation, or fluid samples including blood serum, plasma and exudate samples. Additional aspects will employ the antibodies of the present invention in antibody cloning protocols to obtain cDNAs or genes encoding other Prox-1 protein. They may also be used in inhibition studies to analyze the effects of Prox-1 related peptides in cells or animals. Anti- Prox-1 protein antibodies will also be useful in immunolocalization studies to analyze the distribution of Prox-1 protein during various cellular events, for example, to determine the cellular or tissue-specific distribution of Prox-1 protein polypeptides under different points in the cell cycle. A particularly useful application of such antibodies is in purifying native or recombinant Prox-1 protein, for example, using an antibody affinity column. The operation of all such immunological techniques will be known to those of skill in the art in light of the present disclosure.
  • molecules that are modulators i.e., agonists or antagonists, of the activity of Prox-1 protein.
  • Natural or synthetic molecules that modulate Prox-1 protein may be identified using one or more screening assays, such as those described herein. Such molecules may be administered either in an ex vivo manner, or in an in vivo manner by injection, or by oral delivery, implantation device or the like.
  • Test molecule(s) refers to the molecule(s) that is/are under evaluation for the ability to modulate (i.e., increase or decrease) the activity of Prox-1 protein.
  • a molecule that modulates Prox-1 activity will interact directly with Prox-1. However, it is also contemplated that a molecule may also modulate Prox-1 protein activity indirectly, such as by affecting Prox-1 gene expression, or by binding to a Prox-1 binding partner.
  • a test molecule will bind to a Prox-1 protein with an affinity constant of at least about 10 "6 M, preferably about 10 "8 M, more preferably about 10 "9 M, and even more preferably about 10 "10 M. Methods for identifying compounds which interact with Prox-1 protein are encompassed by the present invention.
  • a Prox-1 protein is incubated with a test molecule under conditions which permit the interaction of the test molecule with a Prox-1 protein, and the extent of the interaction can be measured.
  • the test molecule(s) can be screened in a substantially purified form or in a crude mixture.
  • a Prox-1 protein agonist or antagonist may be a protein, peptide, carbohydrate, lipid or small molecular weight molecule which interacts with Prox-1 to regulate its activity.
  • Molecules which regulate Prox-1 expression include nucleic acids which are complementary to nucleic acids encoding a Prox-1 protein, or are complementary to nucleic acid sequences which direct or control the expression of Prox-1 protein, and which act as anti-sense regulators of expression. Once a set of test molecules has been identified as interacting with Prox-1 protein, the molecules may be further evaluated for their abihty to increase or decrease Prox-1 activity. The measurement of the interaction of test molecules with Prox-1 may be carried out in several formats, including solution-phase assays and immunoassays. In general, test molecules are incubated with Prox-1 for a specified period of time, and Prox-1 protein activity is determined by one or more assays for measuring biological activity.
  • Prox-1 displays biological activity through an interaction with a binding partner
  • a variety of in vitro assays may be used to measure the binding of Prox-1 to the conesponding binding partner. These assays may be used to screen test molecules for their ability to increase or decrease the rate and/or the extent of binding of Prox-1 to its binding partner.
  • a Prox-1 polypeptide is immobilized in the wells of a microtiter plate. Radiolabeled Prox-1 binding partner and the test molecule(s) can then be added either one at a time (in either order) or simultaneously to the wells.
  • the wells can be washed and counted (using a scintillation counter) for radioactivity to determine the extent to which the binding partner bound to Prox-1 polypeptide.
  • the , molecules will be tested over a range of concentrations, and a series of control wells lacking one or more elements of the test assays can be used for accuracy in the evaluation of the results.
  • An alternative to this method involves reversing the "positions" of the proteins, i.e., immobilizing Prox-1 binding partner to the micrbfiter plate wells, incubating with the test molecule and radiolabeled Prox-1 polypeptide, and determining the extent of Prox-1 polypeptide binding.
  • Prox-1 protein or its binding partner may be conjugated to biotin and the presence of biotinylated protein can then be detected using streptavidin linked to an enzyme, such as horseradish peroxidase (HRP) or alkaline phosphatase (AP), that can be detected colorometrically or by fluorescent tagging of streptavidin.
  • HRP horseradish peroxidase
  • AP alkaline phosphatase
  • An antibody directed to Prox-1 or to a Prox-1 binding partner and conjugated to biotin may also be used and can be detected after incubation with enzyme-linked streptavidin linked to AP or HRP.
  • a Prox-1 protein or Prox-1 binding partner can also be immobilized by attachment to agarose beads, acrylic beads or other types of such inert solid phase substrates.
  • the substrate-protein complex can be placed in a solution containing the complementary protein and the test compound. After incubation the beads can be precipitated by centrifugation, and the amount of binding between Prox-1 protein and its binding partner can be assessed using the methods described herein.
  • the substrate-protein complex can be immobilized in a column, and the test molecule and complementary protein are passed through the column. The formation of a complex between an Prox-1 protein and its binding partner can then be assessed using any of the techniques set forth herein, i.e., radiolabeling, antibody binding or the like.
  • a surface plasmon resonance detector system such as the BIAcore assay system (Pharmacia, Piscataway, NJ).
  • the BIAcore system may be carried out using the manufacturer's protocol.
  • This assay essentially involves the covalent binding of either Prox-1 or a Prox-1 binding partner to a dextran-coated sensor chip which is located in a detector. The test compound and the other complementary protein can then be injected, either simultaneously or sequentially, into the chamber containing the sensor chip.
  • the amount of complementary protein that binds can be assessed based on the change in molecular mass which is physically associated with the dextran-coated side of the sensor chip; the change in molecular mass can be measured by the detector system.
  • the assays set forth herein can be readily modified by adding such additional test compound(s) either simultaneous with, or subsequent to, the first test compound. The remainder of the steps in the assay are as set forth herein.
  • In vitro assays such as those described herein may be used advantageously to screen large numbers of compounds for effects on complex formation by Prox-1 polypeptide and a Prox-1 binding partner.
  • the assays may be automated to screen compounds generated in phage display, synthetic peptide, and chemical synthesis libraries.
  • Compounds which increase or decrease the formation of a complex between a Prox-1 polypeptide and a Prox-1 binding partner may also be screened in cell culture using cells and cell lines expressing either Prox-1 polypeptide or a Prox-1 binding partner. Cells and cell lines may be obtained from any mammal.
  • the binding of a Prox-1 protein to cells expressing a Prox-1 binding partner at the surface is evaluated in the presence or absence of test molecules, and the extent of binding may be determined by, for example, flow cytometry using a biotinylated antibody to a Prox-1 binding partner.
  • Cell culture assays can be used advantageously to further evaluate compounds that score positive in protein binding assays described herein. Cell cultures can also be used to screen the impact of a drug candidate.
  • drug candidates may decrease or increase the expression of the Prox-1 like gene.
  • the amount of Prox-1 protein that is produced may be measured after exposure of the cell culture to the drug candidate.
  • the overexpression of a particular gene may have a particular impact on the cell culture.
  • the production of a particular metabolic product such as a fragment of a polypeptide may result in, or be associated with, a disease or pathological condition. In such cases, one may test a drug candidate's ability to decrease the production of such a metabolic product in a cell culture.
  • the tat protein sequence (from HIV) can be used to internalize proteins into a cell. See e.g., Falwell et al, Proc. Natl. Acad. Sci. USA, 91:664-668 (1994).
  • an 11 amino acid sequence (YGRKKRRQRRR; SEQ ID NO: 46) of the HJN tat protein (termed the "protein transduction domain", or TAT PDT) has been described as mediating delivery across the cytoplasmic membrane and the nuclear membrane of a cell.
  • TAT PDT protein transduction domain
  • FITC- constructs are prepared which bind to cells as observed by fluorescence-activated cell sorting (FACS) analysis, and these constructs penetrate tissues after i.p. adminstration.- Next, tat-bgal fusion proteins are constructed. Cells treated with this construct demonstrate b-gal activity. Following injection, a number of tissues, including liver, kidney, lung, heart and brain tissue, have been found to demonstrate expression using these procedures. It is believed that these constructions underwent some degree of unfolding in order to enter the cell; as such, refolding may be required after entering the cell. It will thus be appreciated that the tat protein sequence may be used to internalize a desired protein or polypeptide into a cell.
  • FACS fluorescence-activated cell sorting
  • Prox-1 antagonist such as an anti-Prox-1 binding agent, small molecule, or antisense oligonucleotide
  • Prox-1 antagonist can be administered intracellularly to inhibit the activity of a Prox-1 molecule. See also, Strauss, E., Science, 285:1466-1467 (1999).
  • Rational Drug Design The goal of rational drug design is to produce structural analogs of biologically active polypeptides or compounds with which they interact (agonists, antagonists, inhibitors, peptidomimetics, binding partners, etc.). By creating such analogs, it is possible to fashion drugs which are more active or stable than the natural molecules, which have different susceptibility to alteration or which may affect the function of various other molecules.
  • An alternative approach, "alanine scan” involves the random replacement of residues throughout molecule with alanine, and the resulting affect on function determined.
  • Anti-idiotypes may be generated using the methods described herein for producing antibodies, using an antibody as the antigen.
  • compositions can be administered in therapeutically effective dosages alone or in combination with adjuvant cancer therapy such as surgery, chemotherapy, radiotherapy, thermotherapy, and laser therapy, and may provide a beneficial effect, e.g. reducing tumor size, slowing rate of tumor growth, inhibiting metastasis, or otherwise improving overall clinical condition, without necessarily eradicating the cancer.
  • the composition can also be administered in therapeutically effective amounts as a portion of an anti-cancer cocktail.
  • An anti-cancer cocktail is a mixture of the polypeptide or modulator of the invention with one or more anti-cancer drugs in addition to a pharmaceutically acceptable carrier for delivery.
  • any anti-cancer drugs can be used as a treatment in combination with the polypeptide or modulator of the invention, including: Actinomycin D, Antinoglutethimide, Asparaginase, Bleomycin, Busulfan, Carboplatin, Carmustine, Chlorambucib Cisplatin (cis-DDP), Cyclophosphamide, Cytarabine HCl (Cytosine arabinoside), dacarbazine, Dactinomycin, Daunorubicin HCl, Doxorubicin HCl, Estramustine phosphate sodium, Etoposide (V16-213), Floxuridine, 5-Fluorouracil (5-Fu), Flutamide, Hydroxyurea (hydroxycarbamide), lfosfamide, Interferon Alpha-2a, Interferon Alpha-2b,
  • Leuprolide acetate (LHRH-releasing factor analog), Lomustine, Mechlorethamine HCl (nitrogen mustard), Melphalan, Mercaptopurine, Mesna, Methotrexate (MTX), Mitomycin, Mitoxantrone HCb Octreotide, Plicamycin, Procarbazine HCl, Streptozocin, Tamoxifen citrate, Thioguanine, Thiotepa, Vinblastine sulfate, Vincristine sulfate, Amsacrine, Azacitidine, Hexamethylmelamine, Interleukin-2, Mitoguazone, Pentostatin, Semustine, Teniposide, and Vindesine sulfate.
  • compositions of the invention may be used for prophylactic treatment of cancer.
  • hereditary conditions and/or environmental situations e.g. exposure to carcinogens
  • In vitro and in vivo models can be used to determine the effective doses of the compositions of the invention for cancer treatment.
  • Suitable tumor cells lines are available, e.g. from American Type Tissue Culture Collection catalogs, and/or are described below.
  • compositions Purified nucleic acids, antisense molecules, purified protein, antibodies, antagonists, or inhibitors may all be used as pharmaceutical compositions. Delivery of specific molecules for therapeutic purposes in this invention is further described below.
  • the active compositions of the present invention include classic pharmaceutical preparations. Administration of these compositions according to the present invention will be via any common route so long as the target tissue is available via that route.
  • compositions may be introduced into the subject by any conventional method, e.g., by intravenous, intradermab intramusclar, intramammary, intraperitoneab intrathecal, intraocular, retrobulbar, intrapulmonary (e.g., term release); by oral, sublingual, nasal, anal, vaginal, or transdermal delivery, or by surgical implantation at a particular site, e.g., embedded under the splenic capsule, brain, or in the cornea.
  • the treatment may consist of a single dose or a plurality of doses over a period of time.
  • the active compounds may be prepared for administration as solutions of free base or pharmacologically acceptable salts in water suitably mixed with a surfactant, such as hydroxypropylcellulose.
  • Dispersions also can be prepared in glycerob liquid polyethylene glycols, and 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. 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, ethanob polyol (for example, glycerob propylene glycob and liquid polyethylene glycob and the like), suitable mixtures thereof, and vegetable oils.
  • ethanob polyol for example, glycerob propylene glycob and liquid polyethylene glycob and the like
  • suitable mixtures thereof and vegetable oils.
  • the proper fluidity can 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.
  • 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.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the prefened methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile- filtered solution thereof.
  • pharmaceutically acceptable ca ⁇ ier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents 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 also can be incorporated into the compositions.
  • the active compositions may be incorporated with excipients and used in the form of non-ingestible mouthwashes and dentifrices.
  • a mouthwash may be prepared incorporating the active ingredient in the required amount in an appropriate solvent, such as a sodium borate solution (Dobell's Solution).
  • the active ingredient may be incorporated into an antiseptic wash containing sodium borate, glycerin and potassium bicarbonate.
  • the active ingredient may also be dispersed in dentifrices, including: gels, pastes, powders and slurries.
  • the active ingredient may be added in a therapeutically effective amount to a paste dentifrice that may include water, binders, abrasives, flavoring agents, foaming agents, and humectants.
  • compositions of the present invention may be formulated in a neutral or salt form.
  • Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups also can be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • the compositions of the present invention may be formulated in a neutral or salt form.
  • Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandehc, and the like. Salts formed with the free carboxyl groups also can be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandehc, and the like.
  • Salts formed with the free carboxyl groups also can be derived from inorganic bases such as, for example, sodium,
  • an "effective amount” is an amount sufficient to effect beneficial or desired clinical results.
  • An effective amount can be administered in one or more doses.
  • an "effective amount" of polynucleotide, and/or polypeptide is an amount that results in amelioration of symptoms or a prolongation of survival in a patient.
  • the effective amount is generally determined by the physician on a case-by-case basis and is within the skill of one in the art. Several factors are typically taken into account when determining, an appropriate dosage. These factors include age, sex and weight of the patient, the condition being treated, the severity of the condition and the form of the antibody being administered. For instance, in embodiments in which the antibody compositions of the present invention are being therapeutically administered, it is likely the concentration of a single chain antibody need not be as high as that of native antibodies in order to be therapeutically effective. Determination of the effective amounts is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture (i.e., the concentration of the test compound which achieves a half-maximal inhibition of the C-proteinase activity).
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD50 and ED50.
  • Compounds which exhibit high therapeutic indices are prefened.
  • the data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in human.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. See, e.g., Fingl et al., 1975, in "The Pharmacological Basis of Therapeutics," Ch. 1 p.LDosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the C-proteinase inhibiting effects, of ntinimal effective concentration (MEC).
  • MEC ntinimal effective concentration
  • the MEC will vary for each compound but can be estimated from in vitro data; for example, the concentration necessary to achieve 50-90% inhibition of the C-proteinase using the assays described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations. Dosage intervals can also be determined using MEC value. Compounds should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%. In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration.
  • the present invention is directed at treatment of colon cancer, including colon cancer indicated by the presence of overexpression of Prox-1.
  • routes of administration are contemplated.
  • the discrete tumor mass may be injected.
  • the injections may be single or multiple; where multiple, injections are made at about 1 cm spacings across the accessible surface of the tumor.
  • the subject to be treated may be a mammal, preferably human or other animal.
  • subjects include for example, farm animals including cows, sheep, pigs, horses and goats, companion animals such as dogs and cats, exotic and/or zoo animals, laboratory animals including mice rats, rabbits, guinea pigs and hamsters; and poultry such as chickens, turkey ducks and geese.
  • compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may, for example, comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • transgenic Animals A transgenic animal can be prepared in a number of ways.
  • a transgenic organism is one that has an extra or exogenous fragment of DNA incorporated into its genome, sometimes replacing an endogenous piece of DNA.
  • the integration event In order to achieve stable inheritance of the extra or exogenous DNA, the integration event must occur in a cell type that can give rise to functional germ cells.
  • the two animal cell types that are used for generating transgenic animals are fertilized egg cells and embryonic stem cells.
  • Embryonic stem (ES) cells can be returned from in vitro culture to a "host" embryo where they become incorporated into the developing animal and can give rise to transgenic cells in all tissues, including germ cells. The ES cells are transfected in culture and then the mutation is transmitted into the germline by injecting the cells into an embryo.
  • blastocysts are isolated from pregnant mice at a given stage in development, for example, the blastocyst from mice may be isolated at day 4 of development (where day 1 is defined as the day of plug), into an appropriate buffer that will sustain the ES cells in an undifferentiated, pluripotent state.
  • ES cell tines may be isolated by a number of methods well known to those of skill in the art.
  • the blastocysts may be allowed to attach to the culture dish and approximately 7 days later, the outgrowing inner cell mass picked, trypsinized and transfe ⁇ ed to another culture dish in the same culture media. ES cell colonies appear 2-3 weeks later with between 5-7 individual colonies arising from each explanted inner cell mass.
  • ES cell lines can then be expanded for further analysis.
  • ES cell lines can be isolated using the immunosurgery technique (described in Martin, Proc. Natl. Acad. Sci. USA 78:7634-7638, 1981) where the trophectoderm cells are destroyed using anti-mouse antibodies prior to explanting the inner cell mass.
  • the ES cell lines that have been manipulated by homologous recombination are reintroduced into the embryonic envkonment by blastocyst injection (as described in Wilhams et al, Cell 52: 121-131, 1988). Briefly, blastocysts are isolated from a pregnant mouse and expanded.
  • the expanded blastocysts are maintained in oil-drop cultures at 4°C for 10 minutes prior to culture.
  • the ES cells are prepared by picking individual colonies, which are then incubated in phosphate-buffered saline, 0.5 mM EGTA for 5 minutes; a single cell suspension is prepared by incubation in a trypsin-EDTA solution containing 1% (v/v) chick serum for a further 5 minutes at 4°C.
  • Five to twenty ES cells in Dulbecco's modified Eagle's Medium with 10% (v/v) fetal calf serum and 3,000 units/ml DNAase 1 buffered in 20 mM HEPES [pH 8]) are injected into each blastocyst.
  • the blastocysts are then transfe ⁇ ed into pseudo-pregnant recipients and allowed to develop normally.
  • the transgenic mice are identified by coat markers (Hogan et al. , Manipulating the Mouse Embryo, Cold Spring Harbor, N.Y. (1986)). Additional methods of isolating and propagating ES cells may be found in, for example, U.S. Patent No. 5,166,065; U.S. Patent No. 5,449,620; U.S. Patent No. 5,453,357; U.S. Patent No. 5,670,372; U.S. Patent No. 5,753,506; U.S. Patent No. 5,985,659, each incorporated herein by reference.
  • this method involves injecting DNA into a fertilized egg, or zygote, and then allowing the egg to develop in a pseudo-pregnant mother.
  • the zygote can be obtained using male and female animals of the same strain or from male and female animals of different strains.
  • the transgenic animal that is born, the founder, is bred to produce more animals with the same DNA insertion.
  • the new DNA typically randomly integrates into the genome by a non-homologous recombination event. One to many thousands of copies of the DNA may integrate at a site in the genome Generally, the DNA is injected into one of the pronuclei, usually the larger male pronucleus.
  • DNA clones for microinjection can be prepared by any means known in the art. For example, DNA clones for microinjection can be cleaved with enzymes appropriate for removing the bacterial plasmid sequences, and the DNA fragments electrophoresed on 1% agarose gels in TBE buffer, using standard techniques. The DNA bands are visualized by staining with ethidium bromide, and the band containing the expression sequences is excised.
  • the excised band is then placed in dialysis bags containing 0.3 M sodium acetate, pH 7.0.
  • DNA is electroeluted into the dialysis bags, extracted with a 1:1 phen ⁇ bchloroform solution and precipitated by two volumes of ethanob
  • the DNA is redissolved in 1 ml of low salt buffer (0.2 M NaCb 20 mM Tris, pH 7.4, and 1 mM EDTA) and purified on an Elutip-DTM column.
  • the column is first primed with 3 ml of high salt buffer (1 M Nad, 20 mM Tris, pH 7.4, and 1 mM EDTA) followed by washing with 5 ml of low salt buffer.
  • the DNA solutions are passed through the column three times to bind DNA to the column matrix.
  • DNA is eluted with 0.4 ml high salt buffer and precipitated by two volumes of ethanob DNA concentrations are measured by absorption at 260 nm in a UV spectrophotometer.
  • DNA concentrations are adjusted to 3 mg/ml in 5 mM Tris, pH 7.4 and 0.1 mM EDTA. Additional methods for purification of DNA for microinjection are described in Hogan et al. Manipulating the Mouse Embryo (Cold Spring Harbor
  • the mated females are sacrificed and embryos are recovered from excised oviducts and placed in an appropriate buffer, e.g., Dulbecco's phosphate buffered saline with 0.5% bovine serum albumin (BSA; Sigma). Sunounding cumulus cells are removed with hyaluronidase (1 mg/ml). Pronuclear embryos are then washed and placed in Earle's balanced salt solution containing 0.5 % BSA in a 37.5°C incubator with a humidified atmosphere at 5% C0 2 , 95% ak until the time of injection. Embryos can be implanted at the two-cell stage. Randomly cycling adult female mice are paired with vasectomized males.
  • an appropriate buffer e.g., Dulbecco's phosphate buffered saline with 0.5% bovine serum albumin (BSA; Sigma). Sunounding cumulus cells are removed with hyaluronidase (1 mg/ml).
  • Recipient females are mated at the same time as donor females.
  • the recipient females are anesthetized with an intraperitoneal injection of 0.015 ml of 2.5 % avertin per gram of body weight.
  • the oviducts are exposed by a single midline dorsal incision. An incision is then made through the body wall dkectly over the oviduct. The ovarian bursa is then torn with watchmakers forceps.
  • Embryos to be transfe ⁇ ed are placed in DPBS (Dulbecco's phosphate buffered saline) and in the tip of a transfer pipette (about 10 to 12 embryos). The pipette tip is inserted into the infundibulum and the embryos transfe ⁇ ed. After the transfer, the incision is closed by two sutures. The pregnant animals then give birth to the founder animals which are used to establish the transgenic line.
  • DPBS Dulbecco's phosphate buffered saline
  • the pipette tip is inserted into the infundibulum and the embryos transfe ⁇ ed. After the transfer, the incision is closed by two sutures.
  • the pregnant animals then give birth to the founder animals which are used to establish the transgenic line.
  • prefened aspects of the present invention are dkected to methods of screening and diagnosing colon cancer in an individual.
  • diagnostic methods of the invention are practiced through the detection of the Prox-1 protein.
  • methods for detecting a polypeptide of the invention can comprise contacting a biological sample with a compound that binds to and forms a complex with the polypeptide for a period sufficient to form the complex, and detecting the complex, so that if a complex is detected, a polypeptide of the invention is detected.
  • Prox-1 protein detection can be accomplished using antibodies specific for the protein in any of a number of formats commonly used by those of skill in the art for such detection.
  • Such antibodies may be employed in ELISA-based techniques and Western blotting techniques to detect the presence of Prox-1 in a biological sample from a subject being tested.
  • Methods for setting up ELISA assays and preparing Western blots of a sample are well known to those of skill in the art.
  • the biological sample can be any tissue or fluid in which colon cells or tissue might be present.
  • An anti-Prox-1 antibody or fragment thereof also is useful to monitor expression of this protein in individuals suffering from colon cancer.
  • diagnostic assays entail detecting the formation of a complex resulting from the binding of an antibody or fragment thereof to Prox-1.
  • the antibodies or antigen-binding fragments can be labeled or unlabeled.
  • the antibodies or fragments can be directly labeled.
  • labels can be employed, including, but not limited to, radionuclides, fluorescers, enzymes, enzyme substrates, enzyme cofactors, enzyme inhibitors and ligands (e.g., biotin, haptens).
  • radionuclides e.g., radionuclides
  • fluorescers e.g., fluorescers
  • enzymes e.g., enzyme substrates, enzyme cofactors, enzyme inhibitors and ligands (e.g., biotin, haptens).
  • Numerous appropriate immunoassays are known to the skilled artisan (see, for example, U.S. Pat. Nos. 3,817,827; 3,850,752; 3,901,654 and 4,098,876).
  • the antibodies or fragments can be detected using suitable means, as in agglutination assays,
  • Unlabeled antibodies or fragments can also be used in combination with another (i.e., one or more) suitable reagent which can be used to detect antibody, such as a labeled antibody (e.g., a second antibody) reactive with the first antibody (e.g., anti-idiotype antibodies or other antibodies that are specific for the unlabeled immunoglobulin) or other suitable reagent (e.g., labeled protein A).
  • a labeled antibody e.g., a second antibody
  • the antibodies or fragments of the present invention can be utilized in enzyme immunoassays, wherein the subject antibody or fragment, or second antibodies, are conjugated to an enzyme.
  • a biological sample containing cells expressing a mammalian Prox-1 protein, or biological fluid containing secreted Prox-1 is combined with the subject antibodies, and binding occurs between the antibodies and the Prox-1 protein present in the biological sample comprising an epitope recognized by the antibody.
  • These bound protein can be separated from unbound reagents and the presence of the antibody- enzyme conjugate specifically bound to the Prox-1 protein can be determined, for example, by contacting the sample with a substrate of the enzyme which produces a color or other detectable change when acted on by the enzyme.
  • the subject antibodies can be unlabeled, and a second, labeled antibody can be added which recognizes the subject antibody.
  • the present invention also relates to a method of detecting and/or quantitating expression of a mammalian Prox-1 protein or a portion of the Prox-1 protein by a cell, in which a composition comprising a cell or fraction thereof (e.g., a soluble fraction) is contacted with an antibody or functional fragment thereof which binds to a mammalian Prox-1 protein or a portion of the Prox-1 protein under conditions appropriate for binding of the antibody or fragment thereto, and binding is monitored. Detection of the antibody, indicative of the formation of a complex between antibody and or a portion of the protein, indicates the presence of the protein.
  • a composition comprising a cell or fraction thereof (e.g., a soluble fraction) is contacted with an antibody or functional fragment thereof which binds to a mammalian Prox-1 protein or a portion of the Prox-1 protein under conditions appropriate for binding of the antibody or fragment thereto, and binding is monitored.
  • Detection of the antibody indicative of the formation of a complex between
  • the method can be used to detect expression of Prox-1 from the cells of an individual (e.g., in a sample, such as a body fluid, such as blood, saliva or other suitable sample).
  • a sample such as a body fluid, such as blood, saliva or other suitable sample.
  • the level of expression of in a biological sample of that individual can also be determined, for instance, by flow cytometry, and the level of expression (e.g., staining intensity) can be conelated with disease susceptibility, progression or risk.
  • the polynucleotide sequences encoding Prox-1 protein may be used for the diagnosis of conditions or diseases with which the expression of Prox-1 protein is associated.
  • methods for detecting Prox-1 mRNA can comprise contacting a biological sample with a compound that binds to and forms a complex with Prox-1 mRNA for a period sufficient to form the complex, and detecting the complex in a quantitative or semi- quantitative way.
  • Such methods can also comprise amplification techniques involving contacting a biological sample with nucleic acid primers that anneal to
  • Prox-1 mRNA or its complement amplifying annealed polynucleotides, so that if a polynucleotide is amplified, a polynucleotide of the invention is detected.
  • the biological sample can be any tissue or fluid in which Prox-1 -expressing colon cells might be present.
  • Prox-1 protein may be used in hybridization or PCR assays of fluids or tissues from biopsies to detect Prox-1 protein expression. Such methods may be qualitative or quantitative in nature and may include Southern or northern analysis, dot blot or other membrane-based technologies; PCR technologies; dip stick, pin, chip and ELISA technologies. All of these techniques are well known in the art and are the basis of many commercially available diagnostic kits. One such procedure known in the art is quantitative real-time PCR. Real-time quantitative can be conveniently accomplished using the commercially available ABI PRISMTM 7700 Sequence Detection System, available from PE- Applied Biosystems, Foster City, CA and used according to manufacturer's instructions. PCR reagents can be obtained from PE- Applied Biosystems, Foster City, CA.
  • Gene target quantities obtained by real time RT-PCR may be normalized using either the expression level of GAPDH, a gene whose expression is constant, or by quantifying total RNA using RiboGreenTM (Molecular Probes, Inc. Eugene, OR). GAPDH expression is quantified by real time RT-PCR, by being run simultaneously with the target, multiplexing, or separately. Total RNA is quantified using RiboGreenTM (Molecular Probes, Inc. Eugene, OR).
  • RiboGreenTM RNA quantification reagent from Molecular Probes. Methods of RNA quantification by RiboGreenTM are taught in Jones, L.J., et al, Analytical Biochemistry, 1998, 265, 368-374. Controls are analyzed in parallel to verify the absence of DNA in the RNA preparation (-RT control) as well as the absence of primer dimers in control samples lacking template RNA. In addition, RT-PCR products may be analyzed by gel electrophoresis. A reverse transcriptase PCRTM amplification procedure may be performed in order to quantify the amount of niRNA amplified. Methods of reverse transcribing RNA into cDNA are well known and described in Sambrook et ab, 1989.
  • thermostable DNA polymerases are described in WO 90/07641, filed December 21, 1990.
  • Conditions for incubating a nucleic acid probe or antibody with a test sample vary. Incubation conditions depend on the format employed in the assay, the detection methods employed, and the type and nature of the nucleic acid probe or antibody used in the assay.
  • One skilled in the art will recognize that any one of the commonly available hybridization, amplification or immunological assay formats can readily be adapted to employ the nucleic acid probes or antibodies of the present invention.
  • the tests of the present invention include cells, protein extracts of cells, or biological fluids such as, blood, serum, and plasma.
  • test sample used in the above-described method will vary based on the assay format, nature of the detection method and the tissues, cells or extracts used as the sample to be assayed.
  • Methods for preparing protein extracts or membrane extracts of cells are well known in the art and can be readily be adapted in order to obtain a sample which is compatible with ' the system utilized.
  • assays may be useful in evaluating the efficacy of a particular therapeutic treatment regime in animal studies, in clinical trials, or in monitoring the treatment of an individual patient. In order to provide a basis for the diagnosis of disease, a normal or standard measurement of Prox-1 mRNA or protein expression is established.
  • Prox-1 measurements from healthy colon tissue taken from one or more subjects measured using the same or similar reagents used for the test subjects.
  • the healthy subject preferably is matched for sex and age, and optionally, ethnicity. Deviation between standard and subject values conelates with the presence of precancerous or cancerous tissue.
  • a therapeutic agent is administered; and a treatment profile is generated. Such assays may be repeated on a regular basis to evaluate whether the values in the profile progress toward or return to the normal or standard pattern. Successive treatment profiles may be used to show the efficacy of treatment over a period of several days or several months.
  • Methods to quantify the expression of a particular molecule include radiolabeling (Melby et al, J Immunol Methods 159: 235-44, 1993) or biotinylating (Duplaa et al, Anal Biochem 229-36, 1993) nucleotides, coamphfication of a control nucleic acid, and standard curves onto which the experimental results are interpolated.
  • radiolabeling Melby et al, J Immunol Methods 159: 235-44, 1993
  • biotinylating Duplaa et al, Anal Biochem 229-36, 1993
  • screening methods also may be used in a prognostic manner to monitor the efficacy of treatment.
  • the methods may be performed immediately before, during and after treatment to monitor treatment success.
  • the methods also should be performed at intervals, preferably every three to six months, on disease free patients to insure treatment success.
  • kits which contain the necessary reagents to cany out the assays of the present invention.
  • the invention provides a compartment kit to receive, in close confinement, one or more containers which comprises: (a) a first container comprising one of the probes or antibodies of the present invention; and (b) one or more other containers comprising one or more of the following: wash reagents, reagents capable of detecting presence of a bound probe or antibody.
  • a compartment kit includes any kit in which reagents are contained in separate containers. Such containers include small glass containers, plastic containers, or strips of plastic or paper.
  • Such containers allow one to efficiently transfer reagents from one compartment to another compartment such that the biological sample and reagents are not cross-contaminated, and the agents or solutions of each container can be added in a quantitative fashion from one compartment to another.
  • Such containers will include a container which will accept the test sample, a container which contains, for example, the antibodies used in the assay, containers which contain wash reagents (such as phosphate buffered saline, Tris-buffers, etc.), and containers which contain the reagents used to detect the bound antibody or probe.
  • Types of detection reagents include labeled nucleic acid probes, labeled secondary antibodies, or in the alternative, ifthe primary antibody is labeled, the enzymatic, or antibody binding reagents which are capable of reacting with the labeled antibody.
  • labeled nucleic acid probes labeled secondary antibodies, or in the alternative, ifthe primary antibody is labeled, the enzymatic, or antibody binding reagents which are capable of reacting with the labeled antibody.
  • the disclosed probes and antibodies of the present invention can be readily incorporated into one of the established kit formats which are well known in the art.
  • kits for use in detecting the presence of a mammalian Prox-1 protein can include an antibody or functional fragment thereof which binds to a mammalian Prox-1 protein or portion of this protein, as well as one or more ancillary reagents suitable for detecting the presence of a complex between the antibody or fragment and Prox-1 or portion thereof.
  • the antibody compositions of the present invention can be provided in lyophihzed form, either alone or in combination with additional antibodies specific for other epitopes.
  • the antibodies, which can be labeled or unlabeled can be included in the kits with adjunct ingredients.
  • the antibodies can be provided as a lyophihzed mixture with the adjunct ingredients, or the adjunct ingredients can be separately provided for combination by the user.
  • adjunct materials will be present in less than about 5% weight based on the amount of active antibody, and usually will be present in a total amount of at least about 0.001% weight based on antibody concentration.
  • a second antibody capable of binding to the monoclonal antibody can be provided in the kit, for instance in a separate vial or container.
  • the second antibody if present, is typically labeled, and can be formulated in an analogous manner with the antibody formulations described above.
  • Example 1 provides methods and materials for the subsequent Examples.
  • Example 2 provides experimental results of studies designed to assess Prox-1 expression in colorectal cancer cells.
  • Example 3 details expression of Prox-1 in round but not in adherent subclones of the SW480 colon adenocarcinoma cell line.
  • Example 4 provides experimental results of Prox-1 silencing in
  • Example 5 describes effects of Prox-1 ablation on Notch signaling in SW480R cells.
  • Example 6 describes the effects of suppression of Prox-1 on the growth of SW480R cells in soft agar.
  • Example 7 describes the effect Prox-1 suppression on prostaglandin biosynthesis.
  • Example 8 describes experiments aimed as assessing the effects of altered Notch signaling. 5
  • Example 9 describes experiments aimed at assessing the effects of Prox-1 suppression on the growth of SW480R tumors in nude mice.
  • Example 10 describes analysis of Prox-1 in natural colorectal tumors.
  • Example 11 describes one method for diagnosing or screening for colorectal cancer.
  • Example 12 describes experiments designed to compare Prox-1 expression in normal cololnic epithelium.
  • Example 13 describes experiments aimed at assessing Prox-1 expression in ApeTM + mice.
  • Example 14 describes studies conducted using SW480R cell line as an 15 in vitro model to investigate the role of Prox-1 in colorectal carcinoma.
  • Example 15 describes experiments to characterize the effects of Prox-1 suppression and overexpression in colorectal cancer.
  • Example 16 describes experiments employing dominant negative mutants of Prox- 1.
  • Monoclonal mouse anti-vimentin, ⁇ -catenin (Transduction Laboratories), Ki-67 (Pharmingen) and chromogranin A (Ab-3, NeoMarkers), monoclonal rat anti-BrdU (Harlan Seralab) and polyclonal rabbit anti-Prox-1 were obtained from the indicated commercial sources.
  • the fluorochrome-conjugated secondary antibodies were obtained from Jackson Immunoresearch.
  • Prox-1 antibodies cDNA encoding Prox-1 homeobox domain and prospero domain (amino acids 578-750 of human Prox-1, SEQ J_D NO: 3) was subcloned into pGEX2t vector to produce GST-Prox-1 fusion construct. This construct was expressed in E.
  • coli and the GST-Prox-1 fusion protein from E. coli was purified using glutathione Sepharose according to the manufacturer's instructions (Amersham, Piscataway, NJ). Fusion protein was used to immunize rabbits according to a standard protocol.
  • Prox-1 -specific antibodies were isolated from rabbit serum using sequential columns with GST- and GST-Prox-1 -coupled to vinylsulfone agarose resin (Sigma). Purified antibody recognized an 85 kD protein in lysates from 293T cells transfected with Prox-1 but not from cells transfected with the empty vector.
  • siRNA duplexes were prepared from synthetic 21 nucleotide RNAs
  • siRNA sequences were: 5'-
  • EGFP-targeting control siRNA A18 was essentially as described (Lewis et al., 2002) except that instead of thymidine 3' overhangs uracil overhangs were used; GACGUAAACGGCCACAAGUUU (EGFP siRNA A18 sense) (SEQ ID NO: 8); ACUUGUGGCCGUUUACGUCUU (EGFP siRNA A18 antisense) (SEQ ID NO: 9).
  • siRNAs were 2'-ACE deprotected according to the manufacturer's instructions, dried in vacuum, resuspended in 400 ⁇ l water, dried again, resuspended in water, and annealed to form duplex siRNAs. For anneahng equimolar amounts of siRNA strands (approximately 50-100 ⁇ M) were incubated in anneahng buffer
  • siRNA concentration was measured by spectrometry and siRNA aliquoted and stored at -20°C.
  • SW480 cells were obtained from ATCC (CCL-228) and cultured in
  • RPMI- 1640 supplemented with 10% fetal bovine serum, 1 mM glutamine and antibiotics.
  • HepG2 cells were cultured in DMEM, containing 10% fetal bovine serum 1 mM glutamine and antibiotics.
  • Transfection of siRNAs was canied out using Lipofectamine 2000 (Invitrogen) according to manufacturer's instructions using 0.5% (v/v) lipofectamine 2000 reagent for SW480R and 0.4% (v/v) hpofectamine 2000 for adherent SW480 cells and either 20nM or lOOnM (f.c.) of siRNA. Transfections were carried out in antibiotic-free media for 4-6 hours before changing cells back to normal culture media.
  • RNA transfections were repeated after 48-72h from previous transfection (at protein level the silencing effect was seen to remain efficient for at least 96h). Normally approximately 90-95% transfection efficiency was achieved.
  • Opti-MEM (Invitrogen) medium was used in preparation of transfection mixtures.
  • GFPsi RNA Green Fluorescent Protein small interfering RNA
  • Prox-1 siRNAs 72 h prior to the transfection with the firefly lucefarese reporter constructs CBFl-luc, control pGL2- luc (Promega), TOPFlash and FOPFlash (Upstate).
  • RNeasy columns Qiagen
  • filters were hybridized in ExpressHyb with 32P-labeled probes for LYVE-l and Prox-1 according to the manufacturer's instructions (Clontech).
  • Northern analysis the blots were hybridized in Ultrahyb solution (Ambion) with 32P-labeled probes produced by RT-PCR using RNA from SW480R or SW480A cells.
  • the primers were designed to amplify 300-700 bp of the coding sequence, and all PCR-fragments were sequenced to confirm their identity.
  • sample preparations and hybridizations were carried out as described (Petrova et al. Embo J 21: 4593-9, 2002), using RNA extracted from two clones of SW480R or SW480A cells, or from two independent transfections of two different clones of SW480R cells with GFP siRNA or Prox-1 siRNA A16. To confirm the latter results, another transfection was carried out using Prox siRNA A25. To exclude the non-specific effects due to the transfection itself non-transfected SW480R cells grown in parallel were also analyzed.
  • Prox-1 mRNA is Elevated in Colorectal Tumors
  • a cancer gene profiling a ⁇ ay filter which contains cDNAs from about 250 human cancers and conesponding normal control tissues.
  • Prox-1 mRNA was significantly increased in 35 out of 53 samples of colorectal cancers.
  • Fig. IA In contrast, only rarely or not at all was any increase seen in samples from breast, uterine, lung, kidney, ovarian, or thyroid tumors (Fig. IA, B, and C).
  • Probes for Prox-1 (Fig. IA) and the lymphatic endothelial marker LYVE-l (Fig. IB) were used.
  • Fig. IC demonstrates quantification of dot blot in Fig. IA, the asterisk indicating tumor samples in which Prox-1 expression is significantly different from that of the normal tissue (P ⁇ 0.005). Expression of Prox-1 was low or absent in all kidney cancer samples studied.
  • Prox-1 is a marker for lymphatic vessels, which are abundant both in normal colonic submucosa and around colon carcinomas (White et al., Cancer Res.
  • Staining of a panel of mouse tissues and E12.5 and E17.5 embryos revealed specific nuclear staining for Prox-1 in the previously reported sites of expression such as in lymphatic vessels, lens fiber cells and in a subset of neurons in the neural tube.
  • Staining of eleven human colorectal adenomas and nine carcinomas and adjacent normal mucosa revealed increased expression of Prox-1 in nine adenomas and in six carcinomas (Fig. 2A-I).
  • Increased Prox-1 staining was observed in all cells in seven adenomas and in two carcinomas, whereas in the other lesions a heterogeneous expression of Prox-1 occuned.
  • PROX-1 IS EXPRESSED IN ROUND BUT NOT IN ADHERENT SUBCLONES OF THE SW480 COLON ADENOCARCINOMA CELL LINE. Additional studies were conducted to compare Prox-1 expression in various cells. No Prox-1 expression was seen in the majority of tumor cell lines studied. However, Prox-1 was mRNA was present in hepatocellular carcinoma cell line HepG2 and the colon carcinoma cell line SW480. BEC, blood endothelial cells, CAEC, coronary artery endothelial cells, and LEC, lymphatic endothelial cells, served as negative and positive controls.
  • SW480R round cells displayed anchorage independent growth in vitro and highly malignant phenotype in vivo, whereas the SW480A (adherent) cells did not grow well in soft agar and formed small and well differentiated tumors when implanted into nude mice.
  • SW480R and SW480A clones were isolated, which could be continuously grown for at least 20 passages without conversion of phenotypes.
  • SW480R and SW480A cells differed by the levels of Prox-1, as determined by Northern and Western blotting, with much higher expression in the round cells, and weak, if any, expression in the Adherent ones.
  • SW480R and SW480A cells were compared using oligonucleotide microarrays containing 22,000 annotated human genes, and identified about 1,000 genes whose expression differed by more than fourfold between these two cell types (Table I).
  • SW480 cells were stained for intermediate filament protein vimentin and Prox-1.
  • Northern blotting and hybridization were used for transcripts. Hybridization for
  • GAPDH was used as a control.
  • the SW480R cells lacked many components of the actin, intermediate filament and microtubule networks, such as gelsolin, filamins A and B, ezrin, moesin, vimentin, various integrins, and tubulins (Table I). These cells expressed higher levels of the protoncogene c-met, as well as the receptor tyrosine kinase FGFR-4, which has been associated with malignant transformation in colorectal and other cancer (Bange, J. et ab, Cancer Res.
  • FGFR-4 is a target for therapeutic intervention according to the invention, alone or in combination with Prox-1. Intervention using the same classes of inhibitors as described for Prox-1, as well as antibodies and antibody fragment substances, is specifically contemplated. In addition, all three tissue inhibitors of matrix metalloproteinases were absent from the SW480R cells, which may further account for thek increased tumor growth in vivo.
  • SW480A cells expressed higher levels of the chemokine receptor CXCR4, which is expressed in the normal colonic epithelium (Jordan et ab, J Clin Invest 104, 1061-9, 1999).
  • CXCR4 chemokine receptor 4
  • Table I Examples of groups of genes differentially expressed in round versus adherent SW480 clones. Two round and two adherent clones were analyzed. Gene function and name UniGene Gene Log 2 ratio, St. cluster symbol average dev
  • PDZ and LJM domain 1 (elfin) Hs.75807 PDLEVI1 -2.7 0.2 cadherin 11, type 2, OB-cadherin (osteoblast) Hs.75929 CDH11 -2.6 0.7 myosin IC Hs.286226 MYOIC -2.6 0.6 integrin, alpha 3 Hs.265829 1TGA3 -2.6 0.4 discs, large (Drosophila) homolog 1 Hs.154294 DLG1 -2.5 0.1 integrin, alphaV Hs.295726 LTGAV -2.5 0.3
  • Notch homolog 2 (Drosophila) Hs. .8121 NOTCH2 -1.4 0.15 hairy homolog (Drosophila), HES1 Hs. .250666 HRY -2.1 0.2 jagged 2 Hs. .166154 JAG2 1.6 0.61
  • EXAMPLE 4 PROX-1 SILENCING IN SW480R CELLS LEADS TO A DIFFERENTIATED AND QUIESCENT PHENOTYPE.
  • Prox-1 mRNA and protein in the SW480R cells was suppressed using Prox-1 targeting siRNA. Absence of Prox-1 in Prox-1 siRNA but not the control GFP siRNA transfected cells was confirmed by immunofluorescent staining, and nuclei were visualized with Hoechst 33342.
  • Prox-1 siRNA-transfected cells but not the untransfected or GFP siRNA transfected cells underwent a morphological change, which became visible by 72 hours and persisted at least for 10 days after the transient transfection.
  • the Prox-1 siRNA transfected cells become first more elongated and displayed extensive membrane ruffling.
  • the Prox-1 siRNA cells started to spread on the plate and a number of increased actin stress fibers could be visualized by phalloidine staining.
  • Prox-1 at least partially determines the phenotype of SW480R cells.
  • the ablation of Prox-1 led to upregulation of a number of known epithelial markers, such as annexin Al, CRPB2, S100A3, and EMP1, along with the increase in cell adhesion molecules OB-cadherin and integrins beta7, beta5 and alpha 1.
  • epithelial markers such as annexin Al, CRPB2, S100A3, and EMP1
  • OB-cadherin and integrins beta7, beta5 and alpha 1 In line with the observed growth arrest, also observed was the decrease in c-myc and a strong increase of CDK inhibitor p21Cipl.
  • Asterisk indicates genes that were flagged as absent in either Prox-1 siRNA or GFP siRNA treated cells. Genes differentially expressed between SW480R and SW480ADH cells are shown in bold. Genes down-regulated in the absence of Prox-1 UniGene Gene Log 2 ratio, side 1 cluster symbol average
  • FK506 binding protein 11 19 kDa Hs.24048 FKBP11 -1.4 0.1 caspase recruitment domain family, member 10 Hs.57973 CARDIO -1.3 0.1 inhibin, beta B (activin AB beta polypeptide) Hs.1735 HMHBB -1.3 0.2
  • LI cell adhesion molecule Hs.1757 L1CAM -1.2 0.1 glutathione peroxidase 2 (gastrointestinal) Hs.2704 GPX2 -1.2 0.0 eukaryotic translation elongation factor 1 alpha Hs.2642 EEF1A2 -1.2 0.2
  • KIAA0182 protein Hs.75909 KIAA0182 -1.1 0.0 lectin, galactoside-binding, soluble, 1 (galectin 1) Hs.382367 LGALSl -1.1 0.1
  • CAMP-specific hosphodiesterase 8Bl [Homo Hs.78106 PDE8B 2.3 0.4 sapiens], mRNA sequence* ectodermal-neural cortex (with BTB-like Hs.104925 ENC1 2.3 0.2 domain)* frizzled homolog 1 (Drosophila)* Hs. 94234 FZD1 2.3 0.8
  • C/EBP CCAAT/enhancer binding protein
  • CEBPD 2.1 0.1 sorbin and SH3 domain containing 1 Hs. 108924 SORBS1 2.1 0.5 metallothionein 2A Hs. 118786 MT2A 2.0 0.6
  • RAS guanyl releasing protein 1 (calcium and DAG- Hs. 182591 RASGRPl 2.0 0.4 regulated) checkpoint suppressor 1 Hs .211773 CHES1 2.0 0.4 chondroitin betal,4 N- Hs.11260 ChGn 2.0 0.4 acetylgalactosa inyltransferase* filamin B, beta (actin binding protein 278)* Hs, .81008 FLNB 2.0 0.4 aldehyde dehydrogenase 1 family, member A2* Hs. 95197 ALDH1A2 2.0 0.6 jagged 1 (Alagille syndrome) Hs. 91143 JAG1 2.0 0.1
  • IQ motif containing GTPase activating protein 1 Hs.1742 IQGAPl 1.3 0.5 zinc finger protein 216 Hs.406096 ZNF216 1.3 0.2 cDNA DKFZp564O0122 - - 1.3 0.2 aryl hydrocarbon receptor Hs.170087 AHR 1.2 0.6 neuroepithelial cell transforming gene 1 Hs.25155 NET1 1.2 0.1 sterol-C4-methyl oxidase-like Hs.239926 SC4MOL 1.2 0.1 tubulin, alpha 3 Hs.433394 TUBA3 1.2 0.1
  • HTV-1 Tat interactive protein 2 30kDa Hs.90753
  • HTATIP2 1.0 0.1 insulin receptor substrate 2 Hs.143648 ms2 1.0 0.1 tubulin beta-5 Hs.274398 TUBB-5 1.0 0.0 apoptosis antagonizing transcription factor Hs.16178 AATF 1.0 0.1
  • E2F transcription factor 3 Hs.H89 E2F3 1.0 0.1 hypothetical protein FLJ12542 Hs.236940 FLJ12542 1.0 0.1 phai ⁇ n 2, Pleckstrin homology domain containing, Hs.29724 PLEKHF2 1.0 0.1 family F member 2 proline 4-hydroxylase Hs.3622 P4HA2 1.0 0.1
  • Prox-1 did not affect the activity of ⁇ -catenin TCF-responsive reporter or nuclear localization of ⁇ -catenin.
  • an increased expression of several ⁇ - catenin/TCF-4 target genes, such as CD44, ENC1 and Id2 was observed in the absence of Prox-1 (Table JJ and not shown).
  • ⁇ -catenin/TCF signaling inhibitors may include dominant negative forms of TCF-4, siRNAs and microRNAs targeting TCF-4, ⁇ - catenin, and c-myc, as well as small molecules that would interfere with binding of ⁇ - catenin to TCF-4 or TCF-4 to target DNA sequences. Protocols for making these types of inhibitors are detailed above with respect to Prox-1 inhibition.
  • the DNA and protein sequences for ⁇ -catenin (SEQ ID NOs: 10 and 11, respectively) are published and disclosed as Genbank Accession Number NM_001904.
  • the DNA and protein sequences for TCF-4 (SEQ ED NOs: 12 and 13, respectively) are published and disclosed as Genbank Accession Number NM_003199.
  • APC gene the sequence of which is publicly available as Genbank Accession Number NM_000038.
  • the DNA and amino acid sequences for APC are also provided herein as SEQ ID NOs: 42 and 43, respectively.
  • SEQ ID NOs: 44 and 45, respectively The DNA and protein sequences for C-myc (SEQ ID NOs: 44 and 45, respectively) are published and disclosed as Genbank Accession Number NM_002467. Notch signaling has been shown to be essential for the generation of cell lineages in the crypts of the mouse small intestine.
  • Notch2 and its target transcription factor Hesl levels are higher in SW480A cells in comparison with the SW480R cells, suggesting that this pathway is functionally active in these cells.
  • SW480R cells express higher levels of Notch ligand Jagged2. Suppression of Prox-1 resulted in upregulation of the Notch ligand Jagged 1 and the dkect target of the Notch pathway, the transcription factor Hey2, whereas the expression of Jagged2 and prostaglandin D2 synthase, previously shown to be negatively regulated by Notch signaling was suppressed (Fujimori, K. et al., JBiol Chem 278: 6018-26, 2003).
  • SW480R cells were transfected with GFP siRNA or Prox-1 siRNA and GFBl-luc, TOPFlash or control FOP flash reporters.
  • Fkefly luciferase activity was normalized to Renilla luciferase activity.
  • Up-regulation of Notch-responsive reporter GBFl-luc was observed in SW480R cells transfected with Prox-1 siRNAs.
  • contemplated herein is a method of alleviating the symptoms of colorectal cancer comprising the administration of a Prox-1 suppressor in combination with a Notch agonist or target transcription factor.
  • Notch agonists include Jagged 1, Jagged2, Deltal, Delta3, Delta4, and Senate.
  • Target Notch transcription factors include Heyl, Hey2, and Hesl.
  • the DNA and protein sequences for Notch-1 (SEQ ID NOs: 14 and 15, respectively) are pubhshed and disclosed as Genbank Accession Numbers
  • NM_017617 the DNA and protein sequences for various forms of Notch (including 2-4) are publicly available and included herein as SEQ ID NOs: 16-21.
  • the DNA and protein sequences for various ligands of Notch are publicly available and included herein as SEQ ID NOs: 22- 35, respectively.
  • DNA and protein sequences for target Notch transcription factors Heyl, Hey2, and Hesl are also publicly available and are included herein as SEQ ID NOs: 36-41, respectively.
  • EXAMPLE 6 SUPPRESSION OF PROX-1 mfflBITS GROWTH IN SOFT AGAR.
  • SW480R cells were transfected with GFP siPRNA, Prox-lsiRNA A16 or Prox-1 siRNA A25 repeatedly over an 8-day period, or left untreated, and seeded in soft agar in triplicate. The number of colonies was scored after two weeks of growth. Transfection with both Prox-1 siRNAs but not the control GFP siRNA significantly decreased the number of colonies formed after two weeks of growth in soft agar (Fig. 3A).
  • COX-2 is a key enzyme involved in the conversion of arachidonic acid into the prostaglandin precursors PGG2 and PGH2, which are further transformed into biologically active prostaglandins by the action of conesponding synthases. Prostaglandins acts through binding to the G-protein coupled prostanoid receptors and they are rapidly inactivated by the action of 15-prostaglandin dehydrogenase (15- PGDH). COX-2 is overexpressed in the majority of colorectal cancers and in about half of colonic adenomas, suggesting that the increased PG production is important for tumor growth.
  • contemplated herein is a method of alleviating colorectal cancer via the administration of a Prox-1 suppressor in combination with a COX-2 inhibitor.
  • Cox-2 inhibitors may include the following non-steroid anti-inflammatory drugs: asprin, rofecoxib, celecoxib ,amidophen, analgin, anapyrin, feloran, indomethacin, paracetoamol, pkoxicam, sedalgin, diclofenac sodium, ketoprofan, Acular ® , Ocufen ® , and Voltarol ® .
  • SW480 cells do not express COX-2, contemplated herein are experiments to assess the effects of Prox-1 on prostanoid biosynthesis in the SW480F cells stably transfected with COX-2 or in the cell line which is known to express this enzyme, such as HCA-7.
  • SW480F cells are transfected with a mixture of a COX-2 expressing vector and the plasmid bearing hygromycin resistance gene, such as pCDNA3.1hygro (Invitrogen) using Lipofectamine 2000, as described in Materials and Methods, and the stable clones are selected using 200 ⁇ g/ml hygromycin B (Calbiochem) over a period of 2-3 weeks.
  • COX-2 expressing cells can be infected with AdProx-1 or the control AdGFP virus, as described previously (Petrova et ab, Embo J. 21: 4593-9), and the amount of biologically active PGE2, and total amount of metabolized PGE2 in cell conditioned medium, determined by ELISA (Cayman Chemicals).
  • EXAMPLE 8 EFFECTS OF NOTCH SIGNAL TRANSDUCTION
  • SW480R cells are infected with adenoviruses.
  • Nu/nu mice can be inoculated subcutaneously or intraperitoneally with 1-5 x 10 6 cells/mice using SW480R cells transfected with GFPsi RNA or Prox-1 siRNA, or transduced with the adenoviruses described in Example 8. Tumors are allowed to grow for 3-5 weeks, and tumor size measured twice a week. Animals are sacrificed by cervical dislocation, tumors excised, and processed for immunohistoshemical staining. The tumor histology, expression of differentiation markers, proliferation index and vascularization momtored using the antibodies against KI67 (prohferation), mucin, galectin-2, p21cipl (differentiation), PECAM-1 and vWF (blood vessel markers), and the standard immunostaining protocols.
  • SW480R or HCA-7 cells recombinantly overexpressing 15-PGDH and control cells are implanted subcutaneously into the nu/nu mice, and tumor growth and differentiation studied.
  • 15-PGDH inhibitor CAY10397 administered intravenously, or in drinking water.
  • EXAMPLE 10 ANALYSIS OFPROX-1 INNATURAL COLORECTAL TUMORS
  • Ape min/+ mice bear a truncating mutation in one allele of Ape gene, and develop multiple intestinal polyps, which further progress to adenocarcinoma.
  • Mice are commercially available from JAX.
  • SMAD3 deficient mice which develop invasive colorectal cancer, is available.
  • the DNA and protein sequences for APC (SEQ ID NOs: 42 and 43, respectively) are published and disclosed as Genbank Accession Number NM_000038.
  • Prox-1 inhibitors and administration thereof are described herein.
  • Prox-1 inhibitors available for administration include, but are not hmited to, antisense oligonucleotides, siRNA constructs, or dominant negative proteins.
  • Monitoring of the mice post-administration is contemplated to evaluate the effects of adenocarcinoma and colorectal cancer development and growth. Among the results are measurements of the speed of tumor growth in mice that received the Prox-1 inhibitor versus mice that received the placebo, thus, providing a beneficial efficacy model for the particular Prox-1 inhibitor.
  • methods for screening Prox-1 levels in family members with familial adenomatous polyposis are also contemplated.
  • Prox-1 levels are described herein. Administration of a prophylactic to protect from progression, or the onset of cancer, is contemplated where elevated levels of Prox-1 are observed.
  • EXAMPLE 11 DETECTION OF PROX-1 PROTEIN EXPRESSION IN COLORECTAL CANCER
  • measuring Prox-1 protein expression in colon tissues may be a useful tool for diagnosing colon cancer and/or premalignancies.
  • Prox-1 mRNA can be detected in colorectal cancer tissues as described in Example 2. The following prospective example may be conducted to determine whether Prox-1 protein conelates with Prox-1 transcript expression in colorectal cancer tissue.
  • the immunohistochemical analysis can be carried out as follows using an anti-human Prox-1 antibody dkected against the human Prox-1 peptide, as described in Example 1.
  • the tissues for screening are snap frozen in liquid nitrogen after dissection, embedded in OCT compound, and sectioned. Sections are fixed on -20°C methanol for 10 min, and processed for staining.
  • the tissues may undergo steam induced epitope recovery with a retrieval solution, including several different SHIER solutions with and without enzyme digestion at two different concentrations.
  • the tissues can then be heated in the capillary gap in the upper chamber of a Black and Decker Steamer as described in Ladner et al. (Cancer Research, 60: 3493-3503, 2000).
  • TECHMATE 1000 or TECHMATE 500 BioTek Solutions, Ventura Medical System
  • the tissues are blocked with 3% and 10 % normal goat serum for 15 and 30 minutes respectively.
  • the tissues are incubated with the primary antibody (anti-Prox-1 antibody) for 60 minutes at 3.0 *g/ml.
  • the tissues are stained with the biotinylated goat-anti-rabbit IgG secondary antibody for 25 minutes. Optimal results are obtained with overnight incubation.
  • anti- vimentin is used as a positive control and rabbit IgG is used as a negative control.
  • the antibody binding is detected by an avidin-biotin based tissue staining system where horse-radish peroixidase is used as a reporter enzyme and DAB (3,3'-Diaminobenzididine Tetrahydrochloride) is used as a chromogen. Specifically, the endogenous peroxides are blocked for 30 minutes, the avidin-biotin complex reagent is added and then the tissues are incubated in DAB for a total of 15 minutes. Finally, the tissues are counterstained with hemotoxylin to assess cell and tissue morphology. The slides are mounted in Aquamount, and the tissues are examined visually under a light microscope.
  • DAB 3,3'-Diaminobenzididine Tetrahydrochloride
  • Various options for detecting Prox-1 expression, and, therefore screen for colon cancer include, among others, ELISA-based techniques and Western blotting techniques.
  • EXAMPLE 12 EXPRESSION PATTERN OF PROX-1 IN NORMAL COLONIC EPITHELIUM Studies were conducted to compare Prox-1 expression in normal cololnic epithelium. In normal colonic mucosa, all Prox-1 expressing cells were positive for the intestinal epithelial transcription factor CDX2.
  • Prox-1 may be involved in the regulation of the neuroendocrine cell fate as well as the stem cell phenotype. This hypothesis is supported by the fact that PROX-1 is overexpressed in intestinal neoplasms from Apc ⁇ un/+ mice and that its expression is regulated by TCF/ ⁇ -catenin pathway in vitro (see Examples 13 and 14). This hypothesis is also in agreement with previous results showing that targeted inactivation of Tcf712 gene encoding TCF-4 leads to the depletion of intestinal stem cell compartment and loss of neurodendocrine lineage (Korinek, V.
  • EXAMPLE 14 PROX-1 EXPRESSION IS REGULATED BY ⁇ -CATENIN/TCF PATHWAY AND DNA METHYLATION Further studies were conducted using SW480R cell line as an in vitro model to investigate the role of Prox-1 in colorectal carcinoma.
  • SW480R and SW480A cells were also conducted to compare the activation of ⁇ - catenin/TCF signaling pathway in SW480R and SW480A cells.
  • the SW480R cells had slightly more active ⁇ -catenin and displayed a two-fold increase in the activation of the TCF-responsive promoter TopFLASH; however, both cell lines clearly displayed nuclear localization of ⁇ -catenin as previously reported (Palmer, H. G. et al., / Cell Biol 154, 369-87 (2001)).
  • mice do not develop intestinal adenomas when crossed with Apc m /+ mice. In contrast, they develop lymphomas, demonstrating cell type specific effects of decreased DNA methylation for cancerous growth (Gaudet, F. et al., Science 300, 489-92 (2003), Eads, C. A. et ab, Cancer Res 62, 1296-9 (2002)).
  • EXAMPLE 15 PROX-1 SUPPRESSION AND OVEREXPRESSION IN COLORECTAL CANCER
  • stable colorectal cancer cell hne clones inducibly expressing Prox-1 or Prox-1 targeting siRNAs are employed.
  • Prox-1 or Prox-1 siRNA expressing lentivkuses are employed to provide long-term expression in colorectal cancer cell lines in vitro and in vivo.
  • Prox-1 cDNA was subcloned in pTetOS vector (Sarao and Dumont, Trans enics Res., 1998) , where it is placed under the control of doxycycline regulated promoter.
  • Prox-1 siRNAs were subcloned in pTer vector (van der Wetering et al., Embo Reports, 2003). Colorectal carcinoma cells stably expressing tTA activator may be transfected with Prox-1/TetOS or Prox-1 siRNS/pTer vectors. Clones may be selected in the presence of blasticidine and G480 and further tested for the expression of Prox-1 by immunostaining or Prox-1 siRNA by suppression of co-transfected Prox- 1 in the presence of doxycycline. For production of Prox-1 lentiviruses, Prox-1 cDNA was subcloned into FUkesGFPW (Lois et al., Science , 2002).
  • Prox-1 siRNAs 1 and 2 were subcloned into lentiviral vector pLL3.7 (Rubinson et ab, Nat Genet., 2003). Sequences of the DNA oligos used in the cloning of pLL3J-Prox-l : sense: TGGTCATCTGCAAGCTGGATTTCAAGAGAATCCAGCTTGCAG ATGACCTTTTTC (SEQ ID NO 47). antisense: TCGAGAAAAAAGGTCATCTGCAAGCTGGATTCTCTTGAAATCCAGCTTGC AGTGACCA (SEQ ID NO 48).
  • pLL3J PROX1-2 sense: TGAGCCAGTTTGATATGGATTTCAAGAGAATCCATATCAAACTGGCTCTTT TTTC (SEQ ID NO 49).
  • antisense TCGAGAAAAAAGAGCCAGTTTGATATGGATTCTCTTGAAAT CCATATCAAACTGCTCA (SEQ ID NO 50).
  • Inducible Prox-1 targeting short hairpin RNA (“shRNA”) expression may also be achieved via CRE recombinase activated induction system whereby an inactivating stuffer DNA sequence sunounded by modified loxP sites is removed from an shRNA expression cassette by the CRE recombinase activity, thus activating the shRNA expression.
  • a similar system may be used to inactivate shRNA expression upon introduciton of CRE recombinase. Tiscornia et al PNAS 2004, and Coumoul et al NAR 2004) described these systems.
  • shRNA or "short hairpin RNA” is a short sequence of RNA which makes a tight hairpin turn and can be used to silence gene expression.
  • This small hairpin RNA was first used in a lentiviral vector. (Abbas-Terki T. et al, Hum. Gene Ther. 13(18):2197-201 (2002)). shRNA generates siRNA in cells (An DS et & ⁇ ., Hum. Gene Ther. 14(12): 1207-12 (2003)).
  • intestinal-specific promoter such as villin, CyplA or FABPi are created using standard techniques.
  • the proliferation and differentiation status of intestinal epithelial cells is studied by staining of intestinal tissues for PCNA, Ki67, CDKNIA, mucins, lysozyme, chromogranin A and carboxipeptidases II and IV.
  • the crossing of Prox-1 transgenic animals with Apc 1 " 111 ⁇ mice permits determination of whether Prox-1 overexpression influences the number and size of intestinal polyps in this mouse model of colorectal cancer.
  • Prox-1 cDNA was subcloned in pl2.4Vill plasmid, which places it under the control of 12.4 kb mous villin promoter (Madison et al., J.Biol.Chem.2002, genomic contig NT_039170).
  • the construct may be used for the production of villin- Prox-1 transgenic mice, which will overexpress Prox-1 at the sites of villin expression, i.e. intestinal epithelial cells.
  • Also contemplated is subcloning Prox-1 cDNA into the vector z/AP (Lobe et al., Dev.
  • Prox-1 cDNA is placed between the loxP sites, and it is not expressed until Cre recombinase is present in the same cell. Excision of loxP sites places the transgene under the control of chicken ⁇ -actin promoter .
  • the transgenic animals containing z/AP- Prox-1 expression cassettes in thek genomes may be crossed with villin-Cre mice (Madison et al., J.Biol.Chem.2002). The latter approach may be preferable to the villin-PROXl overexpression because of potentially higher expression levels of the transgene.
  • a Prox-1 mutant protein lacking the transactivation domains or DNA binding domains may act in a dominant negative manner.
  • Disruption of the DNA binding domain entails truncation of the protein to exclude amino acids 572-634 of SEQ ID NO. 3, based on homology to Prospero (Drosophila).
  • Disruption of the transactivation domain entails the deletion of amino acids 635-737.
  • the construct can be used for the generation of transgenic animals with the purpose of suppression of Prox-1 effects in vivo, or for the anti- Prox-1 therapies in colorectal cancer.
  • the foregoing examples are intended to be illustrative of the invention and not intended to limit the claims which define the invention. All patent, journal, and other literature cited herein is incorporated herein by reference in the entirety. While the invention is described specifically with respect to Prox-1, there are other genes described in tables herein that are differentially expressed. All materials and methods described herein are applicable to the genes described in the tables.

Abstract

The invention provides materials and methods for colorectal cancer screening, diagnosis, and therapy.

Description

MATERIALS AND METHODS FOR COLORECTAL CANCER SCREENING, DIAGNOSIS, AND THERAPY
Fl-ELD OF THE INVENTION The present invention relates generally to methods and materials for altering colorectal cancer progression. The present invention also relates to techniques for screening for colon cancer and/or premalignancies.
BACKGROUND The transcription factor Prox-1 is expressed in a number of tissues during embryonic development, including lens fiber cells, subpbpulation of neurons in brains and neural tube, skeletal muscle, heart, liver, pancreas and lymphatic endothelial cells. Targeted inactivation of Prox-1 results in the defects of eye development because of the failure of lens fiber cells to elongate (Wigle et al., Nat. Genet. 21: 318-22, 1999). Prox-1 is also necessary for the migration of hepatocytes during liver development (Sosa-Pineda et al., Nat. Genet. 25: 254-5, 2000). In addition, Prox-1 deficient embryos lack lymphatic vasculature, while the blood vessel development is not affected (Wigle et ab, Cell 98: 769-778, 1999). Recently, others and we have demonstrated the essential role of Prox-1 in the regulation of the lymphatic endothelial phenotype. Overexpression of Prox-1 in blood vascular endothelial cells, where it is otherwise absent, leads to the increased expression of lymphatic endothelial markers and to the suppression of the genes characteristic for the blood vascular endothehal lineage (Petrova et al., Embo J. 21: 4593-9, 2002; Hong et al., Dev. Dyn. 225: 351-7, 2002). Notch is a transmembrane protein that acts as a receptor in a cell-cell signaling mechanism, and in combination with other cellular factors, influences differentiation, proliferation and apoptotic events at all stages of development (Artavanis-Tsakonas, Science 284: 770-776, 1999). In animal models, mutations in the Notch receptor have resulted in developmental abnormalities (Joutel et al., Nature 383: 707, 1996; Lb et al., Nature Genet. 16:243, 1997). Cancer treatments generally promote tumor regression by inhibiting tumor cell proliferation, inhibiting angiogenesis (growth of new blood vessels that is necessary to support tumor growth) and/or prohibiting metastasis by reducing tumor cell motility or invasiveness. Colon cancers are a very common malignancy and colon cancers are typically adenocarcinomas, or sometimes carcinoid tumors. Treatment is primarily surgical resection of the colon, although chemotherapy has been found to be beneficial in some cases. These treatment options for colon cancer are of unpredictable and sometimes limited value, especially if the cancer has not been identified and removed at early stages. There continues to exist a need for novel therapies and diagnostic methods for cancer conditions.
SUMMARY OF THE INVENTION The present invention addresses one or more ongoing needs by providing materials and methods for screening for and treating cancerous and precancerous conditions, especially colorectal in nature. As one aspect, the invention provides materials and methods to screen a mammalian subject for a cancerous or precancerous condition based on analysis of Prox-1 expression in cells from the mammalian subject. In particular, materials and methods are provided for screening colon tissue for signs of cancerous or precancerous pathology. For example, the method includes a method of screening colon tissue for a pathological condition, said method comprising: measuring Prox-1 expression in a biological sample that comprises colon tissue from a mammalian subject, wherein elevated Prox-1 expression in the colon tissue correlates with a pathological phenotype. The determination of elevated Prox-1 expression is generally made by way of a comparison, e.g., to a measurement of Prox-1 expression in healthy colon tissue (from the same subject or others of the same species, preferably matched for sex, age, race, or other characteristics); or to a measurement of Prox-1 expression in diseased (especially neoplastic) colon tissue. When comparing Prox-1 expression in the colon tissue to Prox-1 expression in healthy colon tissue, an increased (e.g., elevated) Prox-1 expression in the colon tissue from the mammalian subject correlates with a pathological phenotype. When comparing to diseased tissue, comparable levels of expression in the tissue from the subject coπelates with a pathological phenotype. In another, related example, the invention includes a method of screening colon tissue for a pathological condition, the method comprising steps of:
(a) obtaining a biological sample comprising colon tissue from a mammalian subject;
(b) measuring Prox-1 expression in the colon tissue; and (c) screening for the presence or absence of a pathological condition from the measurement of Prox-1 in the sample. Similarly, the invention includes a method of screening colon tissue for a pathological condition, the method comprising steps of: (a) obtaining a biological sample comprising colon tissue from a mammalian subject; (b) measuring Prox-1 expression in the colon tissue; and (c) comparing Prox-1 expression in the colon tissue to Prox-1 expression in healthy colon tissue, wherein increased Prox-1 expression in the colon tissue corcelates with a pathological phenotype. For this type of method, the term "pathological condition" is intended to include any abnormality or evidence of disease that warrants medical treatment or monitoring due to concern of developing disease. Cancers and precancerous changes in tissue are particularly contemplated. Thus, in preferred embodiments, the method can be characterized as a screen for colon cancer or colorectal cancers, and increased Prox-1 expression in the colon tissue is scored as being indicative of a cancerous or precancerous condition. The method can be combined with any other molecular, cellular, pathological, or patient symptom criteria to assist a medical practitioner in early diagnosis and therapeutic or prophylactic therapy. For example, in one variation, the method further comprises measuring expression of at least one gene or protein selected from the group consisting of CD44, Encl, and ID2 in the colon tissue, wherein elevated Prox-1 expression and elevated expression of the at least one gene/protein in the colon tissue conelate. with a pathological phenotype. In another variation, the method further comprising measuring activation of -catemn/TCF pathway in the colon tissue, wherein activation of the -catemn/TCF pathway and elevated Prox-1 expression in the colon tissue correlate with a pathological phenotype. Activation of the -catemn/TCF pathway can be measured by a variety of indicators, including mutations in an APC gene; mutations in a -catenin gene; and nuclear localization of -catenin. The biological sample is any tissue or fluid sample obtained in any way from a mammalian subject that includes cells from the large intestine. Biopsies or other surgically removed specimens are preferred. Stool or feces may contain sufficient colon tissue for some embodiments of the assay. The assay may be performed on any mammalian subject, including laboratory animals used in cancer research, livestock, and domestic pets. Humans are most prefened. Any available technique can be used for measuring Prox-1 expression, including direct and indirect techniques. For example, in one variation, the measuring comprises measuring Prox-1 protein in the biological sample. Prefened techniques for measuring amounts or concentrations of Prox-1 protein in a sample are immunological techniques that involve use of a polyclonal or monoclonal antibody that specifically binds Prox-1, or use of a Prox-1 -binding fragment of such an antibody. For example, the measuring comprises contacting the colon tissue with a Prox-1 antibody or antigen-binding fragment thereof. Quantification of the amount of bound antibody (e.g., using a label or second, labeled antibody) provides a measurement of Prox-1 protein expressed in the sample, frnmunoassays such as radioimmunoassay, immunoradiometric assay (labeled antibody), or an enzyme- linked immunosorbent assay (ELISA) are contemplated. In another variation, the measuring comprises measuring Prox-1 mRNA in the colon tissue. Elevated levels of Prox-1 mRNA in the sample are scored as elevated Prox-1 expression. Any available assay for measuring specific oligonucleotides is suitable. Prefened materials for such measurements are oligonucleotide probes complementary to all or a portion of the Prox-1 mRNA sequence. Probes of at least 14 and more preferably 18 nucleotides are prefened to assure specificity. One technique for measuring Prox-1 mRNA comprises in situ hybridization to measure Prox-1 mRNA in the colon sample. Other techniques involve steps of isolating mRNA from the colon tissue and measuring Prox-1 mRNA in the isolated mRNA, for example, by Northern hybridization procedures. In still another variation, quantitative reverse transcriptase polymerase chain reaction (PCR), real-time PCR, or other PCR techniques are employed to quantitatively amplify Prox- 1 mRNA (relative to control samples) to provide a quantitative measurement of Prox- 1 mRNA in the colon tissue. In yet another embodiment, Prox-1 expression is measured indirectly by measuring a functional property of Prox-1, such as measuring Prox-1 binding to DNA or downstream Prox-1 transcription factor effects. The screening method further includes a comparing step whereby
Prox-1 expression in the colon tissue is compared to Prox-1 expression in healthy colon tissue, wherein increased Prox-1 expression in the colon tissue coreelates with a pathological phenotype. As described herein, Prox-1 expression is elevated in a statistically significant manner in pathological specimens studied, compared to healthy colon tissue samples. In one variation, the comparison is performed by taking simultaneous or sequential measurements of a test sample and a sample of colon tissue that is known to be taken from healthy tissue. In another variation, data is accumulated on the quantity of Prox-1 mRNA or protein in healthy tissues, and the amount that is measured in the colon tissue from the biological sample is compared to this predetermined amount. It will be appreciated that comparing Prox-1 measurements from a test sample to measurements from a cancerous or precancerous condition can provide an equivalent indication of the presence or absence of the pathological condition, wherein a test sample with Prox-1 expression comparable to the elevated level observed in a cancer correlates with a pathological phenotype. For measurement comparisons, a database of Prox-1 measurements from colon tissues can be developed, preferably containing information about healthiness or disease of the tissue; age, sex, race/ethnicity of the donor, and location from which the sample was taken. With a database of samples, comparisons can be analyzed using statistical analysis to determine the statistical significance of a measurement's deviation from a mean, optionally selecting entries from the database by selecting for the patient's age, sex, ethnicity, and other factors to best match the patient (mammalian subject) being tested. Such statistical analysis permits establishment of one or more "cutoff" values for the Prox-1 measurement that are conelated with a likelihood of having, or developing, a cancerous condition. If elevated Prox-1 is detected, then in a prefened embodiment, the method further comprises a step of administering to a human subject identified as having a pathological condition characterized by increased Prox-1 expression in colon tissue a composition comprising a Prox-1 inhibitor. . In a related embodiment, the invention provides a method of inliibiting the growth of colon cancer cells, such as colon carcinoma cells, colon adenoma cells, or colon adenocarcinoma cells in a mammalian subject comprising a step of: administering to the subject a composition comprising a molecule that suppresses expression of Prox-1, thereby inhibiting the growth of colon carcinoma cells. For reasons of cost, safety, and efficacy, it is becoming increasingly prefened to attempt to identify patients most likely to benefit from a therapeutic regimen before administering it. This is especially true with cancers where it is known that not all patients respond the same to all therapies. Thus, in a prefened variation of the method, steps are taken to identify patients most likely to benefit from this regimen. For example, the method further comprises a step of identifying a mammalian subject with a colon cancer characterized by increased Prox-1 expression. The composition is administered to such a patient after the identifying step, because cancers characterized by the elevated expression are expected to be the cancers most likely to respond to the inhibitors. Exemplary cancers (neoplasms) in which Prox-1 elevation has been observed include colorectal adenomas and colorectal carcinomas, as described below in greater detail. The composition to be administered preferably includes, in addition to the Prox-1 inhibitor, a pharmaceutically acceptable diluent, adjuvant, or carrier medium. The composition optionally includes additional antineoplastic agents. Administration of any Prox-1 inhibitors, alone or in combination, is contemplated for this invention, either alone or in combination with other Prox-1 inhibitors or other antineoplastic agents. Exemplary inhibitor molecules include antisense oligonucleotides that inhibit Prox-1 expression; micro-RNA that inhibits Prox-1 expression; small (short) interfering RNA (siRNA) that inhibit Prox-1 expression (e.g., siRNA that comprise at least one nucleotide sequence set forth in SEQ ID NOS: 4, 5, 6, and 7); zinc finger proteins that inhibit Prox-1 expression; polypeptides that act as dominant negative form of Prox-1 protein, such as Prox-1 forms that have a disrupted DNA binding domain or transactivation domain(s); polynucleotides that encode dominant-negative Prox-1 proteins; Prox-1 antibodies and fragments thereof; polynucleotides that encode Prox-1 antibodies or encode " polypeptides that comprise Prox-1 binding domains; small molecules discovered and- designed through screening based on the teachings herein, and so on. U.S. Patent Application Publication No. 2003/0224516 discloses exemplary molecules for inhibiting Prox-1 expression and is incorporated herein by reference. The inhibitor is preferably administered in an amount and in a regimen that halts or inhibits neoplastic growth of the affected colorectal tissue. As another benchmark, the tissue itself preferably reverts to a non-transformed, more healthy looking phenotype. As described herein, one apparent benchmark of beneficial administration is an increase in Notch-1 signaling. Thus, in one variation, the composition is administered in an amount effective to suppress Prox-1 expression and increase Notch 1 signaling. Other indications of efficacy relate to modulation of prostaglandin synthesis. Thus, in another variation, the composition is administered in an amount effective to increase 15-PDGH activity or decrease prostaglandin D2 synthase activity. As described herein and in literature, colorectal cancers also are often characterized by increases in the -catenin TCF signaling pathway, relative to what is observable in healthy colorectal tissue. Thus, in a prefened variation, in addition to administering a Prox-1 inhibitor composition, the regimen further comprises administering to the subject an inhibitor of the -catenin/TCF signaling pathway. (Optionally, the patient's diseased tissue is first pre-screened for elevated expression/signaling of this pathway.) The categories of inhibitors described above for Prox-1 are specifically contemplated for the -catenin/TCF pathway as well. In one variation, the inhibitor of the -catenin/TCF signaling pathway is dominant negative form of TCF-4. The inhibitor optionally targets (inhibits) TCF-4, β-catenin, or c-myc expression or activity. In yet another variation, administration of the Prox-1 inhibitor is combined with administration of a COX-2 inhibitor, such as any of the increasing class of non-steroidal anti-inflammatory agents. In still another variation, administration of the Prox-1 inhibitor is combined with administration of a Notch signaling pathway agonist, such as a Notch . ligand or expression vector to cause expression of a Notch ligand. Exemplary Notch ligands include Jaggedl, Jagged2, Deltal, Delta3, Delta4, or Senate. Also contemplated is administration of a molecule that comprises an inhibitor of DNA methyltransferases. Such inhibitors are themselves contemplated as efficacious for inhibiting Prox-1 expression, and can be combined with any other Prox-1 inhibitor described herein for combination therapy. An exemplary methyltransferase inhibitor is 5-aza-2'-deoxycytidine. In still another variation, the Prox-1 inhibitor composition is admimstered in combination with any known antineoplastic agent that is used in cancer therapy. In still another variation, the Prox-1 inhibitor and/or Cox-2 inhibitor are combined (in a medicament or as a combination therapy) with an agent that induces differentiation in colorectal cancer cell lines. Exemplary agents include 1,25- dihydroxyvitamin D3 and analogs thereof; butyrate; and retinoids. With respect to any combination treatment or therapy regimens described herein, the Prox-1 inhibitor composition can be administered simultaneously with the other active agents, which may be in admixture with the Prox-1 inhibitor, or may be in a separate composition. Each composition preferably includes a pharmaceutically acceptable diluent, adjuvant, or carrier. When the agents are separately administered, they may be administered in any order. In still another embodiment, the invention includes a method of inhibiting Prox-1 function in a mammalian subject having a disease characterized by of Prox-1 over-expression in cells, comprising the step of administering to said mammalian subject a composition, said composition comprising a compound effective to inhibit Prox-1 function in cells. In still another variation, the invention includes the use of a Prox-1 inhibitor in the manufacture of a medicament for the treatment of a disease characterized by Prox-1 over-expression in cells, especially cancerous or precancerous cells of colorectal origin. The medicament optionally includes the additional agents described above, either in admixture with the Prox-1 inhibitor or separated, yet packaged together (preferably with instructions for treating the disease). In yet another embodiment, the invention provides a method of screening for Prox-1 modulators comprising the steps of: (a) contacting a test molecule with Prox-1 protein, or a nucleic acid comprising a nucleotide sequence that encodes Prox-1 protein, under conditions which permit the interaction of the test molecule with the Prox-1 protein or nucleic acid; and (b) measuring the interaction between the test molecule and Prox-1 protein or the nucleic acid, wherein a test molecule that binds the Prox-1 protein or nucleic acid is identified as a Prox-1 modulator. "Test molecule" refers to the molecule that is under evaluation for the ability to modulate (i.e., increase or decrease) the activity of Prox-1 protein. Most commonly, a test molecule that is a Prox-1 modulator will interact directly with Prox- 1. However, the screens described herein can identify test molecules that modulate Prox-1 protein activity indirectly, such as by affecting Prox-1 gene expression. The screens work with essentially any test molecule, and the invention is not Umited in this manner. In prefened embodiments, the test molecule is a protein, a carbohydrate, a lipid, or a nucleic acid. Molecules which regulate Prox-1 expression include nucleic acids which are complementary to nucleic acids encoding a Prox-1 protein, or are complementary to nucleic acid sequences which direct or control the expression of Prox-1 protein, and which act as anti-sense regulators of expression. The test molecule may be a member of a chemical library, such as libraries commonly maintained in large pharmaceutical companies or libraries generated combinatorially. In alternate embodiments, the test molecule interacts with Prox-1 by binding to the Prox-1 DNA binding domain, thereby effecting Prox-1 activity. With respect to the screening methods described herein, it may be desirable to evaluate two or more test compounds together for their ability to increase or decrease the Prox-1 protein activity or expression. The assays set forth herein can be readily modified by adding such additional test compounds either simultaneous with, or subsequent to, or prior to, the first test compound. Lh additional embodiments, the measurement of the interaction of test molecules with Prox-1 may is carried out using solution-phase assays or immunoassays. In other embodiments, measurement of the interaction of test molecules with Prox- 1 is carried out by evaluating biological activity of Prox- 1. In a related embodiment, the invention provides a method of screening for modulators of binding between a DNA and Prox-1 protein comprising steps of: (a) contacting a DNA with a Prox-1 protein in the presence and in the absence of a putative modulator compound; (b) detecting binding between the DNA and the Prox-1 protein in the presence and absence of the putative modulator compound; and (c) identifying a modulator compound based on a decrease or increase in binding between the DNA and the Prox-1 protein in the presence of the putative modulator compound, as compared to binding in the absence of the putative modulator compound. Lh a related variation, molecules that modulate binding between DNA and Prox-1 are formulated into a composition or a growth media for contacting a cell from a colorectal cancer or colorectal cancer cell Une, and a modulator that inhibits growth of the cell is selected as a prefened modulator for development as a therapeutic. In yet another related embodiment, the invention provides a method of screening for modulators of binding between a DNA and Prox-1 protein comprising steps of: (a) contacting a DNA with a Prox-1 protein in the presence and in the absence of a putative modulator compound; (b) detecting binding between the DNA and the Prox-1 protein in the presence and absence of the putative modulator compound; and (c) identifying a modulator compound based on a decrease or increase in differentiation in the presence of the putative modulator compound, as compared to differentiation in the absence of the putative modulator compound. In vivo screening also is contemplated, either in addition to or in place of in vitro screening. The test compound preferably is formulated into a pharmaceutically acceptable diluent, adjuvant, or earner. In a prefened variation, this formulation is administered to a mammal with pathological (e.g., cancerous) Prox-1 expressing colon tissue, and the efficacy of the formulation at inhibiting disease progression is monitored. For example, a method described above optionally further comprises steps of formulating a composition comprising the selected Prox-1 modulator and a pharmaceutically acceptable carrier; administering the composition to a mammalian subject having a colorectal cancer; and monitoring the m-immalian subject for growth, metastasis, shrinkage, or disappearance of the colorectal cancer. "Putative modulator compounds" are analogous to the "test molecules" described above in that they are alleged to have an effect on Prox-1 protein activity "and are being identified as such using the methods described herein. In certain embodiments detecting DNA binding to Prox-1 protein and identifying an increase or decrease of DNA binding to Prox-1 protein employs immuno-based assays or various other assays that measure biological activity. Likewise, embodied by the invention are methods wherein identifying a modulator compound the use of proliferation and/or differentiation assays. In still another variation of the invention, provided are short interfering RNA (siRNA) molecules that down regulate expression of Prox-1 by RNA interference. The siRNA molecule can be adapted for use to treat colorectal cancer and any other indications that respond to the level of Prox-1. The siRNA molecule comprises a sense region and an antisense region. The antisense region comprises sequence complementary to an RNA sequence encoding Prox-1, or a fragment thereof, and the sense region comprise sequence complementary to the antisense region. In additional embodiments, the siRNA molecule can comprise two nucleic acid fragments, wherein one fragment comprises the sense region and the second fragment comprises the antisense region of said siRNA molecule. In one embodiment, a siRNA molecule of the invention can comprise any contiguous Prox-1 sequence. Preferably, the siRNA constructs are between 18 and 100 nucleotides in length. More preferably, the siRNA constructs are 21 nucleotides in length. In still another embodiment, the sense region of a siRNA molecule of the invention comprises a 3 '-terminal overhang and the antisense region comprises a 3'- terminal overhang. The 3 '-terminal overhangs each are preferably from 1 to 5 nucleo tides. More preferably, the 3 '-terminal overhangs are 2 nucleotides. In a prefened embodiment, the antisense region of the 3 '-terminal nucleotide overhang is complementary to RNA encoding Prox-1. With respect to the antisense region of the siRNA constructs, the antisense region of Prox-1 siRNA constructs can comprise a sequence complementary to sequence having any of SEQ ID NOs. 4 and 6. Further, the antisense region of Prox-1 siRNA constructs can comprise a having any of SEQ ID NOs. 5 and 7. - In yet an additional embodiment of the invention, compounds, particularly antisense oligonucleotides, which are targeted to a nucleic acid encoding Prox-1, and which modulate the expression of Prox-1 are provided. The antisense oligonucleotides of the invention are preferably complementary to (at least a segment of) the genomic Prox-1 sequence set forth as SEQ ID NO:l. mRNA splice sites, i.e., intron-exon junctions, may be prefened target regions. Accordingly, in another embodiment, the antisense oligonucleotides of the invention comprise a region complementary to a promoter or other control region, an exon, an intron, or an exon- intron boundary. Also embodied by the present invention are antisense oligonucleotides that are complementary to a region within 20-200 bases of an exon- intron splice junction. As detailed herein, pharmaceutical compositions comprising antisense oligonucleotides are also provided. The foregoing paragraphs are not intended to define every aspect of the invention, and additional aspects are described in other sections, such as the Detailed Description. The entire document is intended to be related as a unified disclosure, and it should be understood that all combinations of features described herein are contemplated, even if the combination of features are not found together in the same sentence, or paragraph, or section of this document. Where protein therapy is described, embodiments involving polynucleotide therapy (using polynucleotides that encode the protein) are specifically contemplated, and the reverse also is true. In addition to the foregoing, the invention includes, as an additional aspect, all embodiments of the invention naπower in scope in any way than the variations defined by specific paragraphs above. For example, certain aspects of the invention that are described as a genus, and it should be understood that every member of a genus is, individually, an aspect of the invention. Although the applicant(s) invented the full scope of the invention described herein, the applicants do not intend to claim subject matter described in the prior art work of others. Therefore, in the event that statutory prior art within the scope of a claim is brought to the attention of the applicants by a Patent Office or other entity or individual, the applicant(s) reserve the right to exercise amendment rights under applicable patent laws to redefine the subject matter of such a claim to specifically exclude such statutory prior art or obvious variations of statutory prior art from the scope of such a claim. Variations of the invention defined by such amended claims also are intended as aspects of the invention.
BRIEF DESCRIPTION OF THE DRAWINGS Figures IA, IB, and IC depict the elevated Prox-1 mRNA levels in colorectal tumors. A cancer RNA profiling array was hybridized to probes for Prox-1 (Fig. IA) and the lymphatic endothelial marker LYVE-l (Fig. IB). Fig. IC illustrates the quantification of dot blot in Fig. 1 A, the asterisk indicating tumor samples in which Prox-1 expression is significantly different from that of the normal tissue (P<0.005). Figures 2A-2I depict Prox-1 expression patterns in colon cancer and normal colonic epithelium. Frozen sections of colon adenomas (Fig. 2A-C) or adenocarcinomas (Fig. 2D-F) and the conesponding normal tissues (Fig. 2H-1) were stained for Prox-1. Fig. 2C and Fig. 21 show high power magnification of adenoma and normal colon sections. Figure 3 depicts the efficacy of Prox-1 suppression for inhibiting SW480R cell growth in soft agar. SW480R cells were transfected with GFP siPRNA, Prox-1 siRNA A16 or Prox-1 siRNA A25 or left untreated, and seeded in soft agar in triplicate. The number of colonies was scored after two weeks of growth. DETAILED DESCRIPTION Demonstrated herein for the first time is the importance of Prox-1 in cancer. The Prox-1 gene and protein is overexpressed in colorectal cancers, as compared to healthy colon tissue and other cancer tissues. Prox-1 was overexpressed in 68% of colorectal carcinomas and in 80% of premalignant lesions that were examined, indicating that Prox-1 is important for tumorigenesis, and therefore a useful marker for screening and a useful target for intervention. In normal colonic epithelium, Prox-1 expression was restricted to two cell types, neuroendocrine cells and non-proliferating cells at the very base of the colonic crypts, a location that conesponds to the stem cell compartment. Contemplated and provided for in the present invention are polynucleotides and polypeptides for screening and diagnosis of colorectal cancer and/or premalignancies. Intervention to suppress Prox-1 expression in colorectal cells resulted in increased activation of Notch signal transduction. Specifically, ablation of Prox-1 resulted in cell growth arrest and increased expression of epithelial markers. This was accompanied by an upregulation of the cell cycle inhibitor p21cipl , which has been shown to be important for the differentiation of intestinal epithelia (Quaroni et al., Am. J. Physiol. Cell Physiol. 279: C1045-57, 2000; Yang et ab, Cancer Res. 61, 565- 9, 2001), and by an increased expression of components of the Notch signaling pathway. Unexpectedly, this phenotype persisted for up to two weeks after transient transfection with Prox-1 siRNAs, demonstrating profound changes in the transcriptional program induced in the absence of Prox-1. Without intending to be Umited to a particular theory or mechanism, Prox-1 may be involved in the maintenance of an undifferentiated state of colonic intestinal stem cells, and overexpression of Prox-1 in cancer cells and resulting inhibition of the Notch signaling pathway may lead to the de-differentiation frequently observed upon malignant transformation. The suppression of Prox-1 expression also negatively regulates prostaglandin activity in the tumor cell lines studied. It is, therefore, contemplated that suppression of Prox-1 or activation of Notch signaling in tumor cells can provide a differentiation therapy for colon carcinoma. The present invention, more specifically, provides compositions and methods for suppressing Prox-1 expression. A. Inhibitory Nucleic Acid Constructs for the Suppression of Prox-1 Expression As discussed herein, Prox-1 is overexpressed in colorectal cancer cells and suppression of Prox-1 expression results in increased Notch signal transduction and modified expression of enzymes of the prostaglandin biosynthetic pathway. This data provides an indication to disrupt the expression or activity of Prox-1 as a method of alleviating the symptoms of and/or inhibiting the growth or metastasis of colon cancer. Such disruption is achieved using any materials or methods available to - - inhibit Prox-1 mRNA or protein expression, or inhibit Prox-1 binding, and any Prox-1 activity. The present section discusses nucleic acid-based methods of disrupting the expression of Prox-1. Polynucleotide products which are useful in this endeavor include antisense polynucleotides, ribozymes, small interfering RNAs, natural or designed microRNAs, triple helix polynucleotides, and novel transcription factors that modulate the expression of Prox-1 protein. Techniques for making and delivering antisense polynucleotides and ribozymes are well known to those in the art and have been extensively described in scientific, patent, and trade literature. (PCT Publication No. WO 00/32765; (JBiol Chem ;272:626-38. 1997); Kuneck et αb, (Nucleic Acids Res. ;30:1911-8. 2002); Crooke and B. Lebleu, eds. Antisense Research and Applications (1993) CRC Press; and Antisense RNA and DNA (1988) D. A. Melton, Ed. Cold Spring Harbor
Laboratory Cold Spring Harbor, N.Y.) Anti-sense RNA and DNA molecules act to directly block the translation of mRNA by binding to targeted mRNA and preventing protein translation. An example of an antisense polynucleotide is an oligodeoxyribonucleotide derived from the translation initiation site, e.g., between -10 and +10 regions of the relevant nucleotide sequence. Antisense oligonucleotides of 8- 200 nucleotides in length that include at least a portion of this region of the Prox-1 cDNA or genomic sequences set forth as SEQ ID NOs: 1 and 2 (or are complementary to) are prefened Prox-1 inhibitors of the invention. Antisense polynucleotides are typically generated within the cell by expression from antisense constructs that contain the antisense nucleic acid strand as the transcribed strand. Antisense methodology takes advantage of the fact that nucleic acids tend to pair with "complementary" sequences. By complementary, it is meant that polynucleotides are those which are capable of base-pairing according to the standard Watson-Crick complementarity rules. That is, the larger purines will base pair with the smaller pyrimidines to form combinations of guanine paired with cytosine (G:C) and adenine paired with either thymine (A:T) in the case of DNA, or adenine paired with uracil (A:U) in the case of RNA. Inclusion of less common bases such as inosine, 5-methylcytosine, 6-methyladenine, hypoxanthine and others in hybridizing sequences does not interfere with pairing. Targeting double-stranded (ds) DNA with polynucleotides leads to triple-helix formation; targeting RNA will lead to double-helix formation. Antisense polynucleotides, when introduced into a target cell, specifically bind to their target polynucleotide and interfere with transcription, RNA processing, transport, translation and/or stability. Antisense RNA constructs, or DNA encoding such antisense RNA's, may be employed to inhibit gene transcription or translation or both within a host cell, either in vitro or in vivo, such as within a host animal, including a human subject. Antisense constructs may be designed to bind to the promoter and other control regions, exons, introns or even exon-intron boundaries of a gene. Highly effective antisense constructs include regions complementary to intron/exon splice junctions. Thus, a prefened embodiment includes an antisense construct with complementarity to regions within 50-200 bases of an intron-exon splice junction. It has been observed that some exon sequences can be included in the construct without seriously affecting the target selectivity thereof. The amount of exonic material included will vary depending on the particular exon and intron sequences used. One can readily test whether too much exon DNA is included simply by testing the constructs in vitro to determine whether normal cellular function is affected or whether the expression of related genes having complementary sequences is affected. For purposes of making antisense oligonucleotides, polynucleotide sequences that are substantially complementary over their entire length and have zero or very few base mismatches are prefened. For example, sequences of fifteen bases in length preferably have complementary nucleotides at thirteen or fourteen or fifteen positions. Naturally, sequences which are completely complementary will be sequences which are entirely complementary throughout their entire length and have no base mismatches. Other sequences with lower degrees of homology also are contemplated. For example, an antisense construct which has limited regions of high homology, but also contains anon-homologous region (e.g., ribozymes) could be designed. These molecules, though having less than 50% homology, would bind to target sequences under appropriate conditions. Methods for designing and optimizing antisense nucleotides are described in Lima et al., (J Biol Chem; 272:626-38. 1997) and Kurreck et ab, (Nucleic Acids Res.; 30:1911-8. 2002). Additionally, commercial software and online resources are available to optimize antisense sequence selection and also to compare selected sequences to known genomic sequences to help ensure uniqueness/specificity for a chosen gene. (See, e.g., world wide web at sfold.wadsworth.org/index.pl.) Such uniqueness can be further confirmed by hybridization analyses. Antisense nucleic acids are introduced into cells (e.g., by a viral vector or colloidal dispersion system such as a liposome). The genomic contig of chromosome 1 (where Prox-1 is located), cDNA for Prox-1, and protein sequences for Prox-1 (SEQ ID NOs: 1, 2, and 3, respectively) are published and disclosed as Genbank Accession Numbers NT_021877, NM_002763, and NM_002763, respectively. The Genbank Database is accessible on the world wide web at ncbi.nlm.nih.gov. Related Prox-1 protein and/or nucleic acid sequences from other sources may be identified using probes directed at these sequences. Such additional sequences may be useful in certain aspects of the present invention. Although antisense sequences may be full length genomic or cDNA copies, they also may be shorter fragments or oligonucleotides e.g., polynucleotides of 100 or less bases. Although shorter oligomers (8-20) are easier to make and more easily permeable in vivo, other factors also are involved in determining the specificity of base pairing. For example, the binding affinity and sequence specificity of an oligonucleotide to its complementary target increases with increasing length. It is contemplated that oligonucleotides of 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, or more base pairs will be used. Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. The cleavage event renders the mRNA unstable and prevents protein expression. The mechanism of ribozyme action involves sequence specific interaction of the ribozyme molecule to complementary target RNA, followed by an endonucleolytic cleavage. Within the scope of the invention are engineered hammerhead, for which the substrate sequence requirements are minimal, or other motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of RNA sequences encoding protein complex components. Design of the hammerhead ribozyme and the therapeutic uses of ribozymes are disclosed in Usman et al., Cunent Opin. Struct. Biol. (1996) 6:527-533. Ribozymes can also be prepared and used as described in Long et al., FASEB J. (1993) 7:25; Symons, Ann. Rev. Biochem. (1992) 61 :641 ; Peπotta et al., Biochem. (1992) 31:16-17; Ojwang et al., Proc. Natl. Acad. Sci. (USA) (1992) 89:10802-10806; and U.S. Pat. No. 5,254,678. Methods of cleaving RNA using ribozymes is described in U.S. Pat. No. 5,116,742; and methods for increasing the specificity of ribozymes are described in U.S. Pat. No. 5,225,337 and Koizumi et ab, Nucleic Acid Res. (1989) 17:7059-7071. Preparation and use of ribozyme fragments in a hairpin structure are described by Chowrira and Burke, Nucleic Acids Res. (1992) 20:2835. Ribozymes can also be made by rolling transcription (Daubendiek and Kool, Nat. Biotechnol. (1997) 15(3):273-277). The full-length gene need not be known in order to design and use specific inhibitory ribozymes. Specific ribozyme cleavage sites within any potential * RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites which include the following sequences, GUA, GUU, and GUC. Once identified, short RNA sequences of between 15 and 20 ribonucleotides conesponding to the region of the target gene containing the cleavage site may be evaluated for predicted structural features, such as secondary structure, that may render the oligonucleotide sequence unsuitable. The suitability of candidate targets may also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using ribonuclease protection assays (Draper PCT WO 93/23569; and U.S. Pat. No. 5,093,246, incorporated herein by reference). Using the nucleic acid sequences disclosed herein and methods known in the art, ribozymes can be designed to specifically bind and cut the corresponding mRNA species. Ribozymes, . therefore, provide a means to inhibit the expression Prox-1. Alternatively, endogenous gene expression can be reduced by inactivating or "knocking out" the gene or its promoter using targeted homologous recombination. (E.g., see Smithies et al., 1985, Nature 317:230-234; Thomas & Capecchi, 1987, Cell 51:503-512; Thompson et ab, 1989 Cell 5:313-321). For example, a mutant, non-functional gene (or a completely unrelated DNA sequence) flanked by DNA homologous to the endogenous gene (either the coding regions or regulatory regions of the gene) can be used to transfect cells that express that gene in vivo. Insertion of the DNA construct, via targeted homologous recombination, results in inactivation of the gene. Gene expression can also be reduced by targeting deoxyribonucleoti.de sequences complementary to the regulatory region of the target gene (i.e., the gene promoter and/or enhancers) to form triple helical structures that prevent transcription of the gene in target cells in the body. (See generally, Helene, C. 1991, Anticancer Drug Des., 6(6):569-84; Helene, C, et al., 1992, Ann, N.Y. Acad. Sci., 660:27-36; and Maher, L. J., 1992, Bioassays 14(12):807-15). Nucleic acid molecules used in triple helix formation for the inhibition of transcription are generally single stranded deoxyribonucleotides. The base composition must be designed to promote triple helix formation via Hoogsteen base pairing rules, which generally require sizeable stretches of either purines or pyrimidines to be present on one strand of a duplex. Nucleotide sequences may be pyrimidine-based, which will result in TAT and CGC+ triplets across the three associated strands of the resulting triple helix. The pyrimidine-rich molecules provide base complementarity to a purine-rich region of a single strand of the duplex in a parallel orientation to that strand. In addition, nucleic acid molecules may be chosen that are purine-rich, for example, containing a stretch of G residues. These molecules will form a triple helix with a DNA duplex that is rich in GC pairs, in which the majority of the purine residues are located on a single strand of the targeted duplex, resulting in GGC triplets across the three strands in the triplex. Alternatively, the potential sequences that can be targeted for triple helix formation may be increased by creating a so called "switchback" nucleic acid molecule. Switchback molecules are synthesized in an alternating 5'-3', 3'-5' manner, such that they base pair with first one strand of a duplex and then the other, eliminating the necessity for a sizeable stretch of either purines or pyrimidines to be present on one strand of a duplex. Another technique for inhibiting the expression of a gene involves the use of RNA for induction of RNA interference (RNAi), using double stranded (dsRNA) (Fire et al, Nature 391: 806-811. 1998) or small interfering RNA (siRNA) sequences (Elbashir et al, Nature 411, 494 - 498 (2001)); Yu et al, Proc Natl Acad Sci U SA. 99:6047-52 (2002). "RNAi" is the process by which dsRNA induces homology-dependent degradation of complimentary mRNA. The presence of dsRNA in cells triggers the RNAi response though a mechanism that has yet to be fully characterized. Lh one embodiment, a synthetic antisense nucleic acid molecule is hybridized by complementary base pairing with a "sense" ribonucleic acid to form a double stranded RNA. The presence of long dsRNAs in cells stimulates the activity of a ribonuclease III enzyme. The dsRNA antisense and sense nucleic acid molecules are provided that conespond to at least about 20, 25, 50, 100, 250 or 500 nucleotides or an entire Prox-1 coding strand, or to only a portion thereof. In an alternative embodiment, the siRNAs are 30 nucleotides or less in length, and more preferably 21- to 23-nucleotides, with characteristic 2- to 3- nucleotide 3 '-overhanging ends, which are generated by ribonuclease in cleavage from longer dsRNAs. (See e.g. Tuschl T. Nat Biotechnol. 20:446-48. 2002). At notably higher concentrations single stranded 21 nucleotide RNA molecules have been also shown to function as siRNAs (i.e., enter the RNAi pathway and specifically target mRNA for degradation in mammalian cells (Martinez et ab, Cell 110, 563-574, 2002). Cleavage of the target RNA takes place in the middle of the region complementary to the antisense strand of the siRNA duplex (Elbashir et ab, 2001, Genes Dev., 15, 188). Intracellular transcription of small RNA molecules can be achieved by cloning the siRNA templates into RNA polymerase in (Pol DI) transcription units, which normally encode the small nuclear RNA (snRNA) U6 or the human RNAse P RNA HI. Two approaches can be used to express siRNAs: in one embodiment, sense and antisense strands constituting the siRNA duplex are transcribed using constructs with individual promoters (Lee, et al. Nat. Biotechnol. 20, 500-505. 2002); in an alternative embodiment, siRNAs are expressed as stem-loop hairpin RNA structures that give rise to siRNAs after intracellular processing (Brummelkamp et al. Science 296:550-553. 2002, herein incorporated by reference). Alternatively, a stem loop hairpin can be expressed within an umelated Pol IT transcribed mRNA transcript. A stem-loop hairpin designed to contain the siRNA sequence also contains conserved microRNA sequences within the loop and stem regions, thus resembling a natural precursor mRNA structure. Subsequently, the precursor can be processed by the cellular RNAi components to yield mature, functional siRNA/miRNA. (See, generally, Zeng et ab, Mol Cell 9, 1327-1333 (2002); Hutvagner et al., Science 297, 2056-2060 (2002); Kawasake et ab, Nature 423, 838-842 (2003)). RNAi has been studied in a variety of systems. Work in Drosophila embryonic lysates (Elbashir et al., 2001, EMBO J, 20, 6877) has revealed certain requirements for siRNA length, structure, chemical composition, and sequence that are essential to mediate efficient RNAi activity. Twenty-one nucleotide siRNA duplexes are most active when containing two nucleotide 3 '-overhangs. Replacing the 3'-overhanging segments of a 21-mer siRNA duplex having 2 nucleotide 3' overhangs with deoxyribonucleotides has no adverse effect on RNAi activity, while, replacing up to 4 nucleotides on each end of the siRNA with deoxyribonucleotides may be well tolerated. Complete substitution with deoxyribonucleotides results in no RNAi activity (Elbashir et ab, 2001, EMBO J., 20, 6877). Furthermore, complete substitution of one or both siRNA strands with
2'-deoxy (2'-H) or 2'-O-methyl nucleotides results in no RNAi activity, whereas substitution of the 3 '-terminal siRNA overhang nucleotides with deoxy nucleotides (2'-H) is tolerated. Single mismatch sequences in the center of the siRNA duplex may abolish RNAi activity. In addition, studies indicate that the position of the cleavage site in the target RNA is defined by the 5'-end of the siRNA guide sequence rather than the 3'-end (Elbashir et al., 2001, EMBO J, 20, 6877). Other studies indicate that a 5'-phosphate on the target-complementary strand of a siRNA duplex is required for siRNA activity and that ATP is utilized to maintain the 5'-phosphate moiety on the siRNA (Nykanen et ab, 2001, Cell, 107, 309). The dsRNA/siRNA is most commonly administered by annealing sense and antisense RNA strands in vitro before dehvery to the organism. In an alternate embodiment, RNAi may be carried out by administering sense and antisense nucleic acids of the invention in the same solution without annealing prior to administration, and may even be performed by administering the nucleic acids in separate vehicles within a very close timeframe. Genetic control can also be achieved through the design of novel transcription factors for modulating expression of the gene of interest in native cells and animals. For example, the Cys2-His2 zinc finger proteins, which bind DNA via their zinc finger domains, have been shown to be amenable to structural changes that lead to the recognition of different target sequences. These artificial zinc finger proteins recognize specific target sites with high affinity and low dissociation constants, and are able to act as gene switches to modulate gene expression. - Knowledge of the particular target sequence of the present invention facilitates the engineering of zinc finger proteins specific for the target sequence using known methods such as a combination of structure-based modeling and screening of phage display libraries (Segal et ab, (1999) Proc Natl Acad Sci USA 96:2758-2763; Liu et al., (1997) Proc Natl Acad Sci USA 94:5525-30; Greisman and Pabo (1997) Science 275:657-61; Choo et ab, (1997) J Mol Biol 273:525-32). Each zinc finger domain usually recognizes three or more base pairs. Since a recognition sequence of 18 base pairs is generally sufficient in length to render it unique in any known genome, a zinc finger protein consisting of 6 tandem repeats of zinc fingers would be expected to ensure specificity for a particular sequence (Segal et al., (1999) Proc Natl Acad Sci USA 96:2758-2763). The artificial zinc finger repeats, designed based on target sequences, are fused to activation or repression domains to promote or suppress gene expression (Liu et ab, (1997) Proc Natl Acad Sci USA 94:5525-30). Alternatively, the zinc finger domains can be fused to the TATA box-binding factor (TBP) with varying lengths of linker region between the zinc finger peptide and the TBP to create either transcriptional activators or repressors (Kim et al., (1997) Proc Natl Acad Sci USA 94:3616-3620). Such proteins, and polynucleotides that encode them, have utility for modulating expression in vivo in both native cells, animals and humans. The novel transcription factor can be delivered to the target cells by transfecting constructs that express the transcription factor (gene therapy), or by introducing the protein. Engineered zinc finger proteins can also be designed to bind RNA sequences for use in therapeutics as alternatives to antisense or catalytic RNA methods (McColl et al, (1999) Proc Natl Acad Sci USA 96:9521-6; Wu et ab, (1995) Proc Natl Acad Sci USA 92:344-348). Inactivation of Prox-1 function can also be accomplished using an overexpressed dominant negative form of Prox-1. As used herein a "dominant negative protein" is a mutant form of a protein which has the property of inhibiting the function of the endogenous, wild type form of the protein which conesponds to the mutant protein. Typically, dominant negative proteins have amino acid substitutions or are truncated forms of the wild type protein. The mutation may be in a substrate-binding domain (or DNA binding domain), a catalytic domain, or a cellular localization domain. For instance, a dominant negative form of Prox-1 may include a mutant truncated with respect to the DNA binding domain or transactivation domain. Disruption of the DNA binding domain entails truncation of the protein to exclude amino acids 572-634 of SEQ ID NO. 3, based on homology to Prospero (Drosophila). Disruption of the transactivation domain entails the deletion of amino acids 635-737. Other dominant negatives may include truncated forms of Prox-1 lacking the last 60 amino acids or the first 575 amino acids. Preferably, the mutant polypeptide will be overproduced. Point mutations can be made that have such an effect. In addition, fusion of different polypeptides of various lengths to the terminus of a protein can yield dominant negative mutants. General strategies for making dominant negative mutants are described in Herskowitz, Nature (1987) 329:219-222. Anti-sense RNA and DNA molecules, ribozymes, RNAi, triple helix polynucleotides, and novel transcription factors can be prepared by any method known in the art for the synthesis of DNA and RNA molecules. These include techniques for chemically synthesizing oligodeoxyribonucleotides well known in the art including, but not limited to, solid phase phosphoramidite chemical synthesis. Alternatively, RNA molecules may be generated by in vitro and in vivo transcription of DNA sequences encoding the antisense RNA molecule. Such DNA sequences may be incorporated into a wide variety of vectors which incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters. Alternatively, antisense cDNA constructs that synthesize antisense RNA constitutively or inducibly, depending on the promoter used, can be introduced stably or transiently into cells. B. Gene Therapy As described n detail in the preceding section, a variety of genetic manipulations to achieve modulation of Prox-1 protein expression or activity are contemplated. Additionally, where administration of proteins is contemplated, such as zinc finger proteins targeted to Prox-1, administration of a gene therapy vector to cause the protein of interest to be produced in vivo also is contemplated. Where inhibition of proteins is contemplated (e.g., through use of antibodies or small molecule inhibitors), inhibition of protein expression in vivo by genetic techniques, such as knock-out techniques or anti-sense therapy, is contemplated. It is now widely recognized that DNA may be introduced into a cell using a variety of viral vectors. Exemplary vectors that have been described in the literature include replication-deficient retroviral vectors, including but not limited to lentivirus vectors (Kim et al., J. Virol., 72(1): 811-816 (1998); Kingsman & Johnson, Scrip Magazine, October, 1998, pp. 43-46.); adenoviral (see, for example, U.S. Patent No. 5,824,544; U.S. Patent No. 5,707,618; U.S. Patent No. 5,792,453; U.S. Patent No. 5,693,509; U.S. Patent No. 5,670,488; U.S. Patent No. 5,585,362; Quantin et al., Proc. Natl. Acad. Sci. USA, 89: 2581-2584 (1992); Stratford-Perricadet et al., J. Clin. Invest, 90: 626-630 (1992); and Rosenfeld et ab, Cell, 68: 143-155 (1992)), retroviral (see, for example, U.S. Patent No. 5,888,502; U.S. Patent No. 5,830,725; U.S. Patent No. 5,770,414; U.S. Patent No. 5,686,278; U.S. Patent No. 4,861,719), adeno- associated viral (see, for example, U.S. Patent No. 5,474,935; U.S. Patent No. 5,139,941; U.S. Patent No. 5,622,856; U.S. Patent No. 5,658,776; U.S. Patent No. 5,773,289; U.S. Patent No. 5,789,390; U.S. Patent No. 5,834,441; U.S. Patent No. 5,863,541; U.S. Patent No. 5,851,521; U.S. Patent No. 5,252,479; Gnatenko et ab, J. Lhvestig. Med., 45: 87-98 (1997), an adenoviral-adenoassociated viral hybrid (see, for example, U.S. Patent No. 5,856,152) or a vaccinia viral or a herpesviral (see, for example, U.S. Patent No. 5,879,934; U.S. Patent No. 5,849,571; U.S. Patent No. 5,830,727; U.S. Patent No. 5,661,033; U.S. Patent No. 5,328,688); Lipofectin- mediated gene transfer (BRL); liposomal vectors (See, e.g., U.S. Patent No. 5,631,237 (Liposomes comprising Sendai virus proteins)) ; and combinations thereof. All of the foregoing documents are incorporated herein by reference in the entirety. Replication-deficient adenoviral vectors and adeno-associated viral vectors constitute prefened embodiments. In embodiments employing a viral vector, prefened polynucleotides include a suitable promoter and polyadenylation sequence to promote expression in the target tissue of interest. For many applications of the present invention, suitable promoters/enhancers for mammalian cell expression include, e.g., cytomegalovirus promoter/enhancer (Lehner et al., J. Clin. Microbiob, 29:2494-2502 (1991); Boshart et al., Cell, 41:521-530 (1985)); Rous sarcoma virus promoter (Davis et al., Hum. Gene Ther., 4: 151 (1993)); simian virus 40 promoter, long terminal repeat (LTR) of retroviruses, keratin 14 promoter, and myosin heavy chain promoter. In other embodiments, non-viral dehvery is contemplated. These include calcium phosphate precipitation (Graham and Van Der Eb, Virology, 52:456- 467 (1973); Chen and Okayama, Mol. Cell Biol, 7:2745-2752, (1987); Rippe, et al, Mol. Cell Biol, 10:689-695 (1990)), DEAE-dextran (Gopal, Mol. Cell Biol, 5:1188- 1190 (1985)), electroporation (Tur-Kaspa, et al, Mol. Cell Biol, 6:716-718, (1986); Potter, et al, Proc. Nat. Acad. Sci. USA, 81:7161-7165, (1984)), direct microinjection (Harland and Weintraub, J. Cell Biol, 101:1094-1099 (1985)), DNA-loaded liposomes (Nicolau and Sene, Biochim. Biophys. Acta, 721:185-190 (1982); Fraley, et al, Proc. Natl. Acad. Sci. USA, 76:3348-3352 (1979); Feigner, Sci. Am., 276(6):102-6 (1997); Feigner, Hum. Gene Ther., 7(15): 1791-3, (1996)), cell sonication (Fechheimer, et al, Proc. Natl. Acad. Sci. USA, 84:8463-8467 (1987)), gene bombardment using high velocity microprojectiles (Yang, et al, Proc. Natl. Acad. Sci. USA, 87:9568-9572 (1990)), and receptor-mediated transfection (Wu and Wu, J. Biol. Chem., 262:4429-4432 (1987); Wu and Wu, Biochemistry, 27:887-892 (1988); Wu and Wu, Adv. Drug Delivery Rev., 12:159-167 (1993)). In a particular embodiment of the invention, the expression construct (or indeed the peptides discussed above) may be entrapped in a liposome. Liposomes are vesicular structures characterized by a phospholipid bilayer membrane and an inner aqueous medium. Multilamellar liposomes have multiple lipid layers separated by aqueous medium. They form spontaneously when phospholipids are suspended in an excess of aqueous solution. The lipid components undergo self-reanangement before the formation of closed structures and entrap water and dissolved solutes between the lipid bilayers (Ghosh and Bachhawat, "In Liver Diseases, Targeted Diagnosis And Therapy Using Specific Receptors And Ligands," Wu, G., Wu, C, ed., New York: Marcel Dekker, pp. 87-104 (1991)). The addition of DNA to cationic liposomes causes a topological transition from liposomes to optically birefringent Uquid-crystalline condensed globules (Radler, et al, Science, 275(5301):810-4, (1997)). These DNA-lipid complexes are potential non-viral vectors for use in gene therapy and delivery. Liposome-mediated nucleic acid delivery and expression of foreign DNA in vitro has been very successful. Also contemplated in the present invention are various commercial approaches involving "lipofection" technology. In certain embodiments of the invention, the liposome may be complexed with a hemagglutinating virus (HVJ). This has been shown to facilitate fusion with the cell membrane and promote cell entry of liposome-encapsulated DNA (Kaneda, et al, Science, 243:375-378 (1989)). In other embodiments, the liposome may be complexed or employed in conjunction with nuclear nonhistone chromosomal proteins (HMG-1) (Kato, et al, J. Biol. Chem., 266:3361-3364 (1991)). hi yet further embodiments, the liposome may be complexed or employed in conjunction with both HVJ and HMG-1. In that such expression constructs have been successfully employed in transfer and expression of nucleic acid in vitro and in vivo, then they are applicable for the present invention. Other vector delivery systems that can be employed to deliver a nucleic acid encoding a therapeutic gene into cells include receptor-mediated delivery vehicles. These take advantage of the selective uptake of macromolecules by receptor-mediated endocytosis in almost all eukaryotic cells. Because of the cell type-specific distribution of various receptors, the delivery can be highly specific (Wu and Wu (1993), supra). Receptor-mediated gene targeting vehicles generally consist of two components: a cell receptor-specific ligand and a DNA-binding agent. Several ligands have been used for receptor-mediated gene transfer. The most extensively characterized ligands are asialoorosomucoid (ASOR) (Wu and Wu (1987), supra) and transferrin (Wagner, et al, Proc. Natl Acad Sci. USA, 87(9):3410-3414 (1990)). Recently, a synthetic neoglycoprotein, which recognizes the same receptor as ASOR, has been used as a gene dehvery vehicle (Ferkol, et al, FASEB J., 7:1081-1091 (1993); Perales, et al, Proc. Natl Acad. Sc , USA 91:4086-4090 (1994)) and epidermal growth factor (EGF) has also been used to deliver genes to squamous carcinoma cells (Myers, EPO 0273085). In other embodiments, the dehvery vehicle may comprise a ligand and a liposome. For example, Nicolau, et al, Methods Enzymol, 149:157-176 (1987) employed lactosyl-ceramide, a galactose-terminal asialganglioside, incorporated into liposomes and observed an increase in the uptake of the insulin gene by hepatocytes. Thus, it is feasible that a nucleic acid encoding a therapeutic gene also may be specifically delivered into a particular cell type by any number of receptor-ligand systems with or without liposomes. In another embodiment of the invention, the expression construct may simply consist of naked recombinant DNA or plasmids. Transfer of the construct may be performed by any of the methods mentioned above that physically or chemically permeabilize the cell membrane. This is applicable particularly for transfer in vitro, however, it may be applied for in vivo use as well. Dubensky, et al, Proc. Nat. Acad. Sci. USA, 81:7529-7533 (1984) successfully injected polyomavirus DNA in the form of CaPO precipitates into hver and spleen of adult and newborn mice demonstrating active viral replication and acute infection. Benvenisty and Neshif, Proc. Nat. Acad. Sci. USA, 83:9551-9555 (1986) also demonstrated that direct intraperitoneal injection of CaPO precipitated plasmids results in expression of the transfected genes. Another embodiment of the invention for transfening a naked DNA expression construct into cells may involve particle bombardment. This method depends on the ability to accelerate DNA coated microprojectiles to a high velocity allowing them to pierce cell membranes and enter cells without killing them (Klein, et al, Nature, 327:70-73 (1987)). Several devices for accelerating small particles have been developed. One such device relies on a high voltage discharge to generate an electrical cunent, which in turn provides the motive force (Yang, et al, Proc. Natl. Acad. Sci USA, 87:9568-9572 (1990)). The microprojectiles used have consisted of biologically inert substances such as tungsten or gold beads. Well-known techniques exist for gene delivery to in vivo and ex vivo situations. For viral vectors, one generally will prepare a viral vector stock. Depending on the type of virus and the titer attainable, one will deliver 1 x 104, 1 x 105, 1 x 106, 1 x 107, 1 x 10s, 1 x 109, 1 x 1010, 1 x 1011 or 1 x 1012 infectious particles to the patient. Similar figures may be extrapolated for liposomal or other non- viral formulations by comparing relative uptake efficiencies. Formulation as a pharmaceutically acceptable composition is discussed below. Various routes are contemplated for various tumor types. For practically any tumor, systemic delivery is contemplated. This will prove especially important for attacking microscopic or metastatic cancer. Where discrete tumor mass may be identified, a variety of direct, local and regional approaches may be taken. For example, the tumor may be directly injected with the expression vector or protein. A tumor bed may be treated prior to, during or after resection. Following resection, one generally will deliver the vector by a catheter left in place following surgery. One may utilize the tumor vasculature to introduce the vector into the tumor by injecting a supporting vein or artery. A more distal blood supply route also may be utilized. In an ex vivo embodiment, cells from the patient are removed and maintained outside the body for at least some period of time. During this period, a therapy is delivered, after which the cells are reintroduced into the patient; preferably, any tumor cells in the sample have been killed.
C. Antibodies Immunoreactive with Prox-1 Protein In another aspect, the present invention contemplates an antibody that is immunoreactive with a Prox-1 protein molecule of the present invention, or any portion thereof. Such antibodies include, but are not limited to, polyclonal, monoclonal, chimeric, single chain, Fab fragments and fragments produced by a Fab expression library, bifunctional/bispecific antibodies, humanized antibodies, CDR grafted antibodies, human antibodies and antibodies which include portions of CDR sequences specific for Prox-1 protein. The antibodies are useful as diagnostic reagents for measuring Prox-1 expression in a biological sample (e.g., a biopsy of colon tissue), and are useful for binding to Prox-1 protein to inhibit Prox-1 activity where the antibodies are delivered into cells. Neutralizing antibodies, i.e., those which may suppress Prox-1 expression, are especially prefened for therapeutic embodiments. In a prefened embodiment, an antibody is a monoclonal antibody. The invention provides for a pharmaceutical composition comprising a therapeutically effective amount of an antibody directed against Prox-1 protein. The antibody may bind to and neutralize the apoptotic effects of the Prox-1 protein. The antibody may be formulated with a pharmaceutically acceptable adjuvant. Means for preparing and characterizing antibodies are well known in the art (see, e.g., Harlow and Lane, ANTIBODIES: A LABORATORY MANUAL, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1988). Briefly, a polyclonal antibody is prepared by immunizing an animal with an immunogen comprising a polypeptide of the present invention and collecting antisera from that immunized animal. A wide range of animal species can be used for the production of antisera. Typically an animal used for production of anti-antisera is a non-human animal including rabbits, mice, rats, hamsters, goat, sheep, pigs or horses. Because of the relatively large blood volume of rabbits, a rabbit is a prefened choice for production of polyclonal antibodies. Depending on the host species, various adjuvants may be used to increase immunological response. Such adjuvants include but are not limited to Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol. BCG (bacilli Calmette-Guerin) and Corynebacterium parvum are potentially useful human adjuvants. Antibodies, both polyclonal and monoclonal, specific for isoforms of antigen may be prepared using conventional immunization techniques, as will be generally known to those of skill in the art. As used herein, the term "specific for" is intended to mean that the variable regions of the antibodies recognize and bind Prox-1 protein and are capable of distinguishing Prox-1 protein from other antigens, for example other secreted proapoptotic factors. A composition containing antigenic epitopes of the compounds of the present invention can be used to immunize one or more experimental animals, such as a rabbit or mouse, which will then proceed to produce specific antibodies against the compounds of the present invention. Polyclonal antisera may be obtained, after allowing time for antibody generation, simply by bleeding the animal and preparing serum samples from the whole blood. Monoclonal antibodies to Prox-1 protein may be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include but are not Umited to the hybridoma technique originally described by Koehler and Milstein ( Nature 256: 495-497, 1975), the human B-cell hybridoma technique (Kosbor et al, Immunol Today 4:72, 1983 ; Cote et al, Proc Natl Acad Sci 80: 2026-2030, 1983) and the EBV-hybridoma technique (Cole et al, Monoclonal Antibodies and Cancer Therapy, Alan R Liss Inc, New York N.Y., pp 77-96, (1985). When the hybridoma technique is employed, myeloma cell lines may be used. Such cell lines suited for use in hybridoma-producing fusion procedures preferably are non-antibody-producing, have high fusion efficiency, and enzyme deficiencies that render them incapable of growing in certain selective media which support the growth of only the desired fused cells (hybridomas). For example, where the immunized animal is a mouse, one may use P3-X63/Ag8, P3-X63-Ag8.653, NSl/l.Ag 4 1, Sp210-Agl4, FO, NSO/U, MPC-11, MPC11-X45-GTG 1.7 and S194/5XX0 Bui; for rats, one may use R210.RCY3, Y3-Ag 1.2.3, IR983F and 4B210; and U-266, GM1500-GRG2, LICR-LON-HMy2 and UC729-6 are all useful in connection with cell fusions. It should be noted that the hybridomas and cell lines produced by such techniques for producing the monoclonal antibodies are contemplated to be novel compositions of the present invention. An exemplary method for producing monoclonal antibodies against Prox-1 is provided in Example 1. Those of skill in the art will appreciate that such a method may be modified using techniques well known to those of skill in the art and still produce antibodies within the scope of the present invention. In addition to the production of monoclonal antibodies, techniques developed for the production of "chimeric antibodies", the splicing of mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity can be used (Monison et al, Proc Nati Acad Sci 81: 6851-6855, 1984 ; Neuberger et al, Nature 312: 604-608, 1984; Takeda et al, Nature 314: 452-454; 1985). Alternatively, techniques described for the production of single chain antibodies (U.S. Pat. No. 4,946,778) can be adapted to produce Prox-1 protein- specific single chain antibodies. Antibodies may also be produced by inducing in vivo production in the lymphocyte population or by screening recombinant immunoglobulin libraries or panels of highly specific binding reagents as disclosed in Orlandi et al (Proc Natl Acad Sci 86: 3833-3837; 1989), and Winter G and Milstein C (Nature 349: 293-299, 1991). Fully human antibodies relate to antibody molecules in which essentially the entire sequences of both the light chain and the heavy chain, including the CDRs, arise from human genes. Such antibodies are termed "human antibodies/' or "fully human antibodies" herein. Human monoclonal antibodies can be prepared by the trioma technique; the human B-cell hybridoma technique (see Kozbor, et al, Immunol Today 4: 72 (1983)) and the EBV hybridoma technique to produce human monoclonal antibodies (see Cole, et al, 1985 In: MONOCLONAL ANTIBODIES AND
CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96). Human monoclonal antibodies may be utilized in the practice of the present invention and may be produced by using human hybridomas (see Cote, et al., Proc Natl Acad Sci USA 80: 2026-2030 (1983)) or by transforming human B-cells with Epstein Ban Virus in vitro (see Cole, et al., 1985 In: MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96). In addition, human antibodies can also be produced using additional techniques, including phage display libraries (Hoogenboom and Winter, J. Mol. Biol 227:381 (1991); Marks et al, J. Mol. Biol. 222:581 (1991)). Similarly, human antibodies can be made by introducing human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, which closely resembles that seen in humans in all respects, including gene reaπangement, assembly, and antibody repertoire. This approach is described, for example, in U.S. Patent Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; 5,661,016, and in Marks et al. (Bio/Technology 10, 779-783 (1992)); Lonberg et al. (Nature 368 856-859 (1994)); Morrison ( Nature 368:812-13 (1994)); Fishwild et al,( Nature Biotechnology 14, 845-51 (1996)); Neuberger (Nature Biotechnology 14:826 (1996)); and Lonberg and Huszar (Intern. Rev. Immunol. 13:65-93 (1995)). Human antibodies may additionally be produced using transgenic nonhuman animals which are modified so as to produce fully human antibodies rather than the animal's endogenous antibodies in response to challenge by an antigen. (See PCT publication WO94/02602). The endogenous genes encoding the heavy and tight immunoglobulin chains in the nonhuman host have been incapacitated, and active loci encoding human heavy and tight chain immunoglobuhns are inserted into the host's genome. The human genes are incorporated, for example, using yeast artificial chromosomes containing the requisite human DNA segments. An animal which provides all the desired modifications is then obtained as progeny by crossbreeding intermediate transgenic animals containing fewer than the full complement of the modifications. The prefened embodiment of such a nonhuman animal is a mouse, and is termed the Xenomouse™ as disclosed in PCT publications WO 96/33735 and WO 96/34096. This animal produces B cells which secrete fully human immunoglobuhns. The antibodies can be obtained directly from the animal after immunization with an immunogen of interest, as, for example, a preparation of a polyclonal antibody, or alternatively from immortalized B cells derived from the animal, such as hybridomas producing monoclonal antibodies. Additionally, the genes encoding the immunoglobuhns with human variable regions can be recovered and expressed to obtain the antibodies directly, or can be further modified to obtain analogs of antibodies such as, for example, single chain Fv molecules. An example of a method of producing a nonhuman host, exemplified as a mouse, lacking expression of an endogenous immunoglobulin heavy chain is disclosed in U.S. Patent No. 5,939,598. It can be obtained by a method including deleting the J segment genes from at least one endogenous heavy chain locus in an embryonic stem cell to prevent reanangement of the locus and to prevent formation of a transcript of a reaπanged immunoglobulin heavy chain locus, the deletion being effected by a targeting vector containing a gene encoding a selectable marker; and producing from the embryonic stem cell a transgenic mouse whose somatic and germ cells contain the gene encoding the selectable marker. Antibodies as described herein are useful in standard immunochemical procedures, such as ELISA, radioimmuno assays, and Western blot methods and in immunohistochemical procedures such as tissue staining, as well as in other procedures which may utihze antibodies specific to Prox-1 protein -related antigen epitopes. Additionally, it is proposed that monoclonal antibodies specific to the particular Prox-1 protein of different species may be utilized in other useful applications. In general, both polyclonal and monoclonal antibodies against Prox-1 protein may be used in a variety of embodiments. In certain aspects, the antibodies may be employed for therapeutic purposes in which the inhibition of Prox-1 protein activity is desired (e.g., to reduce apoptosis in neuronal cells). Antibodies may be used to block Prox-1 protein action. Antibodies of the present invention also may prove useful in diagnostic purposes in order, for example, to detect increases or decreases in Prox-1 protein in tissue samples including samples for sites of inflammation, or fluid samples including blood serum, plasma and exudate samples. Additional aspects will employ the antibodies of the present invention in antibody cloning protocols to obtain cDNAs or genes encoding other Prox-1 protein. They may also be used in inhibition studies to analyze the effects of Prox-1 related peptides in cells or animals. Anti- Prox-1 protein antibodies will also be useful in immunolocalization studies to analyze the distribution of Prox-1 protein during various cellular events, for example, to determine the cellular or tissue-specific distribution of Prox-1 protein polypeptides under different points in the cell cycle. A particularly useful application of such antibodies is in purifying native or recombinant Prox-1 protein, for example, using an antibody affinity column. The operation of all such immunological techniques will be known to those of skill in the art in light of the present disclosure.
D. Assaying for Other Modulators of Prox-1 Activity and/or Expression In some situations, it may be desirable to identify molecules that are modulators, i.e., agonists or antagonists, of the activity of Prox-1 protein. Natural or synthetic molecules that modulate Prox-1 protein may be identified using one or more screening assays, such as those described herein. Such molecules may be administered either in an ex vivo manner, or in an in vivo manner by injection, or by oral delivery, implantation device or the like. "Test molecule(s)" refers to the molecule(s) that is/are under evaluation for the ability to modulate (i.e., increase or decrease) the activity of Prox-1 protein. Most commonly, a molecule that modulates Prox-1 activity will interact directly with Prox-1. However, it is also contemplated that a molecule may also modulate Prox-1 protein activity indirectly, such as by affecting Prox-1 gene expression, or by binding to a Prox-1 binding partner. In one embodiment, a test molecule will bind to a Prox-1 protein with an affinity constant of at least about 10"6 M, preferably about 10"8 M, more preferably about 10"9 M, and even more preferably about 10"10 M. Methods for identifying compounds which interact with Prox-1 protein are encompassed by the present invention. In certain embodiments, a Prox-1 protein is incubated with a test molecule under conditions which permit the interaction of the test molecule with a Prox-1 protein, and the extent of the interaction can be measured. The test molecule(s) can be screened in a substantially purified form or in a crude mixture. In certain embodiments, a Prox-1 protein agonist or antagonist may be a protein, peptide, carbohydrate, lipid or small molecular weight molecule which interacts with Prox-1 to regulate its activity. Molecules which regulate Prox-1 expression include nucleic acids which are complementary to nucleic acids encoding a Prox-1 protein, or are complementary to nucleic acid sequences which direct or control the expression of Prox-1 protein, and which act as anti-sense regulators of expression. Once a set of test molecules has been identified as interacting with Prox-1 protein, the molecules may be further evaluated for their abihty to increase or decrease Prox-1 activity. The measurement of the interaction of test molecules with Prox-1 may be carried out in several formats, including solution-phase assays and immunoassays. In general, test molecules are incubated with Prox-1 for a specified period of time, and Prox-1 protein activity is determined by one or more assays for measuring biological activity. In the event that Prox-1 displays biological activity through an interaction with a binding partner, a variety of in vitro assays may be used to measure the binding of Prox-1 to the conesponding binding partner. These assays may be used to screen test molecules for their ability to increase or decrease the rate and/or the extent of binding of Prox-1 to its binding partner. In one assay, a Prox-1 polypeptide is immobilized in the wells of a microtiter plate. Radiolabeled Prox-1 binding partner and the test molecule(s) can then be added either one at a time (in either order) or simultaneously to the wells. After incubation, the wells can be washed and counted (using a scintillation counter) for radioactivity to determine the extent to which the binding partner bound to Prox-1 polypeptide. Typically, the , molecules will be tested over a range of concentrations, and a series of control wells lacking one or more elements of the test assays can be used for accuracy in the evaluation of the results. An alternative to this method involves reversing the "positions" of the proteins, i.e., immobilizing Prox-1 binding partner to the micrbfiter plate wells, incubating with the test molecule and radiolabeled Prox-1 polypeptide, and determining the extent of Prox-1 polypeptide binding. See, for example, chapter 18, Current Protocols in Molecular Biology, Ausubel et al, eds., John Wiley & Sons, New York, NY (1995). As an alternative to radiolabeling, Prox-1 protein or its binding partner may be conjugated to biotin and the presence of biotinylated protein can then be detected using streptavidin linked to an enzyme, such as horseradish peroxidase (HRP) or alkaline phosphatase (AP), that can be detected colorometrically or by fluorescent tagging of streptavidin. An antibody directed to Prox-1 or to a Prox-1 binding partner and conjugated to biotin may also be used and can be detected after incubation with enzyme-linked streptavidin linked to AP or HRP. A Prox-1 protein or Prox-1 binding partner can also be immobilized by attachment to agarose beads, acrylic beads or other types of such inert solid phase substrates. The substrate-protein complex can be placed in a solution containing the complementary protein and the test compound. After incubation the beads can be precipitated by centrifugation, and the amount of binding between Prox-1 protein and its binding partner can be assessed using the methods described herein. Alternatively, the substrate-protein complex can be immobilized in a column, and the test molecule and complementary protein are passed through the column. The formation of a complex between an Prox-1 protein and its binding partner can then be assessed using any of the techniques set forth herein, i.e., radiolabeling, antibody binding or the like. Another in vitro assay that is useful for identifying a test molecule which increases or decreases the formation of a complex between Prox-1 and a Prox-1 binding partner is a surface plasmon resonance detector system such as the BIAcore assay system (Pharmacia, Piscataway, NJ). The BIAcore system may be carried out using the manufacturer's protocol. This assay essentially involves the covalent binding of either Prox-1 or a Prox-1 binding partner to a dextran-coated sensor chip which is located in a detector. The test compound and the other complementary protein can then be injected, either simultaneously or sequentially, into the chamber containing the sensor chip. The amount of complementary protein that binds can be assessed based on the change in molecular mass which is physically associated with the dextran-coated side of the sensor chip; the change in molecular mass can be measured by the detector system. In some cases, it may be desirable to evaluate two or more test compounds together for their abihty to increase or decrease the formation of a complex between Prox-1 polypeptide and a Prox-1 binding partner. In these cases, the assays set forth herein can be readily modified by adding such additional test compound(s) either simultaneous with, or subsequent to, the first test compound. The remainder of the steps in the assay are as set forth herein. In vitro assays such as those described herein may be used advantageously to screen large numbers of compounds for effects on complex formation by Prox-1 polypeptide and a Prox-1 binding partner. The assays may be automated to screen compounds generated in phage display, synthetic peptide, and chemical synthesis libraries. Compounds which increase or decrease the formation of a complex between a Prox-1 polypeptide and a Prox-1 binding partner may also be screened in cell culture using cells and cell lines expressing either Prox-1 polypeptide or a Prox-1 binding partner. Cells and cell lines may be obtained from any mammal. The binding of a Prox-1 protein to cells expressing a Prox-1 binding partner at the surface is evaluated in the presence or absence of test molecules, and the extent of binding may be determined by, for example, flow cytometry using a biotinylated antibody to a Prox-1 binding partner. Cell culture assays can be used advantageously to further evaluate compounds that score positive in protein binding assays described herein. Cell cultures can also be used to screen the impact of a drug candidate.
For example, drug candidates may decrease or increase the expression of the Prox-1 like gene. In certain embodiments, the amount of Prox-1 protein that is produced may be measured after exposure of the cell culture to the drug candidate. In certain embodiments, one may detect the actual impact of the drug candidate on the cell culture. For example, the overexpression of a particular gene may have a particular impact on the cell culture. In such cases, one may test a drug candidate's ability to increase or decrease the expression of the gene or its ability to prevent or inhibit a particular impact on the cell culture. In other examples, the production of a particular metabolic product such as a fragment of a polypeptide may result in, or be associated with, a disease or pathological condition. In such cases, one may test a drug candidate's ability to decrease the production of such a metabolic product in a cell culture.
E. Internalizing Proteins The tat protein sequence (from HIV) can be used to internalize proteins into a cell. See e.g., Falwell et al, Proc. Natl. Acad. Sci. USA, 91:664-668 (1994). For example, an 11 amino acid sequence (YGRKKRRQRRR; SEQ ID NO: 46) of the HJN tat protein (termed the "protein transduction domain", or TAT PDT) has been described as mediating delivery across the cytoplasmic membrane and the nuclear membrane of a cell. See Schwarze et al, Science, 285:1569-1572 (1999); and Νagahara et al, Nature Medicine, 4:1449-1452 (1998). In these procedures, FITC- constructs are prepared which bind to cells as observed by fluorescence-activated cell sorting (FACS) analysis, and these constructs penetrate tissues after i.p. adminstration.- Next, tat-bgal fusion proteins are constructed. Cells treated with this construct demonstrate b-gal activity. Following injection, a number of tissues, including liver, kidney, lung, heart and brain tissue, have been found to demonstrate expression using these procedures. It is believed that these constructions underwent some degree of unfolding in order to enter the cell; as such, refolding may be required after entering the cell. It will thus be appreciated that the tat protein sequence may be used to internalize a desired protein or polypeptide into a cell. For example, using the tat protein sequence, Prox-1 antagonist (such as an anti-Prox-1 binding agent, small molecule, or antisense oligonucleotide) can be administered intracellularly to inhibit the activity of a Prox-1 molecule. See also, Strauss, E., Science, 285:1466-1467 (1999).
F. Rational Drug Design The goal of rational drug design is to produce structural analogs of biologically active polypeptides or compounds with which they interact (agonists, antagonists, inhibitors, peptidomimetics, binding partners, etc.). By creating such analogs, it is possible to fashion drugs which are more active or stable than the natural molecules, which have different susceptibility to alteration or which may affect the function of various other molecules. In one approach, one generates a three- , dimensional structure for Prox-1 protein or a fragment thereof. This is accomplished by x-ray crystallography, computer modeling or by a combination of both approaches. An alternative approach, "alanine scan," involves the random replacement of residues throughout molecule with alanine, and the resulting affect on function determined. It also is possible to isolate a specific antibody, selected by a functional assay, and then solve its crystal structure. In principle, this approach yields a pharmacore upon which subsequent drug design can be based. It is possible to bypass protein crystallography altogether by generating anti-idiotypic antibodies to a functional, pharmacologically active antibody. As a mirror image of a niirror image, the binding site of anti-idiotype would be expected to be an analog of the original antigen. The anti-idiotype could then be used to identify and isolate peptides from banks of chemically- or biologically-produced peptides. Selected peptides would then serve as the pharmacore. Anti-idiotypes may be generated using the methods described herein for producing antibodies, using an antibody as the antigen. Thus, one may design drugs which have activity as stimulators, inhibitors, agonists, antagonists of Prox-1 protein or molecules affected by Prox-1 protein function. Such rational drug design may start with lead compounds identified by the present invention. By virtue of the availability of cloned Prox-1 protein sequences, sufficient amounts of the related proteins can be produced to perform crystallographic studies. In addition, knowledge of the polypeptide sequences permits computer employed predictions of structure-function relationships. G. Therapeutic Methods As discussed herein, polynucleotides or modulators of Prox-1 (including inhibitors of Prox-1) are administered to treat cancer. Therapeutic compositions can be administered in therapeutically effective dosages alone or in combination with adjuvant cancer therapy such as surgery, chemotherapy, radiotherapy, thermotherapy, and laser therapy, and may provide a beneficial effect, e.g. reducing tumor size, slowing rate of tumor growth, inhibiting metastasis, or otherwise improving overall clinical condition, without necessarily eradicating the cancer. The composition can also be administered in therapeutically effective amounts as a portion of an anti-cancer cocktail. An anti-cancer cocktail is a mixture of the polypeptide or modulator of the invention with one or more anti-cancer drugs in addition to a pharmaceutically acceptable carrier for delivery. Any anti-cancer drugs can be used as a treatment in combination with the polypeptide or modulator of the invention, including: Actinomycin D, Antinoglutethimide, Asparaginase, Bleomycin, Busulfan, Carboplatin, Carmustine, Chlorambucib Cisplatin (cis-DDP), Cyclophosphamide, Cytarabine HCl (Cytosine arabinoside), Dacarbazine, Dactinomycin, Daunorubicin HCl, Doxorubicin HCl, Estramustine phosphate sodium, Etoposide (V16-213), Floxuridine, 5-Fluorouracil (5-Fu), Flutamide, Hydroxyurea (hydroxycarbamide), lfosfamide, Interferon Alpha-2a, Interferon Alpha-2b,
Leuprolide acetate (LHRH-releasing factor analog), Lomustine, Mechlorethamine HCl (nitrogen mustard), Melphalan, Mercaptopurine, Mesna, Methotrexate (MTX), Mitomycin, Mitoxantrone HCb Octreotide, Plicamycin, Procarbazine HCl, Streptozocin, Tamoxifen citrate, Thioguanine, Thiotepa, Vinblastine sulfate, Vincristine sulfate, Amsacrine, Azacitidine, Hexamethylmelamine, Interleukin-2, Mitoguazone, Pentostatin, Semustine, Teniposide, and Vindesine sulfate. In addition, therapeutic compositions of the invention may be used for prophylactic treatment of cancer. There are hereditary conditions and/or environmental situations (e.g. exposure to carcinogens) known in the art that predispose an individual to developing cancers. Under these circumstances, it may be beneficial to treat these individuals with therapeutically effective doses of compositions of the invention to reduce the risk of developing cancers. In vitro and in vivo models can be used to determine the effective doses of the compositions of the invention for cancer treatment. These in vitro models include proliferation and differentiation assays of cultured tumor cells, growth of cultured tumor cells in soft agar (see Freshney, (1987) Culture of Animal Cells: A Manual of Basic Technique, Wily-Liss, New York, NY Ch 18 and Ch 21), tumor systems in nude mice as described in Giovanella et ab, J. Nati. Can. Inst, 52: 921-30 (1974), mobility and invasive potential of tumor cells in Boyden Chamber assays as described in Pilkington et al., Anticancer Res., 17: 4107-9 (1997), and angiogenesis assays such as induction of vascularization of the chick chorioallantoic membrane or induction of vascular endothelial cell migration as described in Ribatta et al., Intl. J. Dev. Biol., 40: 1189-97 (1999) and Li et al., Clin. Exp. Metastasis, 17:423-9 (1999) respectively. Suitable tumor cells lines are available, e.g. from American Type Tissue Culture Collection catalogs, and/or are described below.
H. Pharmaceutical Compositions Purified nucleic acids, antisense molecules, purified protein, antibodies, antagonists, or inhibitors may all be used as pharmaceutical compositions. Delivery of specific molecules for therapeutic purposes in this invention is further described below. The active compositions of the present invention include classic pharmaceutical preparations. Administration of these compositions according to the present invention will be via any common route so long as the target tissue is available via that route. The pharmaceutical compositions may be introduced into the subject by any conventional method, e.g., by intravenous, intradermab intramusclar, intramammary, intraperitoneab intrathecal, intraocular, retrobulbar, intrapulmonary (e.g., term release); by oral, sublingual, nasal, anal, vaginal, or transdermal delivery, or by surgical implantation at a particular site, e.g., embedded under the splenic capsule, brain, or in the cornea. The treatment may consist of a single dose or a plurality of doses over a period of time. The active compounds may be prepared for administration as solutions of free base or pharmacologically acceptable salts in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions also can be prepared in glycerob liquid polyethylene glycols, and 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. 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, ethanob polyol (for example, glycerob propylene glycob and liquid polyethylene glycob and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can 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. Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the prefened methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile- filtered solution thereof. As used herein, "pharmaceutically acceptable caπier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents 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 also can be incorporated into the compositions. For oral administration the active compositions may be incorporated with excipients and used in the form of non-ingestible mouthwashes and dentifrices. A mouthwash may be prepared incorporating the active ingredient in the required amount in an appropriate solvent, such as a sodium borate solution (Dobell's Solution). Alternatively, the active ingredient may be incorporated into an antiseptic wash containing sodium borate, glycerin and potassium bicarbonate. The active ingredient may also be dispersed in dentifrices, including: gels, pastes, powders and slurries. The active ingredient may be added in a therapeutically effective amount to a paste dentifrice that may include water, binders, abrasives, flavoring agents, foaming agents, and humectants. The compositions of the present invention may be formulated in a neutral or salt form. Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups also can be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. The compositions of the present invention may be formulated in a neutral or salt form. Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandehc, and the like. Salts formed with the free carboxyl groups also can be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations are easily administered in a variety of dosage forms such as injectable solutions, drug release capsules and the like. 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 and intraperitoneal administration. In the clinical setting an "effective amount" is an amount sufficient to effect beneficial or desired clinical results. An effective amount can be administered in one or more doses. In terms of treatment, an "effective amount" of polynucleotide, and/or polypeptide is an amount that results in amelioration of symptoms or a prolongation of survival in a patient. The effective amount is generally determined by the physician on a case-by-case basis and is within the skill of one in the art. Several factors are typically taken into account when determining, an appropriate dosage. These factors include age, sex and weight of the patient, the condition being treated, the severity of the condition and the form of the antibody being administered. For instance, in embodiments in which the antibody compositions of the present invention are being therapeutically administered, it is likely the concentration of a single chain antibody need not be as high as that of native antibodies in order to be therapeutically effective. Determination of the effective amounts is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. For example, a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture (i.e., the concentration of the test compound which achieves a half-maximal inhibition of the C-proteinase activity). Such information can be used to more accurately determine useful doses in humans. Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD50 and ED50. Compounds which exhibit high therapeutic indices are prefened. The data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. See, e.g., Fingl et al., 1975, in "The Pharmacological Basis of Therapeutics," Ch. 1 p.LDosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the C-proteinase inhibiting effects, of ntinimal effective concentration (MEC). The MEC will vary for each compound but can be estimated from in vitro data; for example, the concentration necessary to achieve 50-90% inhibition of the C-proteinase using the assays described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations. Dosage intervals can also be determined using MEC value. Compounds should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%. In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration. Refinement of the calculations necessary to determine the appropriate treatment dose is routinely made by those of ordinary skill in the art without undue experimentation, especially in light of the dosage information and assays disclosed herein as well as the pharmacokinetic data observed in animals or human clinical trials. As studies are conducted, further information will emerge regarding appropriate dosage levels and duration of treatment for specific diseases and conditions. In a prefened embodiment, the present invention is directed at treatment of colon cancer, including colon cancer indicated by the presence of overexpression of Prox-1. A variety of different routes of administration are contemplated. For example, in the case of a tumor, the discrete tumor mass may be injected. The injections may be single or multiple; where multiple, injections are made at about 1 cm spacings across the accessible surface of the tumor. Alternatively, targeting the tumor vasculature by direct, local or regional intra-arterial injection are contemplated. The lymphatic systems, including regional lymph nodes, present another likely target for delivery. Further, systemic injection may be prefened. It will be appreciated that the pharmaceutical compositions and treatment methods of the invention may be useful in fields of human medicine and veterinary medicine. Thus the subject to be treated may be a mammal, preferably human or other animal. For veterinary purposes, subjects include for example, farm animals including cows, sheep, pigs, horses and goats, companion animals such as dogs and cats, exotic and/or zoo animals, laboratory animals including mice rats, rabbits, guinea pigs and hamsters; and poultry such as chickens, turkey ducks and geese. The compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may, for example, comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
H. Transgenic Animals A transgenic animal can be prepared in a number of ways. A transgenic organism is one that has an extra or exogenous fragment of DNA incorporated into its genome, sometimes replacing an endogenous piece of DNA. In order to achieve stable inheritance of the extra or exogenous DNA, the integration event must occur in a cell type that can give rise to functional germ cells. The two animal cell types that are used for generating transgenic animals are fertilized egg cells and embryonic stem cells. Embryonic stem (ES) cells can be returned from in vitro culture to a "host" embryo where they become incorporated into the developing animal and can give rise to transgenic cells in all tissues, including germ cells. The ES cells are transfected in culture and then the mutation is transmitted into the germline by injecting the cells into an embryo. The animals carrying mutated germ cells are then bred to produce transgenic offspring. The use of ES cells to make genetic changed in the mouse germline is well recognized. For a reviews of this technology, those of skill in the art are refened to Bronson & Smithies, J. Biol. Chem., 269(44), 27155-27158, 1994; Tones, Curr. Top. Dev. Biol, 36, 99-114; 1998 and the references contained therein! Generally, blastocysts are isolated from pregnant mice at a given stage in development, for example, the blastocyst from mice may be isolated at day 4 of development (where day 1 is defined as the day of plug), into an appropriate buffer that will sustain the ES cells in an undifferentiated, pluripotent state. ES cell tines may be isolated by a number of methods well known to those of skill in the art. For example, the blastocysts may be allowed to attach to the culture dish and approximately 7 days later, the outgrowing inner cell mass picked, trypsinized and transfeπed to another culture dish in the same culture media. ES cell colonies appear 2-3 weeks later with between 5-7 individual colonies arising from each explanted inner cell mass. The ES cell lines can then be expanded for further analysis. Alternatively, ES cell lines can be isolated using the immunosurgery technique (described in Martin, Proc. Natl. Acad. Sci. USA 78:7634-7638, 1981) where the trophectoderm cells are destroyed using anti-mouse antibodies prior to explanting the inner cell mass. In generating transgenic animals, the ES cell lines that have been manipulated by homologous recombination are reintroduced into the embryonic envkonment by blastocyst injection (as described in Wilhams et al, Cell 52: 121-131, 1988). Briefly, blastocysts are isolated from a pregnant mouse and expanded. The expanded blastocysts are maintained in oil-drop cultures at 4°C for 10 minutes prior to culture. The ES cells are prepared by picking individual colonies, which are then incubated in phosphate-buffered saline, 0.5 mM EGTA for 5 minutes; a single cell suspension is prepared by incubation in a trypsin-EDTA solution containing 1% (v/v) chick serum for a further 5 minutes at 4°C. Five to twenty ES cells (in Dulbecco's modified Eagle's Medium with 10% (v/v) fetal calf serum and 3,000 units/ml DNAase 1 buffered in 20 mM HEPES [pH 8]) are injected into each blastocyst. The blastocysts are then transfeπed into pseudo-pregnant recipients and allowed to develop normally. The transgenic mice are identified by coat markers (Hogan et al. , Manipulating the Mouse Embryo, Cold Spring Harbor, N.Y. (1986)). Additional methods of isolating and propagating ES cells may be found in, for example, U.S. Patent No. 5,166,065; U.S. Patent No. 5,449,620; U.S. Patent No. 5,453,357; U.S. Patent No. 5,670,372; U.S. Patent No. 5,753,506; U.S. Patent No. 5,985,659, each incorporated herein by reference. An alternative method involving zygote injection method for making transgenic animals is described in, for example, U.S. Patent No. 4,736,866, incorporated herein by reference. Additional methods for producing transgenic animals are generally described by Wagner and Hoppe (U.S. Patent No. 4,873,191; which is incorporated herein by reference), Brinster et al. Proc. Nat'lAcad. Sci. USA, 82(13) 4438-4442, 1985; which is incorporated herein by reference in its entirety) and in Manipulating the Mouse Embryo; A Laboratory Manual, 2nd edition (eds., Hogan, Beddington, Costantimi and Long, Cold Spring Harbor Laboratory Press, 1994; which is incorporated herein by reference in its entkety). Briefly, this method involves injecting DNA into a fertilized egg, or zygote, and then allowing the egg to develop in a pseudo-pregnant mother. The zygote can be obtained using male and female animals of the same strain or from male and female animals of different strains. The transgenic animal that is born, the founder, is bred to produce more animals with the same DNA insertion. In this method of making transgenic animals, the new DNA typically randomly integrates into the genome by a non-homologous recombination event. One to many thousands of copies of the DNA may integrate at a site in the genome Generally, the DNA is injected into one of the pronuclei, usually the larger male pronucleus. The zygotes are then either transfeπed the same day, or cultured overnight to form 2-cell embryos and then transfeπed into the oviducts of pseudo-pregnant females. The animals born are screened for the presence of the desired integrated DNA. DNA clones for microinjection can be prepared by any means known in the art. For example, DNA clones for microinjection can be cleaved with enzymes appropriate for removing the bacterial plasmid sequences, and the DNA fragments electrophoresed on 1% agarose gels in TBE buffer, using standard techniques. The DNA bands are visualized by staining with ethidium bromide, and the band containing the expression sequences is excised. The excised band is then placed in dialysis bags containing 0.3 M sodium acetate, pH 7.0. DNA is electroeluted into the dialysis bags, extracted with a 1:1 phenόbchloroform solution and precipitated by two volumes of ethanob The DNA is redissolved in 1 ml of low salt buffer (0.2 M NaCb 20 mM Tris, pH 7.4, and 1 mM EDTA) and purified on an Elutip-D™ column. The column is first primed with 3 ml of high salt buffer (1 M Nad, 20 mM Tris, pH 7.4, and 1 mM EDTA) followed by washing with 5 ml of low salt buffer. The DNA solutions are passed through the column three times to bind DNA to the column matrix. After one wash with 3 ml of low salt buffer, the DNA is eluted with 0.4 ml high salt buffer and precipitated by two volumes of ethanob DNA concentrations are measured by absorption at 260 nm in a UV spectrophotometer. For microinjection, DNA concentrations are adjusted to 3 mg/ml in 5 mM Tris, pH 7.4 and 0.1 mM EDTA. Additional methods for purification of DNA for microinjection are described in Hogan et al. Manipulating the Mouse Embryo (Cold Spring Harbor
Laboratory, Cold Spring Harbor, NY, 1986), in Palmiter et al. Nature 300:611 (1982); in The Qiagenologist, Application Protocols, 3rd edition, published by Qiagen, Inc., Chatsworth, CA.; and in Sambrook et al. Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 1989). hi an exemplary microinjection procedure, female mice six weeks of age are induced to superovulate. The superovulating females are placed with males and allowed to mate. After approximately 21 hours, the mated females are sacrificed and embryos are recovered from excised oviducts and placed in an appropriate buffer, e.g., Dulbecco's phosphate buffered saline with 0.5% bovine serum albumin (BSA; Sigma). Sunounding cumulus cells are removed with hyaluronidase (1 mg/ml). Pronuclear embryos are then washed and placed in Earle's balanced salt solution containing 0.5 % BSA in a 37.5°C incubator with a humidified atmosphere at 5% C02, 95% ak until the time of injection. Embryos can be implanted at the two-cell stage. Randomly cycling adult female mice are paired with vasectomized males. C57BL/6 or Swiss mice or other comparable strains can be used for this purpose. Recipient females are mated at the same time as donor females. At the time of embryo transfer, the recipient females are anesthetized with an intraperitoneal injection of 0.015 ml of 2.5 % avertin per gram of body weight. The oviducts are exposed by a single midline dorsal incision. An incision is then made through the body wall dkectly over the oviduct. The ovarian bursa is then torn with watchmakers forceps. Embryos to be transfeπed are placed in DPBS (Dulbecco's phosphate buffered saline) and in the tip of a transfer pipette (about 10 to 12 embryos). The pipette tip is inserted into the infundibulum and the embryos transfeπed. After the transfer, the incision is closed by two sutures. The pregnant animals then give birth to the founder animals which are used to establish the transgenic line. I. Use of Prox-1-based Compositions for Diagnostic Purposes The demonstration that Prox-1 is overexpressed in precancerous and colon cancer cells also indicates that detection of Prox-1 polynucleotides and polypeptides (including variants thereof) are useful for diagnostic purposes. Therefore, prefened aspects of the present invention are dkected to methods of screening and diagnosing colon cancer in an individual. In one prefened embodiment, diagnostic methods of the invention are practiced through the detection of the Prox-1 protein. In general, methods for detecting a polypeptide of the invention can comprise contacting a biological sample with a compound that binds to and forms a complex with the polypeptide for a period sufficient to form the complex, and detecting the complex, so that if a complex is detected, a polypeptide of the invention is detected. Prox-1 protein detection can be accomplished using antibodies specific for the protein in any of a number of formats commonly used by those of skill in the art for such detection. For example, elsewhere in the present application, the production and characterization of monoclonal antibodies specific for Prox-1 is described. Such antibodies may be employed in ELISA-based techniques and Western blotting techniques to detect the presence of Prox-1 in a biological sample from a subject being tested. Methods for setting up ELISA assays and preparing Western blots of a sample are well known to those of skill in the art. The biological sample can be any tissue or fluid in which colon cells or tissue might be present. An anti-Prox-1 antibody or fragment thereof also is useful to monitor expression of this protein in individuals suffering from colon cancer. Typically, diagnostic assays entail detecting the formation of a complex resulting from the binding of an antibody or fragment thereof to Prox-1. For diagnostic purposes, the antibodies or antigen-binding fragments can be labeled or unlabeled. The antibodies or fragments can be directly labeled. A variety of labels can be employed, including, but not limited to, radionuclides, fluorescers, enzymes, enzyme substrates, enzyme cofactors, enzyme inhibitors and ligands (e.g., biotin, haptens). Numerous appropriate immunoassays are known to the skilled artisan (see, for example, U.S. Pat. Nos. 3,817,827; 3,850,752; 3,901,654 and 4,098,876). When unlabeled, the antibodies or fragments can be detected using suitable means, as in agglutination assays, for example. Unlabeled antibodies or fragments can also be used in combination with another (i.e., one or more) suitable reagent which can be used to detect antibody, such as a labeled antibody (e.g., a second antibody) reactive with the first antibody (e.g., anti-idiotype antibodies or other antibodies that are specific for the unlabeled immunoglobulin) or other suitable reagent (e.g., labeled protein A). In one embodiment, the antibodies or fragments of the present invention can be utilized in enzyme immunoassays, wherein the subject antibody or fragment, or second antibodies, are conjugated to an enzyme. When a biological sample comprising a Prox-1 protein is combined with the subject antibodies, binding occurs between the antibodies and the Prox-1 protein. In one embodiment, a biological sample containing cells expressing a mammalian Prox-1 protein, or biological fluid containing secreted Prox-1 is combined with the subject antibodies, and binding occurs between the antibodies and the Prox-1 protein present in the biological sample comprising an epitope recognized by the antibody. These bound protein can be separated from unbound reagents and the presence of the antibody- enzyme conjugate specifically bound to the Prox-1 protein can be determined, for example, by contacting the sample with a substrate of the enzyme which produces a color or other detectable change when acted on by the enzyme. In another embodiment, the subject antibodies can be unlabeled, and a second, labeled antibody can be added which recognizes the subject antibody. Similarly, the present invention also relates to a method of detecting and/or quantitating expression of a mammalian Prox-1 protein or a portion of the Prox-1 protein by a cell, in which a composition comprising a cell or fraction thereof (e.g., a soluble fraction) is contacted with an antibody or functional fragment thereof which binds to a mammalian Prox-1 protein or a portion of the Prox-1 protein under conditions appropriate for binding of the antibody or fragment thereto, and binding is monitored. Detection of the antibody, indicative of the formation of a complex between antibody and or a portion of the protein, indicates the presence of the protein. The method can be used to detect expression of Prox-1 from the cells of an individual (e.g., in a sample, such as a body fluid, such as blood, saliva or other suitable sample). The level of expression of in a biological sample of that individual can also be determined, for instance, by flow cytometry, and the level of expression (e.g., staining intensity) can be conelated with disease susceptibility, progression or risk. In certain other diagnostic embodiments, the polynucleotide sequences encoding Prox-1 protein may be used for the diagnosis of conditions or diseases with which the expression of Prox-1 protein is associated. In general, methods for detecting Prox-1 mRNA can comprise contacting a biological sample with a compound that binds to and forms a complex with Prox-1 mRNA for a period sufficient to form the complex, and detecting the complex in a quantitative or semi- quantitative way. Such methods can also comprise amplification techniques involving contacting a biological sample with nucleic acid primers that anneal to
Prox-1 mRNA or its complement, and amplifying annealed polynucleotides, so that if a polynucleotide is amplified, a polynucleotide of the invention is detected. The biological sample can be any tissue or fluid in which Prox-1 -expressing colon cells might be present. In the amplification procedures, polynucleotide sequences encoding
Prox-1 protein may be used in hybridization or PCR assays of fluids or tissues from biopsies to detect Prox-1 protein expression. Such methods may be qualitative or quantitative in nature and may include Southern or northern analysis, dot blot or other membrane-based technologies; PCR technologies; dip stick, pin, chip and ELISA technologies. All of these techniques are well known in the art and are the basis of many commercially available diagnostic kits. One such procedure known in the art is quantitative real-time PCR. Real-time quantitative can be conveniently accomplished using the commercially available ABI PRISM™ 7700 Sequence Detection System, available from PE- Applied Biosystems, Foster City, CA and used according to manufacturer's instructions. PCR reagents can be obtained from PE- Applied Biosystems, Foster City, CA. Gene target quantities obtained by real time RT-PCR may be normalized using either the expression level of GAPDH, a gene whose expression is constant, or by quantifying total RNA using RiboGreen™ (Molecular Probes, Inc. Eugene, OR). GAPDH expression is quantified by real time RT-PCR, by being run simultaneously with the target, multiplexing, or separately. Total RNA is quantified using
RiboGreen™ RNA quantification reagent from Molecular Probes. Methods of RNA quantification by RiboGreen™ are taught in Jones, L.J., et al, Analytical Biochemistry, 1998, 265, 368-374. Controls are analyzed in parallel to verify the absence of DNA in the RNA preparation (-RT control) as well as the absence of primer dimers in control samples lacking template RNA. In addition, RT-PCR products may be analyzed by gel electrophoresis. A reverse transcriptase PCR™ amplification procedure may be performed in order to quantify the amount of niRNA amplified. Methods of reverse transcribing RNA into cDNA are well known and described in Sambrook et ab, 1989. Alternative methods for reverse transcription utilize thermostable DNA polymerases. These methods are described in WO 90/07641, filed December 21, 1990. Conditions for incubating a nucleic acid probe or antibody with a test sample vary. Incubation conditions depend on the format employed in the assay, the detection methods employed, and the type and nature of the nucleic acid probe or antibody used in the assay. One skilled in the art will recognize that any one of the commonly available hybridization, amplification or immunological assay formats can readily be adapted to employ the nucleic acid probes or antibodies of the present invention. Examples of such assays can be found in Chard, T., An Introduction to Radioimmunoassay and Related Techniques, Elsevier Science Publishers, Amsterdam, The Netherlands (1986); Bullock, G.R. et al., Techniques in Immunocytochemistry, Academic Press, Orlando, FL Vol. 1 (1982), Vol. 2 (1983), Vol. 3 (1985); Tijssen, P., Practice and Theory of immunoassays: Laboratory Techniques in Biochemistry and Molecular Biology, Elsevier Science Publishers, Amsterdam, The Netherlands (1985). The tests of the present invention include cells, protein extracts of cells, or biological fluids such as, blood, serum, and plasma. The test sample used in the above-described method will vary based on the assay format, nature of the detection method and the tissues, cells or extracts used as the sample to be assayed. Methods for preparing protein extracts or membrane extracts of cells are well known in the art and can be readily be adapted in order to obtain a sample which is compatible with' the system utilized. In addition, such assays may be useful in evaluating the efficacy of a particular therapeutic treatment regime in animal studies, in clinical trials, or in monitoring the treatment of an individual patient. In order to provide a basis for the diagnosis of disease, a normal or standard measurement of Prox-1 mRNA or protein expression is established. This generally involves Prox-1 measurements from healthy colon tissue taken from one or more subjects, measured using the same or similar reagents used for the test subjects. The healthy subject preferably is matched for sex and age, and optionally, ethnicity. Deviation between standard and subject values conelates with the presence of precancerous or cancerous tissue. Once disease is established, a therapeutic agent is administered; and a treatment profile is generated. Such assays may be repeated on a regular basis to evaluate whether the values in the profile progress toward or return to the normal or standard pattern. Successive treatment profiles may be used to show the efficacy of treatment over a period of several days or several months. Methods to quantify the expression of a particular molecule include radiolabeling (Melby et al, J Immunol Methods 159: 235-44, 1993) or biotinylating (Duplaa et al, Anal Biochem 229-36, 1993) nucleotides, coamphfication of a control nucleic acid, and standard curves onto which the experimental results are interpolated. In addition to being used as diagnostic methods, screening methods also may be used in a prognostic manner to monitor the efficacy of treatment. The methods may be performed immediately before, during and after treatment to monitor treatment success. The methods also should be performed at intervals, preferably every three to six months, on disease free patients to insure treatment success. In another embodiment of the present invention, kits are provided which contain the necessary reagents to cany out the assays of the present invention. Specifically, the invention provides a compartment kit to receive, in close confinement, one or more containers which comprises: (a) a first container comprising one of the probes or antibodies of the present invention; and (b) one or more other containers comprising one or more of the following: wash reagents, reagents capable of detecting presence of a bound probe or antibody. In detail, a compartment kit includes any kit in which reagents are contained in separate containers. Such containers include small glass containers, plastic containers, or strips of plastic or paper. Such containers allow one to efficiently transfer reagents from one compartment to another compartment such that the biological sample and reagents are not cross-contaminated, and the agents or solutions of each container can be added in a quantitative fashion from one compartment to another. Such containers will include a container which will accept the test sample, a container which contains, for example, the antibodies used in the assay, containers which contain wash reagents (such as phosphate buffered saline, Tris-buffers, etc.), and containers which contain the reagents used to detect the bound antibody or probe. Types of detection reagents include labeled nucleic acid probes, labeled secondary antibodies, or in the alternative, ifthe primary antibody is labeled, the enzymatic, or antibody binding reagents which are capable of reacting with the labeled antibody. One skilled in the art will readily recognize that the disclosed probes and antibodies of the present invention can be readily incorporated into one of the established kit formats which are well known in the art. In further detail, kits for use in detecting the presence of a mammalian Prox-1 protein can include an antibody or functional fragment thereof which binds to a mammalian Prox-1 protein or portion of this protein, as well as one or more ancillary reagents suitable for detecting the presence of a complex between the antibody or fragment and Prox-1 or portion thereof. The antibody compositions of the present invention can be provided in lyophihzed form, either alone or in combination with additional antibodies specific for other epitopes. The antibodies, which can be labeled or unlabeled, can be included in the kits with adjunct ingredients. For example, the antibodies can be provided as a lyophihzed mixture with the adjunct ingredients, or the adjunct ingredients can be separately provided for combination by the user. Generally these adjunct materials will be present in less than about 5% weight based on the amount of active antibody, and usually will be present in a total amount of at least about 0.001% weight based on antibody concentration. Where a second antibody capable of binding to the monoclonal antibody is employed, such antibody can be provided in the kit, for instance in a separate vial or container. The second antibody, if present, is typically labeled, and can be formulated in an analogous manner with the antibody formulations described above.
J. Examples The present invention is illustrated in the following examples, which - are intended to be illustrative and not limiting. Upon consideration of the present disclosure, one of skill in the art will appreciate that many other embodiments and variations may be made in the scope of the present invention. Example 1 provides methods and materials for the subsequent Examples. Example 2 provides experimental results of studies designed to assess Prox-1 expression in colorectal cancer cells. Example 3 details expression of Prox-1 in round but not in adherent subclones of the SW480 colon adenocarcinoma cell line. Example 4 provides experimental results of Prox-1 silencing in
SW480R cells. Example 5 describes effects of Prox-1 ablation on Notch signaling in SW480R cells. Example 6 describes the effects of suppression of Prox-1 on the growth of SW480R cells in soft agar. Example 7 describes the effect Prox-1 suppression on prostaglandin biosynthesis. Example 8 describes experiments aimed as assessing the effects of altered Notch signaling. 5 Example 9 describes experiments aimed at assessing the effects of Prox-1 suppression on the growth of SW480R tumors in nude mice.
- - - - - -- - Example 10 describes analysis of Prox-1 in natural colorectal tumors. Example 11 describes one method for diagnosing or screening for colorectal cancer.
10 Example 12 describes experiments designed to compare Prox-1 expression in normal cololnic epithelium. Example 13 describes experiments aimed at assessing Prox-1 expression in Ape™ + mice. Example 14 describes studies conducted using SW480R cell line as an 15 in vitro model to investigate the role of Prox-1 in colorectal carcinoma. Example 15 describes experiments to characterize the effects of Prox-1 suppression and overexpression in colorectal cancer. Example 16 describes experiments employing dominant negative mutants of Prox- 1.
20 EXAMPLE 1 METHODS AND MATERIALS Methods and material used or referred to in subsequent examples are set forth dkectly below.
25 Antibodies Monoclonal mouse anti-vimentin, β-catenin (Transduction Laboratories), Ki-67 (Pharmingen) and chromogranin A (Ab-3, NeoMarkers), monoclonal rat anti-BrdU (Harlan Seralab) and polyclonal rabbit anti-Prox-1 were obtained from the indicated commercial sources. The fluorochrome-conjugated secondary antibodies were obtained from Jackson Immunoresearch. For production of Prox-1 antibodies cDNA encoding Prox-1 homeobox domain and prospero domain (amino acids 578-750 of human Prox-1, SEQ J_D NO: 3) was subcloned into pGEX2t vector to produce GST-Prox-1 fusion construct. This construct was expressed in E. coli and the GST-Prox-1 fusion protein from E. coli was purified using glutathione Sepharose according to the manufacturer's instructions (Amersham, Piscataway, NJ). Fusion protein was used to immunize rabbits according to a standard protocol. Prox-1 -specific antibodies were isolated from rabbit serum using sequential columns with GST- and GST-Prox-1 -coupled to vinylsulfone agarose resin (Sigma). Purified antibody recognized an 85 kD protein in lysates from 293T cells transfected with Prox-1 but not from cells transfected with the empty vector.
Synthetic siRNAs siRNA duplexes were prepared from synthetic 21 nucleotide RNAs
(Dharmacon Research). siRNA sequences were: 5'-
CUGCAAGCUGGAUAGUGAAGU-3' (Prox-1 siRNA A16 sense) (SEQ ID NO: 4);
5'-UUCACUAUCCAGCUUGCAGAU-3' (Prox-1 siRNA A16 antisense) (SEQ ID
NO: 5); 5'-CUAUGAGCCAGUUUGAUAUUU-3' (Prox-1 siRNA A25 sense) (SEQ ID NO: 6); 5'- AUAUCAAACUGGCUCAUAGUU-3' (Prox-1 siRNA A25 antisense) (SEQ JJD NO: 7). EGFP-targeting control siRNA A18 was essentially as described (Lewis et al., 2002) except that instead of thymidine 3' overhangs uracil overhangs were used; GACGUAAACGGCCACAAGUUU (EGFP siRNA A18 sense) (SEQ ID NO: 8); ACUUGUGGCCGUUUACGUCUU (EGFP siRNA A18 antisense) (SEQ ID NO: 9). siRNAs were 2'-ACE deprotected according to the manufacturer's instructions, dried in vacuum, resuspended in 400μl water, dried again, resuspended in water, and annealed to form duplex siRNAs. For anneahng equimolar amounts of siRNA strands (approximately 50-100μM) were incubated in anneahng buffer
(lOOmM potassium acetate 30mM Hepes-KOH pH 7.4, 2mM magnesium acetate) for 5 min at +95°C followed by 30 min at +37°C and 30 min at +25°C. After annealing the siRNA concentration was measured by spectrometry and siRNA aliquoted and stored at -20°C.
Cell culture, transfection, and soft agar assay SW480 cells were obtained from ATCC (CCL-228) and cultured in
RPMI- 1640 supplemented with 10% fetal bovine serum, 1 mM glutamine and antibiotics. HepG2 cells were cultured in DMEM, containing 10% fetal bovine serum 1 mM glutamine and antibiotics. Transfection of siRNAs was canied out using Lipofectamine 2000 (Invitrogen) according to manufacturer's instructions using 0.5% (v/v) lipofectamine 2000 reagent for SW480R and 0.4% (v/v) hpofectamine 2000 for adherent SW480 cells and either 20nM or lOOnM (f.c.) of siRNA. Transfections were carried out in antibiotic-free media for 4-6 hours before changing cells back to normal culture media. For long-term experiments siRNA transfections were repeated after 48-72h from previous transfection (at protein level the silencing effect was seen to remain efficient for at least 96h). Normally approximately 90-95% transfection efficiency was achieved. Opti-MEM (Invitrogen) medium was used in preparation of transfection mixtures. For luciferase assays, cells were transfected with Green Fluorescent Protein small interfering RNA (GFPsi RNA) or Prox-1 siRNAs 72 h prior to the transfection with the firefly lucefarese reporter constructs CBFl-luc, control pGL2- luc (Promega), TOPFlash and FOPFlash (Upstate). To normalize the transfection efficiency, cells were co-transfected with the Renilly firefly reporter pRL-TK (Promega). 36 h after the last transfection cells were lysed and lysates were analyzed for the luciferase activity using Dual-Luciferase™1 kit according to the manufacturer' s instructions (Promega). For soft agar assay, 2xl03 and 2xl04 cells were seeded in triplicate in 1 ml of 0.33% (w/v) agar (Difco) containing D-MEM, 10% fetal bovine serum, 1 mM glutamine and antibiotics in 6-well plates containing 1ml of 0.5% bottom agar layer. Cells were fed twice a week, and number of colonies per plate was scored after two weeks in culture. RNA isolation, Northern, and Western blotting Total RNA was isolated and DNAsel treated in RNeasy columns (Qiagen). For Cancer Array analysis, filters were hybridized in ExpressHyb with 32P-labeled probes for LYVE-l and Prox-1 according to the manufacturer's instructions (Clontech). For Northern analysis, the blots were hybridized in Ultrahyb solution (Ambion) with 32P-labeled probes produced by RT-PCR using RNA from SW480R or SW480A cells. The primers were designed to amplify 300-700 bp of the coding sequence, and all PCR-fragments were sequenced to confirm their identity. For the Affymetrix® gene expression analysis, sample preparations and hybridizations were carried out as described (Petrova et al. Embo J 21: 4593-9, 2002), using RNA extracted from two clones of SW480R or SW480A cells, or from two independent transfections of two different clones of SW480R cells with GFP siRNA or Prox-1 siRNA A16. To confirm the latter results, another transfection was carried out using Prox siRNA A25. To exclude the non-specific effects due to the transfection itself non-transfected SW480R cells grown in parallel were also analyzed. For Western blotting 2xl05 cells were lysed in 500 μl of sample buffer, lysates were separated using 10% PAGE and transfeπed to the nitrocellulose membranes (Schleisher&SchulI) using semi-dry transfer method for 1 h at 300 rnA. Membranes were blocked in 5% non-fat dry milk, 0.1% Tween-20 in 50 mM TrisHCb pH 7.4, 150 mM NaCb and incubated overnight with primary antibodies. Bound primary antibodies were detected using HRP-coηjugated conesponding secondary antibodies and the ECL detection method (KPL).
Immunofluorescence and immunohistochemistry The cells were cultured on coverslips, fixed with MetOH and stained with the primary antibodies and fluorochrome-conjugated secondary antibody. F- actin was stained using TexasRed-conjugated phalloidin (Molecular Probes). Cells were counterstained with Hoechst 33258 fluorochrome (Sigma) and viewed in Zeiss Axioplan 2 fluorescent microscope. For tissue staining staining, colon tumors and normal colon samples were embedded in Tissue-Tek® (Sakura), frozen and sectioned. The 4μm sections were fixed in cold methanol for 10 min and stained with the primary antibodies followed by peroxidase staining using Vectastain Elite ABC kit (Vector Laboratories) and 3-amino-9-ethyl carbazole (Sigma), or by detection using fluorchrome conjugated secondary antibodies. EXAMPLE 2 Prox-1 mRNA is Elevated in Colorectal Tumors Experiments were conducted to assess the expression of Prox-1 mRNA in human cancers using a cancer gene profiling aπay filter, which contains cDNAs from about 250 human cancers and conesponding normal control tissues. Prox-1 mRNA was significantly increased in 35 out of 53 samples of colorectal cancers. In contrast, only rarely or not at all was any increase seen in samples from breast, uterine, lung, kidney, ovarian, or thyroid tumors (Fig. IA, B, and C). Probes for Prox-1 (Fig. IA) and the lymphatic endothelial marker LYVE-l (Fig. IB) were used. Fig. IC demonstrates quantification of dot blot in Fig. IA, the asterisk indicating tumor samples in which Prox-1 expression is significantly different from that of the normal tissue (P<0.005). Expression of Prox-1 was low or absent in all kidney cancer samples studied. Prox-1 is a marker for lymphatic vessels, which are abundant both in normal colonic submucosa and around colon carcinomas (White et al., Cancer Res. 62: 1669-75 (2002)). Therefore, the filter to the probe for the lymphatic endothehal hyaluronan receptor LYVE-l was hybridized. Unlike Prox-1, LYVE-l levels were higher in the normal samples, suggesting that the increased expression cannot be attributed to the lymphatic vessels (Fig. IB). Experiments were further conducted to assess the expression of Prox-1 in colon cancers and premalignant colonic lesions using affinity purified antibodies raised against Prox-1 homeobox and prospero domains, which are conserved between the mouse and human proteins. Staining of a panel of mouse tissues and E12.5 and E17.5 embryos revealed specific nuclear staining for Prox-1 in the previously reported sites of expression such as in lymphatic vessels, lens fiber cells and in a subset of neurons in the neural tube. Staining of eleven human colorectal adenomas and nine carcinomas and adjacent normal mucosa revealed increased expression of Prox-1 in nine adenomas and in six carcinomas (Fig. 2A-I). Increased Prox-1 staining was observed in all cells in seven adenomas and in two carcinomas, whereas in the other lesions a heterogeneous expression of Prox-1 occuned. In one tumor sample, no specific staining for Prox-1 was seen, while strong expression was observed in intratumoral lymphatic vessels. Double immunofluorescent staining for Prox-1 and the neuroendocrine marker chromogranin A or proliferation marker Ki-67 was conducted in normal colonic epithelial cells. Nuclei were visualized with Hoechst 333421. In the normal colonic mucosa, Prox-1 was strongly expressed in some epithelial cells, a subset of which was positive for the pan-neuroendocrine marker chromogranin A. In addition, a weaker but significant Prox-1 expression was observed in the bottom of the crypts below the cell prohferation zone identified by staining for the Ki-67 antigen. The location of Prox-1 positive cells at the base of the crypts conesponds to the position of the intestinal stem cells (Bach et ab, Carcinogenesis 21: 469-76 (2000)). EXAMPLE 3
PROX-1 IS EXPRESSED IN ROUND BUT NOT IN ADHERENT SUBCLONES OF THE SW480 COLON ADENOCARCINOMA CELL LINE. Additional studies were conducted to compare Prox-1 expression in various cells. No Prox-1 expression was seen in the majority of tumor cell lines studied. However, Prox-1 was mRNA was present in hepatocellular carcinoma cell line HepG2 and the colon carcinoma cell line SW480. BEC, blood endothelial cells, CAEC, coronary artery endothelial cells, and LEC, lymphatic endothelial cells, served as negative and positive controls. Immunofluorescent staining of Prox-1 revealed strong expression in all HepG2 cells, whereas only a subset of SW480 cells were Prox-1 positiveDouble immunofluorescent staining for Prox-1 and for β-catenin or for the F-actin marker phalloidin demonstrated that Prox-1 expression is restricted to weakly adherent round SW480 cells which did not display focal adhesions or actin stress fibers, and that Prox-1 was very weakly expressed the adherent cells. The existence of two subtypes of cells in the SW480 cultures has been reported previously (Palmer, H. G. et ab, J Cell Biol. 154: 369-87, 2001; Tornita, N. et ab, Cancer Res. 52; 6840-7, 1992). The SW480R (round) cells displayed anchorage independent growth in vitro and highly malignant phenotype in vivo, whereas the SW480A (adherent) cells did not grow well in soft agar and formed small and well differentiated tumors when implanted into nude mice. Several SW480R and SW480A clones were isolated, which could be continuously grown for at least 20 passages without conversion of phenotypes. SW480R and SW480A cells differed by the levels of Prox-1, as determined by Northern and Western blotting, with much higher expression in the round cells, and weak, if any, expression in the Adherent ones. The gene expression profiles of SW480R and SW480A cells were compared using oligonucleotide microarrays containing 22,000 annotated human genes, and identified about 1,000 genes whose expression differed by more than fourfold between these two cell types (Table I). SW480 cells were stained for intermediate filament protein vimentin and Prox-1. Northern blotting and hybridization were used for transcripts. Hybridization for
GAPDH was used as a control. A striking difference was observed in the expression of cytoskeletal and cell adhesion proteins. In agreement with thek decreased adhesion and round cell shape, the SW480R cells lacked many components of the actin, intermediate filament and microtubule networks, such as gelsolin, filamins A and B, ezrin, moesin, vimentin, various integrins, and tubulins (Table I). These cells expressed higher levels of the protoncogene c-met, as well as the receptor tyrosine kinase FGFR-4, which has been associated with malignant transformation in colorectal and other cancer (Bange, J. et ab, Cancer Res. 62: 840-7, 2002; Cavallaro, TJ., Niedermeyer, J., Fuxa, M. & Christofori, G., Nat. Cell Biol. 3: 650-7, 2001; Yamada, S. M. et al., Neurol Res. 24: 244-8, 2002), and low levels of the tumor suppressor p21Cipl. FGFR-4 is a target for therapeutic intervention according to the invention, alone or in combination with Prox-1. Intervention using the same classes of inhibitors as described for Prox-1, as well as antibodies and antibody fragment substances, is specifically contemplated. In addition, all three tissue inhibitors of matrix metalloproteinases were absent from the SW480R cells, which may further account for thek increased tumor growth in vivo. In contrast, the SW480A cells expressed higher levels of the chemokine receptor CXCR4, which is expressed in the normal colonic epithelium (Jordan et ab, J Clin Invest 104, 1061-9, 1999). In summary, the gene expression profile of the SW480R cells conelates well with a highly aggressive transformed phenotype, whereas the SW480A cells display more differentiated features typical of cells in the colonic crypts. Table I. Examples of groups of genes differentially expressed in round versus adherent SW480 clones. Two round and two adherent clones were analyzed. Gene function and name UniGene Gene Log2 ratio, St. cluster symbol average dev
1. Cytoskeleton and adhesion collagen, type XIJJ, alpha 1 Hs.211933 COL13A1 -5.6 0.9 fibronectin 1 Hs.287820 FN1 -5.2 0.5 integrin, alpha 7 Hs.74369 1TGA7 -4.3 0.3 vimentin Hs.297753 V -4.1 0.6 filamin B, beta (actin binding protein 278) . . Hs.81008 FLΝB -3.8 0.7 integrin, beta 5 Hs.149846 ITGB5 -3.6 0.5 tubulin, beta polypeptide Hs.274398 TUBB -3.3 0.7 PTPLl-associated RhoGAP 1 Hs.70983 PARG1 -3.0 0.5 collagen, type LX, alpha 3 Hs.53563 COL9A3 -2.8 0.8 paralemmin Hs.78482 PALM -2.7 0.2
PDZ and LJM domain 1 (elfin) Hs.75807 PDLEVI1 -2.7 0.2 cadherin 11, type 2, OB-cadherin (osteoblast) Hs.75929 CDH11 -2.6 0.7 myosin IC Hs.286226 MYOIC -2.6 0.6 integrin, alpha 3 Hs.265829 1TGA3 -2.6 0.4 discs, large (Drosophila) homolog 1 Hs.154294 DLG1 -2.5 0.1 integrin, alphaV Hs.295726 LTGAV -2.5 0.3
CDC42 effector protein (Rho GTPase Hs.260024 binding) 3 CDC42EP3 -2.4 0.4 ephrin-Bl Hs.144700 EFΝB1 -2.3 0.4
PERM, RhoGEF (ARHGEF) and pleckstrin Hs.183738 domain protein 1 FARP1 -2.3 0.4 myosin ID Hs.39871 MYOID -2.1 0.2
PDZ and LJM domain 2 (mystique) Hs.379109 PDLJVI2 -2.1 0.4 tubulin beta-5 Hs.274398 TUBB-5 -1.9 0.3 erythrocyte membrane protein band 4.1 -like 1 Hs.26395 EPB41L1 -1.9 0.1 gelsolin (amyloidosis, Finnish type) Hs.290070 GSΝ -1.9 0.3 laminin, gamma 1 (formerly LAMB2) Hs.432855 LAMCl -1.8 0.1 ras homolog gene family, member E Hs.6838 ARHE -1.7 0.2 IQ motif containing GTPase activating Hs.1742 protein 1 IQGAP1 -1.7 0.3 tight junction protein 1 (zona occludens 1) Hs.74614 TJP1 -1.7 0.4 catenin (cadherin-associated protein), alphaHs.58488 like 1 CTΝΝAL1 -1.7 0.6 collagen, type XVIII, alpha 1 Hs.78409 COL18A1 -1.6 .0.1 filamin A, alpha (actin binding protein 280) Hs.195464 FLΝA -1.6 0.2 actin related protein 2/3 complex, subunit IA, Hs.90370 41kDa ARPCIA -1.5 0.3 alpha integrin binding protein 63 - AIBP63 -1.4 0.3 spectrin, alpha, non-erythrocytic 1 (alpha- Hs.77196 fodrin) SPTAΝ1 -1.4 0.2 villin 2 (ezrin) Hs.155191 VIL2 -1.4 0.3 actin related protein 2/3 complex, subunit IB, Hs.433506 41kDa ARPCIB -1.3 0.1 plakophilin 4 Hs.152151 PKP4 -1.3 0.3 ras homolog gene family, member C Hs.179735 ARHC -1.1 0.1 moesin Hs.170328 MSΝ -1.1 0.1 myristoylated alanine-rich protein kinase C Hs.75607 substrate MARCKS -1.1 0.2
2. Tumor growth and invasion tissue inhibitor of metalloproteinase 2 Hs 6441 TLMP2 -2.3 0.21 tissue inhibitor of metalloproteinase 3 Hs.245188 TLMP3 -1.5 0.14 Cyclin-dependent kinase irihibitor IA (p21, Hs 179665 Cipl) CDKN1A -2.5 0 tissue inhibitor of metalloproteinase 1 Hs .5831 TLMP1 -1.5 0.4 met proto-oncogene (hepatocyte growth factorHs316752 receptor) MET 2.6 0.46 Fibroblast growth receptor 4 Hs 165950 FGFR4 3.9 0.76
3. Expressed in normal intestinal epithelium CXCR4 Hs 89414 CXCR4 -1.3 0.1 solute carrier family 7 (cationic amino acid Hs 22891 transporter, y+ system), member 8 SLC7A8 -1.8
4. Notch pathway
Notch homolog 2 (Drosophila) Hs. .8121 NOTCH2 -1.4 0.15 hairy homolog (Drosophila), HES1 Hs. .250666 HRY -2.1 0.2 jagged 2 Hs. .166154 JAG2 1.6 0.61
5. Wnt pathway wingless-type MMTV integration site family, Hs. 152213 0.12 member 5A WNT5A -5.8 dickkopf homolog 3 Hs. ,4909 DKK3 -5.6 1.21 wingless-type MMTV integration site family, Hs.29764 0.23 member 6 WNT6 -4.2 frizzled homolog 7 (Drosophila) Hs. 173859 FZD7 -4.1 0.65 frizzled homolog 2 (Drosophila) Hs 81217 FZD2 -3.1 0.56 frizzled homolog 10 (Drosophila) Hs 31664 FZD10 2.97 0.86 dickkopf homolog 4 Hs.159311 DKK4 7.37 0.71
EXAMPLE 4 PROX-1 SILENCING IN SW480R CELLS LEADS TO A DIFFERENTIATED AND QUIESCENT PHENOTYPE. Experiments were conducted to investigate whether Prox-1 plays role in the generation and maintenance of the highly transformed phenotype. Prox-1 mRNA and protein in the SW480R cells was suppressed using Prox-1 targeting siRNA. Absence of Prox-1 in Prox-1 siRNA but not the control GFP siRNA transfected cells was confirmed by immunofluorescent staining, and nuclei were visualized with Hoechst 33342. Prox-1 siRNA-transfected cells but not the untransfected or GFP siRNA transfected cells underwent a morphological change, which became visible by 72 hours and persisted at least for 10 days after the transient transfection. The Prox-1 siRNA transfected cells become first more elongated and displayed extensive membrane ruffling. Eventually the Prox-1 siRNA cells started to spread on the plate and a number of increased actin stress fibers could be visualized by phalloidine staining. BrdU incorporation experiments demonstrated that the Prox- 1 siRNA transfected cells proliferated at the lower rate than GFPsi or nontransfected cells (22+0.5% of BrdU positive cells in Prox-1 siRNA A16, 18+1 % Prox-1 siRNA A25 vs 34±4% GFP siRNA). Changes in the gene expression profiles of the SW480R and SW480A cells 120 and 240 h posttransfection, when the morphological changes were apparent, were also analyzed. Only 29 down-regulated and 120 upregulated genes in Prox-1 siRNA versus GFP siRNA transfected cells (Table II) were identified. 41 % of these genes were differentially expressed between the SW480R and SW480A cells, suggesting that Prox-1 at least partially determines the phenotype of SW480R cells. The ablation of Prox-1 led to upregulation of a number of known epithelial markers, such as annexin Al, CRPB2, S100A3, and EMP1, along with the increase in cell adhesion molecules OB-cadherin and integrins beta7, beta5 and alpha 1. In line with the observed growth arrest, also observed was the decrease in c-myc and a strong increase of CDK inhibitor p21Cipl. Highly similar changes in gene expression profile were observed when another unrelated Prox-1 si RNA was used, suggesting that the cellular effects are due to the specific targeting of Prox-1, and they did not result from off-target silencing. In addition, titration experiments demonstrated that the induction of p21 and other target genes occuned even at the low (20 nM) concentration of Prox-1 siRNAs but not of the control GFP siRNA. Also, the mentioned gene changes were not observed in Prox-1 negative SW480A cells transfected with siRNAs at 100 nm concentration. The transfection efficiency was controlled using another siRNA, which successfully suppressed the expression of the target gene in SW480A cells. Table JJ. Genes regulated by Prox-1 in SW480R cells. Asterisk indicates genes that were flagged as absent in either Prox-1 siRNA or GFP siRNA treated cells. Genes differentially expressed between SW480R and SW480ADH cells are shown in bold. Genes down-regulated in the absence of Prox-1 UniGene Gene Log2 ratio, side1 cluster symbol average
Nebulette Hs.5025 NEBL -2.0 0.4 transforming growth factor, beta-induced, Hs.H8787 TGFBI -1.9 0.1
68kDa trinucleotide repeat containing 9 Hs.H0826 TNRC9 -1.9 0.2 insulin-like growth factor binding protein 3 Hs.77326 IGFBP3 -1.6 0.0 calpain 1, (mu/l) large subunit Hs.2575 CAPNl -1.5 0.3 inhibitor of DNA binding 1 Hs.75424 ΓDI -1.5 0.3 midkine (neurite growth-promoting factor 2) Hs.82045 MDK -1.5 0.1
FK506 binding protein 11, 19 kDa Hs.24048 FKBP11 -1.4 0.1 caspase recruitment domain family, member 10 Hs.57973 CARDIO -1.3 0.1 inhibin, beta B (activin AB beta polypeptide) Hs.1735 HMHBB -1.3 0.2
LI cell adhesion molecule Hs.1757 L1CAM -1.2 0.1 glutathione peroxidase 2 (gastrointestinal) Hs.2704 GPX2 -1.2 0.0 eukaryotic translation elongation factor 1 alpha Hs.2642 EEF1A2 -1.2 0.2
2 hypothetical protein FLJl 1149 Hs.37558 FLJl 1149 -1.2 0.2 potassium voltage-gated channel, subfamily H Hs.188021 KCNH2 -1.1 0.1
(eag-related), member 2
KIAA0182 protein Hs.75909 KIAA0182 -1.1 0.0 lectin, galactoside-binding, soluble, 1 (galectin 1) Hs.382367 LGALSl -1.1 0.1
Homo sapiens cDNA ELJ41000 fis, - - -1.1 0.3 ephrin-B2 Hs.30942 EFNB2 -1.1 0.1 v-myc myelocytomatosis viral oncogene Hs.79070 MYC -1.1 0.1 homolog (avian)
S100 calcium binding protein A14 Hs.288998 S100A14 -1.1 0.2
Alpha one globin [Homo sapiens], mRNA -1.1 0.1 sequence* hypothetical protein FLJ10986* Hs.273333 FLJ10986 -1.0 0.0 hypothetical protein FLJ11149 Hs.37558 FLJ11149 -1 0.0 myelin transcription factor 1* MYT1 -1.0 0.0 nucleolar autoantigen (55kD) similar to rat Hs.446459 SC65 -1.0 0.1 synaptonemal complex protein* tumor necrosis factor receptor superfamily, Hs.455817 TNFRSF6B -1.0 0.1 member 6b, decoy jagged 2 Hs.166154 JAG2 -1.0 0.1 mitochondrial ribosomal protein S2 Hs.20776 MRPS2 -1.0 0.1
Total: 29 genes Genes up-regulated in the absence of Proxl UniGene Gene Log2 ratio, Stdev cluster symbol average insulin-like growth factor binding protein 7* Hs.119206 IGFBP7 5.8 0.4 chitinase 3-like 1 (cartilage glycoprotein-39)* HsJ5184 CHI3L1 5.3 0.8 chemokine (C-X-C motif) receptor 4* Hs.89414 CXCR4 4.5 1.1 semaphorin 3C* Hs.171921 SEMA3C 4.5 4.5 cadherin 11, type 2, OB-cadherin (osteoblast)* Hs.75929 CDH11 3.8 0.3 annexin Al Hs.78225 ANXA1 3.7 1.1 hypothetical protein MGC10796* MGC1079 3.3 0.4 6
CD44 antigen Hs.169610 CD44 2.6 1.1
Homo sapiens clone 23785 mRNA sequence 2.9 0.4 epithelial membrane protein 1* Hs.79368 EMP1 2.9 0.1 inhibitor of DNA binding 2, dominant negative Hs.180919 ID2 2.8 0.1 helix-loop-helix protein*
Human HepG23' region cDNA, clone hmdlfOό, - 2.8 0.3 mRNA sequence tumor necrosis factor receptor superfamily, Hs.81791 TNFRSF1 2.6 0.7 member *llb (osteoprotegerin) IB likely homolog of mouse glucuronyl C5-epimerase* Hs.183006 GLCE 2.6 1.1 ribonuclease, RNase A family, 1 (pancreatic)* Hs.78224 RNASE1 2.6 0.1 apolipoprotein B mRNA editing enzyme, catalyticHs.226307 APOBEC 2.5 0.1 polypeptide-like 3B* 3B hydroxyprostaglandin dehydrogenase 15-(NAD)* Hs.77348 HPGD 2.5 1.1
NPD009 protein Hs.283675 NPD009 2.5 0.6 integrin, beta 7* Hs.1741 ITGB7 2.4 0.1 fibroblast growth factor 20* Hs.154302 FGF20 2.3 1.0
KIAA0455 gene product Hs.13245 KIAA0455 2.3 1.3
CAMP-specific hosphodiesterase 8Bl [Homo Hs.78106 PDE8B 2.3 0.4 sapiens], mRNA sequence* ectodermal-neural cortex (with BTB-like Hs.104925 ENC1 2.3 0.2 domain)* frizzled homolog 1 (Drosophila)* Hs. 94234 FZD1 2.3 0.8
S100 calcium binding protein A3* Hs. 433168 S100A3 2.2 0.6 zeta-chain (TCR) associated protein kinase 70kDa* Hs. 234569 ZAF70 2.2 1.1 platelet derived growth factor C* Hs, 43080 PDGFC 2.1 0.1 cystatin D * Hs. 121489 CST5 2.1 0.3
CCAAT/enhancer binding protein (C/EBP), deltaHs .76722 CEBPD 2.1 0.1 sorbin and SH3 domain containing 1 Hs. 108924 SORBS1 2.1 0.5 metallothionein 2A Hs. 118786 MT2A 2.0 0.6
RAS guanyl releasing protein 1 (calcium and DAG- Hs. 182591 RASGRPl 2.0 0.4 regulated) checkpoint suppressor 1 Hs .211773 CHES1 2.0 0.4 chondroitin betal,4 N- Hs.11260 ChGn 2.0 0.4 acetylgalactosa inyltransferase* filamin B, beta (actin binding protein 278)* Hs, .81008 FLNB 2.0 0.4 aldehyde dehydrogenase 1 family, member A2* Hs. 95197 ALDH1A2 2.0 0.6 jagged 1 (Alagille syndrome) Hs. 91143 JAG1 2.0 0.1
A kinase (PRKA) anchor protein (gravin) 12* Hs. .788 AKAP12 1.9 0.1 metallothionein IX* Hs. 380778 MT1X 1.9 0.8 creatine kinase, mitochondrial 2 (sarcomeric) Hs, ,80691 CKMT2 1.8 0.6 serum-inducible kinase Hs.3838 SNK 1.8 0.1
CGI-130 protein Hs.32826 CGI-130 1.8 0.1 guanine nucleotide binding protein (G protein), Hs.203862 GNAI1 1.8 0.4 alpha inhibiting activity polypeptide 1 related to the N terminus of tre* Hs.278526 RNTRE 1.7 0.4 solute carrier family 12 (sodium/potassium/chloride Hs.110736 SLC12A2 1.7 0.3 transporters), member 2
Human clone 23612 mRNA sequence _ _ 1.7 1.0 ankyrin repeat and SOCS box-containing 4 Hs.248062 ASB4 1.7 0.8 apolipoprotein B mRNA editing enzyme, catalytic Hs.8583 APOBEC3 1.7 0.1 polypeptide-like 3C C cellular retinoic acid binding protein 2* Hs.183650 CRABP2 1.7 0.1
KIAA0657 protein* Hs.6654 KIAA065 1.7 1.1 7 phosphodiesterase 4D, cAMP-specilϊc Hs.172081 PDE4D 1.7 0.1 (phosphodiesterase E3 dunce homolog, Drosophila) autism susceptibility candidate 2 Hs.32168 AUTS2 1.6 0.4 hairy/enhancer-of-split related with YRPW motif 2* Hs.144287 HEY2 1.6 0.0 immediate early response 5 Hs.15725 IER5 1.6 0.1 E3 ubiquitin ligase SMURF2 Hs.194477 SMURF2 1.6 0.4 ADP-ribosylation factor-like 7* Hs.111554 ARL7 1.6 1.0 Ras and Rab interactor 2* Hs.62349 RIN2 1.6 0.4 GS3955 protein, Tribbles homolog 2 Hs.155418 TRB2 1.6 0.5 metallothionein IL Hs.448357 MT1L 1.5 0.6 glutamate receptor, metabotropic 8 Hs.86204 GRM8 1.5 0.2 klotho Hs.94592 KL 1.5 0.1 calmoduhn-like 3 Hs.239600 CALML3 1.4 0.6 integrin, alpha 1 Hs.116774 ΓTGAI 1.4 0.1 lymphoid enhancer-binding factor 1 Hs.44865 LEF1 1.4 0.4 epithelial V-like antigen 1 Hs.116651 EVA1 1.4 0.1 likely ortholog of mouse limb-bud and heart Hs.57209 LBH 1.4 0.1 gene* insulin induced protein 2 Hs.7089 ISG2 1.4 0.2 patched homolog (Drosophila) Hs.159526 PTCH 1.4 0.1 chemokine-like factor super family 6 Hs.380627 CKLFSF6 1.3 0.3 lipoma HMGIC fusion partner Hs.93765 LHFP 1.3 0.4 transforming growth factor, alpha Hs.170009 TGFA 1.3 0.4
Homo sapiens mRNA; cDNA DKFZp762M127 - - 1.3 0.6
(from clone DKFZp762M127), mRNA sequence cyclin I Hs.79933 CCNI 1.3 0.1 hyaluronan synthase 2 Hs.159226 HAS2 1.3 0.5
IQ motif containing GTPase activating protein 1 Hs.1742 IQGAPl 1.3 0.5 zinc finger protein 216 Hs.406096 ZNF216 1.3 0.2 cDNA DKFZp564O0122 - - 1.3 0.2 aryl hydrocarbon receptor Hs.170087 AHR 1.2 0.6 neuroepithelial cell transforming gene 1 Hs.25155 NET1 1.2 0.1 sterol-C4-methyl oxidase-like Hs.239926 SC4MOL 1.2 0.1 tubulin, alpha 3 Hs.433394 TUBA3 1.2 0.1
BCG-induced gene in monocytes, clone 103 Hs.284205 BIGM103 1.2 0.0 cathepsin B Hs.297939 CTSB 1.2 0.0 keratin 6A Hs.367762 KRT6A 1.2 0.4 paraoxonase 2 Hs.169857 PON2 1.2 0.4 suppressor of cytokine signaling 5 Hs.169836 SOCS5 1.2 0.4
KIAA0877 protein Hs.H217 KIAA0877 1.2 0.2 propionyl Coenzyme A carboxylase alpha Hs.80741 PCCA 1.2 0.2 solute carrier family 2 Hs.7594 SLC2A3 1.2 0.1 solute carrier family 7 Hs.22891 SLC7A8 1.2 0.1
Homo sapiens mRNA; cDNA DKFZp762M127 - - 1.2 0.1 aryl hydrocarbon receptor nuclear translocator-like Hs.74515 ARNTL 1.1 0.3 DnaJ (Hsp40) homolog, subfamily B, member 6 Hs.181195 DNAJB6 1.1 0.3 hypothetical protein ELJ21276 - FLJ21276 1.1 0.1 integrin, beta 5 Hs.149846 ITGB5 1.1 0.1 PTK7 protein tyrosine kinase 7 Hs.90572 PTK7 1.1 0.3 transforming growth factor, beta receptor II Hs.82028 TGFBR2 1.1 0.1 Homo sapiens cDNA FLJ25134 fis Hs.301306 1.1 0.0 DKFZP564A2416 protein Hs.5297 DKFZP56 1.1 0.1 4A2416 dual specificity phosphatase 6 Hs.180383 DUSP6 1.1 0.4 idline 1 (Opitz/BBB syndrome) Hs.27695 MIDI 1.1 0.1 membrane protein, palmitoylated 1, 55kDa Hs.1861- MPP1 1.1 0.1
LIM domain protein Hs.424312 REL 1.1 0.1
SH3-domain binding protein 5 Q3TK-associated) Hs.109150 SH3BP5 1.1 0.1
SIPL protein Hs.64322 SIPL 1.1 0.1 tumor protein D52-like 1 Hs.16611 TPD52L1 1.1 0.4
3-hydroxy-3-methylglutaryl-Coenzyme A reductase Hs.H899 HMGCR 1.0 0.1 homeo box B7 Hs.819 HOXB7 1.0 0.1
HTV-1 Tat interactive protein 2, 30kDa Hs.90753 HTATIP2 1.0 0.1 insulin receptor substrate 2 Hs.143648 ms2 1.0 0.1 tubulin beta-5 Hs.274398 TUBB-5 1.0 0.0 apoptosis antagonizing transcription factor Hs.16178 AATF 1.0 0.1
E2F transcription factor 3 Hs.H89 E2F3 1.0 0.1 hypothetical protein FLJ12542 Hs.236940 FLJ12542 1.0 0.1 phaiϊn 2, Pleckstrin homology domain containing, Hs.29724 PLEKHF2 1.0 0.1 family F member 2 proline 4-hydroxylase Hs.3622 P4HA2 1.0 0.1
Homo sapiens G21VN02 mRNA, mRNA Hs.324787 1.0 0.1 sequence, solute carrier family 5 (inositol SLC5A3 transporters), member 3
EXAMPLE 5 ABLATION OF PROX-1 LEADS TO DD?FERENTIATION THROUGH UPREGULATION OF NOTCH SIGNALING IN THE SW480R CELLS. Activation of β-catenin/TCF pathway plays a central role in colon tumorigenesis (Giles, R. H., van Es, J. H. & Clevers, H, Biochim Biophys Acta 1653: 1-24, 2003). Of interest for this study, suppression of β-catenin/TCF signahng in colon cancer cells decreases the levels of c-myc, increases p21Cipl levels and induces cell cycle aπest (van de Wetering et ab, Cell 111:, 241-50, 2002). However, suppression of Prox-1 did not affect the activity of β-catenin TCF-responsive reporter or nuclear localization of β-catenin. Moreover, an increased expression of several β- catenin/TCF-4 target genes, such as CD44, ENC1 and Id2 was observed in the absence of Prox-1 (Table JJ and not shown). These data suggest that Prox-1 may act via an alternative pathway to promote growth of colon cancer cells, and that both β- catenin/TCF activation and overexpression of Prox-1 are necessary for cell transformation. Accordingly, contemplated herein are methods of alleviating colorectal cancer whereby a Prox-1 suppressor is administered in combination with a β-catenin/TCF signaling inhibitor. β-catenin/TCF signaling inhibitors may include dominant negative forms of TCF-4, siRNAs and microRNAs targeting TCF-4, β- catenin, and c-myc, as well as small molecules that would interfere with binding of β- catenin to TCF-4 or TCF-4 to target DNA sequences. Protocols for making these types of inhibitors are detailed above with respect to Prox-1 inhibition. The DNA and protein sequences for β-catenin (SEQ ID NOs: 10 and 11, respectively) are published and disclosed as Genbank Accession Number NM_001904. The DNA and protein sequences for TCF-4 (SEQ ED NOs: 12 and 13, respectively) are published and disclosed as Genbank Accession Number NM_003199. Related to the β-catenin/TCF signahng pathway is the APC gene, the sequence of which is publicly available as Genbank Accession Number NM_000038. The DNA and amino acid sequences for APC are also provided herein as SEQ ID NOs: 42 and 43, respectively. The DNA and protein sequences for C-myc (SEQ ID NOs: 44 and 45, respectively) are published and disclosed as Genbank Accession Number NM_002467. Notch signaling has been shown to be essential for the generation of cell lineages in the crypts of the mouse small intestine. High levels of Notch are thought to suppress the expression of the basic helix-loop-helix transcription factor Mathl via the induction of the transcriptional repressor Hesl, which will lead to the differentiation of progenitor cells into enterocytes. Conversely, high levels of Mathl result in the differentiation towards the neuroendocrine, Goblet and Paneth cell types in the small intestine (Jensen, J. et ab, Nat Genet 24: 36-44, 2000; Yang, Q., Bermingham, N. A., Finegold, M. J. & Zoghbi, H. Y., Science 294: 2155-8, 2001). Among Notch signaling components, Notch2 and its target transcription factor Hesl levels are higher in SW480A cells in comparison with the SW480R cells, suggesting that this pathway is functionally active in these cells. Interestingly, SW480R cells express higher levels of Notch ligand Jagged2. Suppression of Prox-1 resulted in upregulation of the Notch ligand Jagged 1 and the dkect target of the Notch pathway, the transcription factor Hey2, whereas the expression of Jagged2 and prostaglandin D2 synthase, previously shown to be negatively regulated by Notch signaling was suppressed (Fujimori, K. et al., JBiol Chem 278: 6018-26, 2003). SW480R cells were transfected with GFP siRNA or Prox-1 siRNA and GFBl-luc, TOPFlash or control FOP flash reporters. Fkefly luciferase activity was normalized to Renilla luciferase activity. Up-regulation of Notch-responsive reporter GBFl-luc was observed in SW480R cells transfected with Prox-1 siRNAs. Accordingly, contemplated herein is a method of alleviating the symptoms of colorectal cancer comprising the administration of a Prox-1 suppressor in combination with a Notch agonist or target transcription factor. Notch agonists include Jagged 1, Jagged2, Deltal, Delta3, Delta4, and Senate. Target Notch transcription factors include Heyl, Hey2, and Hesl. The DNA and protein sequences for Notch-1 (SEQ ID NOs: 14 and 15, respectively) are pubhshed and disclosed as Genbank Accession Numbers
NM_017617. Likewise, the DNA and protein sequences for various forms of Notch (including 2-4) are publicly available and included herein as SEQ ID NOs: 16-21. In addition, the DNA and protein sequences for various ligands of Notch (including Jaggedl, Jagged2, Jagged2 (transcript variant 2), Deltal, Delta3, Delta4, and Jagged2 (transcript variant 1)) are publicly available and included herein as SEQ ID NOs: 22- 35, respectively. DNA and protein sequences for target Notch transcription factors Heyl, Hey2, and Hesl are also publicly available and are included herein as SEQ ID NOs: 36-41, respectively. EXAMPLE 6 SUPPRESSION OF PROX-1 mfflBITS GROWTH IN SOFT AGAR. Since anchorage-independent growth is one of the hallmarks of malignant transformation, experiments were conducted to assess the effects of Prox-1 suppression on the growth of SW480R cells in soft agar. SW480R cells were transfected with GFP siPRNA, Prox-lsiRNA A16 or Prox-1 siRNA A25 repeatedly over an 8-day period, or left untreated, and seeded in soft agar in triplicate. The number of colonies was scored after two weeks of growth. Transfection with both Prox-1 siRNAs but not the control GFP siRNA significantly decreased the number of colonies formed after two weeks of growth in soft agar (Fig. 3A). EXAMPLE 7 REGULATION OF PROSTAGLANDIN BIOSYNTHESIS BY PROX-1 COX-2 is a key enzyme involved in the conversion of arachidonic acid into the prostaglandin precursors PGG2 and PGH2, which are further transformed into biologically active prostaglandins by the action of conesponding synthases. Prostaglandins acts through binding to the G-protein coupled prostanoid receptors and they are rapidly inactivated by the action of 15-prostaglandin dehydrogenase (15- PGDH). COX-2 is overexpressed in the majority of colorectal cancers and in about half of colonic adenomas, suggesting that the increased PG production is important for tumor growth. In support of this view, treatment with non-steroid anti- inflammatory drugs, which acts as inhibitors of COX-2, significantly reduces the risk of developing colon cancer (Gupta, R. A. & Dubois, R. N., Nat Rev Cancer 1: 11-21, 2001). Accordingly, contemplated herein is a method of alleviating colorectal cancer via the administration of a Prox-1 suppressor in combination with a COX-2 inhibitor. Cox-2 inhibitors may include the following non-steroid anti-inflammatory drugs: asprin, rofecoxib, celecoxib ,amidophen, analgin, anapyrin, feloran, indomethacin, paracetoamol, pkoxicam, sedalgin, diclofenac sodium, ketoprofan, Acular®, Ocufen®, and Voltarol®. Experiments were conducted which found that suppression of Prox-1 in SW480R cells resulted in the up-regulation of the expression of 15-PGDH and downregulation of prostaglandin D2 synthase, whereas overexpression of Prox-1 in SW480F cells down-regulated 15-PGDH and up-regulated PGD2 synthase (Affymetrix results). These data suggest that Prox-1 may be important for the control of the balance of the total PG production in tumor cells, i.e., in the presence of Prox-1 decreased expression of 15-PGDH will result in higher net amounts of biologically active prostaglandins and enhanced tumor growth. Because SW480 cells do not express COX-2, contemplated herein are experiments to assess the effects of Prox-1 on prostanoid biosynthesis in the SW480F cells stably transfected with COX-2 or in the cell line which is known to express this enzyme, such as HCA-7. To generate COX-2 expressing cells, SW480F cells are transfected with a mixture of a COX-2 expressing vector and the plasmid bearing hygromycin resistance gene, such as pCDNA3.1hygro (Invitrogen) using Lipofectamine 2000, as described in Materials and Methods, and the stable clones are selected using 200 μg/ml hygromycin B (Calbiochem) over a period of 2-3 weeks. Individual clones are isolated and the expression of COX-2 protein is tested using Western blotting. Functionality of COX-2 may be further verified in COX-2 expressing clones in comparison to the control cells, using ELISA to monitor PGE2 production according to the manufacturer's instructions (Cayman Chemical). To test the effects of Prox-1 on prostaglandin biosynthesis, COX-2 expressing cells can be infected with AdProx-1 or the control AdGFP virus, as described previously (Petrova et ab, Embo J. 21: 4593-9), and the amount of biologically active PGE2, and total amount of metabolized PGE2 in cell conditioned medium, determined by ELISA (Cayman Chemicals). If overexpression of Prox-1 increases levels of the biologically active PGE2 in vitro, contemplated herein are studies to assess the link between Prox- 1 overexpression and prostanoid biosynthesis in vivo. SW480R or HCA-7 stably overexpressing 15-PGDH will be produced using the protocol described above, and the tumorigenic potential of these cells in nude mice will be determined. In addition, contemplated are studies regarding the effects of the treatment with 15-PGDH inhibitor on growth of Prox-1 expressing or control xenografts in nude mice. EXAMPLE 8 EFFECTS OF NOTCH SIGNAL TRANSDUCTION To investigate the effects of altered Notch signaling in SW480R cells described herein are experiments that overexpress constitutively active Notchl, Notch2, Notch3, and Notch4 intracellular domains, as well as Jagged 1, soluble Jaggedl, and Jagged2 using recombinant adenoviruses. Replication-deficient adenoviruses for the expression of constitutively active Notch 1-4 intracellular domains, and Notch ligands Jaggedl, Jagged2, Deltal, Delta3, Delta4, and Senate are produced. SW480R cells are infected with adenoviruses. 48-72 h postinfection cells are seeded in soft agar as described previously, and the number of colonies are scored after two weeks in culture. In parallel, total RNA is isolated and analysis of gene expression changes is conducted using Affymetrix® microaπay according to the previously described procedures. If overexpression of Notch or its ligand results in the inhibition of cell growth in soft agar, further studies are conducted to investigate the effects of activation of Notch signaling on growth of tumors in nude mice. EXAMPLE 9 EFFECTS OF PROX-1 SUPPRESSION ON SW480R IN NUDE MICE Also contemplated herein are studies to assess the effects of Prox-1 suppression on the growth of SW480R tumors in nude mice. Nu/nu mice can be inoculated subcutaneously or intraperitoneally with 1-5 x 106 cells/mice using SW480R cells transfected with GFPsi RNA or Prox-1 siRNA, or transduced with the adenoviruses described in Example 8. Tumors are allowed to grow for 3-5 weeks, and tumor size measured twice a week. Animals are sacrificed by cervical dislocation, tumors excised, and processed for immunohistoshemical staining. The tumor histology, expression of differentiation markers, proliferation index and vascularization momtored using the antibodies against KI67 (prohferation), mucin, galectin-2, p21cipl (differentiation), PECAM-1 and vWF (blood vessel markers), and the standard immunostaining protocols. To assess of the effects of Prox-1 -dependent genes, such as 15-PGDH, on prostaglandin metabolism and tumor growth in vivo, SW480R or HCA-7 cells recombinantly overexpressing 15-PGDH and control cells, are implanted subcutaneously into the nu/nu mice, and tumor growth and differentiation studied. In order to confirm the specificity of 15-PGDH effects, a subset of the control and 15- PGDH overexpressing tumor-bearing animals are treated with the 15-PGDH inhibitor CAY10397, administered intravenously, or in drinking water. EXAMPLE 10 ANALYSIS OFPROX-1 INNATURAL COLORECTAL TUMORS Experiments to asses the expression of Prox-1 in a mouse model of human famihal adenomatous polyposis, Ape min/+ are also contemplated herein. The Ape min/+ mice bear a truncating mutation in one allele of Ape gene, and develop multiple intestinal polyps, which further progress to adenocarcinoma. Mice are commercially available from JAX. As another cancer model, SMAD3 deficient mice, which develop invasive colorectal cancer, is available. The DNA and protein sequences for APC (SEQ ID NOs: 42 and 43, respectively) are published and disclosed as Genbank Accession Number NM_000038. Administration of a'Prox-1 inhibitor and a placebo to mice of the above-described models is also contemplated. Prox-1 inhibitors and administration thereof are described herein. Prox-1 inhibitors available for administration include, but are not hmited to, antisense oligonucleotides, siRNA constructs, or dominant negative proteins. Monitoring of the mice post-administration is contemplated to evaluate the effects of adenocarcinoma and colorectal cancer development and growth. Among the results are measurements of the speed of tumor growth in mice that received the Prox-1 inhibitor versus mice that received the placebo, thus, providing a beneficial efficacy model for the particular Prox-1 inhibitor. Also contemplated are methods for screening Prox-1 levels in family members with familial adenomatous polyposis. Methods for screening Prox-1 levels are described herein. Administration of a prophylactic to protect from progression, or the onset of cancer, is contemplated where elevated levels of Prox-1 are observed. EXAMPLE 11 DETECTION OF PROX-1 PROTEIN EXPRESSION IN COLORECTAL CANCER As described above, measuring Prox-1 protein expression in colon tissues may be a useful tool for diagnosing colon cancer and/or premalignancies. Prox-1 mRNA can be detected in colorectal cancer tissues as described in Example 2. The following prospective example may be conducted to determine whether Prox-1 protein conelates with Prox-1 transcript expression in colorectal cancer tissue. The immunohistochemical analysis can be carried out as follows using an anti-human Prox-1 antibody dkected against the human Prox-1 peptide, as described in Example 1. The tissues for screening are snap frozen in liquid nitrogen after dissection, embedded in OCT compound, and sectioned. Sections are fixed on -20°C methanol for 10 min, and processed for staining. To enhance epitope recovery, the tissues may undergo steam induced epitope recovery with a retrieval solution, including several different SHIER solutions with and without enzyme digestion at two different concentrations. The tissues can then be heated in the capillary gap in the upper chamber of a Black and Decker Steamer as described in Ladner et al. (Cancer Research, 60: 3493-3503, 2000). Automated immunohistochemistry is carried out with the TECHMATE 1000 or TECHMATE 500 (BioTek Solutions, Ventura Medical System). Specifically, the tissues are blocked with 3% and 10 % normal goat serum for 15 and 30 minutes respectively. Subsequently, the tissues are incubated with the primary antibody (anti-Prox-1 antibody) for 60 minutes at 3.0 *g/ml. The tissues are stained with the biotinylated goat-anti-rabbit IgG secondary antibody for 25 minutes. Optimal results are obtained with overnight incubation. To ensure the staining procedure is working appropriately, anti- vimentin is used as a positive control and rabbit IgG is used as a negative control. The antibody binding is detected by an avidin-biotin based tissue staining system where horse-radish peroixidase is used as a reporter enzyme and DAB (3,3'-Diaminobenzididine Tetrahydrochloride) is used as a chromogen. Specifically, the endogenous peroxides are blocked for 30 minutes, the avidin-biotin complex reagent is added and then the tissues are incubated in DAB for a total of 15 minutes. Finally, the tissues are counterstained with hemotoxylin to assess cell and tissue morphology. The slides are mounted in Aquamount, and the tissues are examined visually under a light microscope. Tissue that is positive for increased Prox-1 protein expression as compared to healthy colon tissue, or other cancer tissues, indicate colorectal cancer and/or premalignant lesions. While this prospective example provide one means of detecting colon cancer, other means will be obvious to those with skill in the art. Various options for detecting Prox-1 expression, and, therefore screen for colon cancer, include, among others, ELISA-based techniques and Western blotting techniques. EXAMPLE 12 EXPRESSION PATTERN OF PROX-1 IN NORMAL COLONIC EPITHELIUM Studies were conducted to compare Prox-1 expression in normal cololnic epithelium. In normal colonic mucosa, all Prox-1 expressing cells were positive for the intestinal epithelial transcription factor CDX2. There was no overlap with the expression of MUC2, expressed by the goblet cells; however, a subset of Prox-1 positive cells also expressed the pan-neuroendocrine marker chromogranin A. Also observed was weaker but significant Prox-1 expression in the bottom of the crypts below the cell prohferation zone identified by staining for the Ki67 antigen. Colonic epithelium is composed of the slowly dividing stem cells located in the bottom of the crypt, the cell proliferation zone with transient amplifying cells, which give rise to the three main colonic epithelial cell types, and terminally differentiated cells, located in the upper part of the crypts. The location of Prox-1 positive cells at the base of the crypts, therefore, conesponded to the position of the intestinal stem cells. (Bach, S. P., Renehan, A. G. & Potten, C. S., Carcinogenesis 21, 469-76 (2000); Potten, C. S., Kellett, M., Roberts, S. A., Rew, D. A. & Wilson, G. D., Gut 33, 71-8 (1992)) A similar staining pattern was observed in the murine descending colon, whereas the duodenal epithelium was negative for Prox-1. Expression of p2iCIP1/WAF1 marks the differentiated compartment of colonic crypts independently of the cell type (Doglioni, C. et ab, J Pathol 179, 248-53 (1996)). Accordingly, studies were conducted regarding the expression of Prox-1 in relation to p21cπ>1 WAF1. All Prox-1 positive cells located at the bottom of the crypts were negative for p21CIP1/WAF1; however, most of the rare Prox-1 positive cells present in the upper parts of the crypts were also negative for ρ21cπ>1 WAF1, demonstrating a mutually exclusive relation between Prox-1 expression and terminal differentiation. p21(CIPl)/(WAFl) (CDKNl) sequences are published and disclosed as Genbank Accession Numbers NM_078467 and NM_000389. These variants (1) and (2) encode the same protein. Based on the data implicating Prospero/Prox- 1 in cell fate determination in other cell types, and on its expression pattern in colonic epithelial cells it is contemplated that Prox-1 may be involved in the regulation of the neuroendocrine cell fate as well as the stem cell phenotype. This hypothesis is supported by the fact that PROX-1 is overexpressed in intestinal neoplasms from Apcπun/+ mice and that its expression is regulated by TCF/β-catenin pathway in vitro (see Examples 13 and 14). This hypothesis is also in agreement with previous results showing that targeted inactivation of Tcf712 gene encoding TCF-4 leads to the depletion of intestinal stem cell compartment and loss of neurodendocrine lineage (Korinek, V. et al., Nat Genet 19, 379-83 (1998)). EXAMPLE 13 PROX-1 IS OVEREXPRESSED IN INTESTINAL NEOPLASMS FROM APC"11117* MICE BUT NQT FROM Ltbp4 -/- DEFICIENT MICE Studies were also conducted to assess Prox-1 expression in Apcιmπ/+ mice. A truncating germline mutation in the Ape gene together with somatic inactivation of the remaining wild type allele, lead to abnormal β-catenin/TCF signaling in intestinal epithelial cells of Apcπun/+ mice and development of multiple intestinal polyps (Luongo, C, Moser, A. R., Gledhilb S. & Dove, W. F., Cancer Res 54, 5947-52 (1994); Su, L. K. et al., Science 256, 668-70 (1992)). High levels of Prox-1 in intestinal neoplasms of Apcπun + mice were observed. Prox-1 mRNA and protein were present in tumor cells with high cytoplasmic and nuclear β-catenin levels, but not in the differentiating cells of the neighboring normal glands with membrane localization of β-catenin. Mutation in genes regulating TGFβ signaling pathway, such as TGFRJJ and SMAD4 occur in human colorectal cancer, and targeted inactivation of TGF-βl binding protein LTBP-4 leads to colon cancer in mice (White, R. L., Cell 92, 591-2 (1998); Sterner-Kock, A. et al., Genes Dev. 16, 2264-73 (2002)). Studies were conducted to assess Prox-1 expression in Ltpb4J- mice. In contrast to the results from Ape nιin/+, accumulation of Prox-1 in the colonic adenocarinomas from Ltpb4- /- mice, which generally preserve normal distribution of β-catenin, was not observed. These results strongly suggest that Prox-1 is a target of APC/ β-catenin/TCF pathway in vivo. Tumors from Ltpb4-/- mice had strongly increased number of lymphatic vessels, positive both for Prox-1 and LYVE-l. EXAMPLE 14 PROX-1 EXPRESSION IS REGULATED BY β-CATENIN/TCF PATHWAY AND DNA METHYLATION Further studies were conducted using SW480R cell line as an in vitro model to investigate the role of Prox-1 in colorectal carcinoma. Suppression of Prox-1 expression using two different siRNAs (SEQ ID NOS: 4, 5, 6, and 7) did not affect the activity of a β-catenin/TCF-responsive reporter, the nuclear localization of β- catenin, or the cellular content of active, non-phosphorylated β-catenin, confirming that Prox-1 is not acting upstream of this pathway, ha contrast, suppression of β- catenin using two independent siRNAs resulted in almost complete disappearance of Prox-1 mRNA and protein. In line with this finding, suppression of Prox-1 was also observed in SW480R cells transfected with dominant negative mutant of TCF4, which disrupts β-catenin/TCF mediated transcription (Morin PJ, et al., Science 1997 Mar 21 ;275(5307): 1787-90). However, overexpression of p21cπ>1 AF1, shown to induce cell differentiation in colorectal carcinoma cells (van de Wetering, M. et al., Cell 111, 241-50 (2002)), did not modify Prox-1 levels. Taken together, these data show that Prox-1 lies downstream of β-catenin/TCF4 and upstream of p21CIP1 AF1. Also observed was increased expression of several known β- catenin/TCF-4 target genes, such as CD44, ENC1 and 1D2 in the absence of Prox-1 (Table H, (Fujita et al, 2001 ; Rockman et al, 2001 ; Wielenga et al, 1999)), while others such as p21CIP1/WAF1, annexin Al, and OB-cadherin were induced upon suppression of either β-catenin or Prox-1. These results underline the complexity of the regulatory cascade initiated by β-catenin/TCF in CRC cells and suggest that concerted regulation by Prox-1 and other β-catenin/TCF targets is necessary for. neoplastic growth. Studies were also conducted to compare the activation of β- catenin/TCF signaling pathway in SW480R and SW480A cells. The SW480R cells had slightly more active β-catenin and displayed a two-fold increase in the activation of the TCF-responsive promoter TopFLASH; however, both cell lines clearly displayed nuclear localization of β-catenin as previously reported (Palmer, H. G. et al., / Cell Biol 154, 369-87 (2001)). These observations, together with the fact that abnormal β-catenin/TCF pathway signaling is a feature of the majority of colorectal cancer cell lines, suggest that β-catenin/TCF activation is necessary but not sufficient for the induction of Prox-1 expression in colorectal cancer. DNA methylation is frequently abnormal in colorectal cancer, and it was reported recently that Prox-1 expression is suppressed in human hematological cell lines due to hypermethylation of CpG islands in intron 1 of Prox-1 (Nagai, H. et al., Genes Chromosomes Cancer 38, 13-21 (2003)). Treatment of SW480A cells with the inhibitor of DNA methyltransferases 5-aza-2' -deoxycytidme did not result in the increase of Prox-1 RNA, while there was increase in the expression of TJMP3. In contrast, 5-aza-2' -deoxycytidme almost completely suppressed Prox-1 expression in SW480R cells, suggesting that, at least in this cell type, the regulation of Prox-1 by DNA methylation is opposite to the one observed in leukemic cells. Our finding that DNA demethylation decreases Prox-1 mRNA levels suggests the existence of a putative suppressor of Prox-1 transcription, whose expression becomes relieved upon treatment with 5-aza-2' -deoxycytidme. Since 5- aza-2' -deoxycytidme is used for the treatment of human cancers, our data also suggest that Prox-1 could be used as marker to identify the colorectal tumors which would respond favorably to this drug. Such screening of patients/tumors is intended as an aspect of the invention. The role of DNA methylation in the growth of intestinal neoplasms was previously demonstrated in mice heterozygous or hypomorphic for DNA methyltransf erase 1 , a major enzyme involved in the methylation of DNA.
These mice do not develop intestinal adenomas when crossed with Apcm /+ mice. In contrast, they develop lymphomas, demonstrating cell type specific effects of decreased DNA methylation for cancerous growth (Gaudet, F. et al., Science 300, 489-92 (2003), Eads, C. A. et ab, Cancer Res 62, 1296-9 (2002)). EXAMPLE 15 PROX-1 SUPPRESSION AND OVEREXPRESSION IN COLORECTAL CANCER To characterize the effects of Prox-1 suppression and overexpression in colorectal cancer, stable colorectal cancer cell hne clones inducibly expressing Prox-1 or Prox-1 targeting siRNAs are employed. Cells are implanted into laboratory animals, such as nu/nu mice, and tumor growth is studied in control mice and mice treated with doxycyctine. As an alternative approach, Prox-1 or Prox-1 siRNA expressing lentivkuses are employed to provide long-term expression in colorectal cancer cell lines in vitro and in vivo. To inducibly suppress and overexpress Prox-1 or Prox-1 siRNAs, Prox-1 cDNA was subcloned in pTetOS vector (Sarao and Dumont, Trans enics Res., 1998) , where it is placed under the control of doxycycline regulated promoter. Prox-1 siRNAs were subcloned in pTer vector (van der Wetering et al., Embo Reports, 2003). Colorectal carcinoma cells stably expressing tTA activator may be transfected with Prox-1/TetOS or Prox-1 siRNS/pTer vectors. Clones may be selected in the presence of blasticidine and G480 and further tested for the expression of Prox-1 by immunostaining or Prox-1 siRNA by suppression of co-transfected Prox- 1 in the presence of doxycycline. For production of Prox-1 lentiviruses, Prox-1 cDNA was subcloned into FUkesGFPW (Lois et al., Science , 2002). For production of Prox-1 siRNA lentiviruses, Prox-1 siRNAs 1 and 2 were subcloned into lentiviral vector pLL3.7 (Rubinson et ab, Nat Genet., 2003). Sequences of the DNA oligos used in the cloning of pLL3J-Prox-l : sense: TGGTCATCTGCAAGCTGGATTTCAAGAGAATCCAGCTTGCAG ATGACCTTTTTC (SEQ ID NO 47). antisense: TCGAGAAAAAAGGTCATCTGCAAGCTGGATTCTCTTGAAATCCAGCTTGC AGTGACCA (SEQ ID NO 48). pLL3J PROX1-2: sense: TGAGCCAGTTTGATATGGATTTCAAGAGAATCCATATCAAACTGGCTCTTT TTTC (SEQ ID NO 49). antisense: TCGAGAAAAAAGAGCCAGTTTGATATGGATTCTCTTGAAAT CCATATCAAACTGCTCA (SEQ ID NO 50).
Inducible Prox-1 targeting short hairpin RNA ("shRNA") expression may also be achieved via CRE recombinase activated induction system whereby an inactivating stuffer DNA sequence sunounded by modified loxP sites is removed from an shRNA expression cassette by the CRE recombinase activity, thus activating the shRNA expression. Alternatively a similar system may be used to inactivate shRNA expression upon introduciton of CRE recombinase. Tiscornia et al PNAS 2004, and Coumoul et al NAR 2004) described these systems. shRNA or "short hairpin RNA" is a short sequence of RNA which makes a tight hairpin turn and can be used to silence gene expression. This small hairpin RNA was first used in a lentiviral vector. (Abbas-Terki T. et al, Hum. Gene Ther. 13(18):2197-201 (2002)). shRNA generates siRNA in cells (An DS et &\., Hum. Gene Ther. 14(12): 1207-12 (2003)). To study the effects of Prox-1 overexpression in vivo, transgenic mice overexpressing Prox-1 under the control of intestinal-specific promoter, such as villin, CyplA or FABPi are created using standard techniques. The proliferation and differentiation status of intestinal epithelial cells is studied by staining of intestinal tissues for PCNA, Ki67, CDKNIA, mucins, lysozyme, chromogranin A and carboxipeptidases II and IV. The crossing of Prox-1 transgenic animals with Apc1"111^ mice permits determination of whether Prox-1 overexpression influences the number and size of intestinal polyps in this mouse model of colorectal cancer. Specifically, for in vivo studies of Prox-1 in intestinal differentiation, Prox-1 cDNA was subcloned in pl2.4Vill plasmid, which places it under the control of 12.4 kb mous villin promoter (Madison et al., J.Biol.Chem.2002, genomic contig NT_039170). The construct may be used for the production of villin- Prox-1 transgenic mice, which will overexpress Prox-1 at the sites of villin expression, i.e. intestinal epithelial cells. Also contemplated is subcloning Prox-1 cDNA into the vector z/AP (Lobe et al., Dev. Biol, 1999), to be able conditionally express Prox-1 in any given tissue. In this approach Prox-1 cDNA is placed between the loxP sites, and it is not expressed until Cre recombinase is present in the same cell. Excision of loxP sites places the transgene under the control of chicken β-actin promoter . To achieve intestinal specific overexpression of Prox-1 the transgenic animals containing z/AP- Prox-1 expression cassettes in thek genomes may be crossed with villin-Cre mice (Madison et al., J.Biol.Chem.2002). The latter approach may be preferable to the villin-PROXl overexpression because of potentially higher expression levels of the transgene. Also contemplated in cloning Prox-1 cDNA under the control of rat Fabpi promoter (Rottman and Gordon, J. Biol. Chem., 1993, genomic contig NW_047627) or CyplA promoter (Sansom et ab, Genes Dev., 2004, genomic contig NT_039474). The latter promoter has an advantage of being inducible upon administration of β- naphtoflavone. All of these transgenic mice are contemplated as aspects of the invention. EXAMPLE 16 DOMINANT NEGATIVE MUTANTS OF PROX-1 Further contemplated herein are dominant negative mutants of Prox-1. Specifically, a Prox-1 mutant protein lacking the transactivation domains or DNA binding domains may act in a dominant negative manner. Experiments to investigate this hypothesis may be conducted by producing a truncated form of Prox-1 lacking the last 60 amino acids or the first 575 amino acids. Disruption of the DNA binding domain entails truncation of the protein to exclude amino acids 572-634 of SEQ ID NO. 3, based on homology to Prospero (Drosophila). Disruption of the transactivation domain entails the deletion of amino acids 635-737. These proteins may then be tested for their ability to repress the induction of Prox-1 target genes upon co-transfection with the wt Prox-1. If such an effect is observed, the construct can be used for the generation of transgenic animals with the purpose of suppression of Prox-1 effects in vivo, or for the anti- Prox-1 therapies in colorectal cancer. The foregoing examples are intended to be illustrative of the invention and not intended to limit the claims which define the invention. All patent, journal, and other literature cited herein is incorporated herein by reference in the entirety. While the invention is described specifically with respect to Prox-1, there are other genes described in tables herein that are differentially expressed. All materials and methods described herein are applicable to the genes described in the tables.

Claims

Claims: 1. A method of screening colon tissue for a pathological condition, said method comprising: measuring Prox-1 expression in a biological sample that comprises colon tissue from a mammalian subject, wherein elevated Prox-1 expression in the colon tissue conelates with a pathological phenotype.
2. A method according to claim 1, comprising comparing Prox-1 expression in the colon tissue to Prox-1 expression in healthy colon tissue, wherein increased Prox-1 expression in the colon tissue from the mammalian subject conelates with a pathological phenotype.
3.. A method according to claim 1 or 2, further comprising a step, prior to said measuring step, of obtaining the biological sample comprising colon tissue from a mammalian subject.
4. The method according to any one of claims 1-3, wherein the pathological condition is colon cancer, and wherein increased Prox-1 expression in the colon tissue is indicative of a cancerous or precancerous condition.
5. The method according to any one of claims 1-4, wherein the measuring comprises measuring Prox-1 protein in the biological sample.
6. The method of claim 5, wherein the measuring comprises contacting the colon tissue with a Prox-1 antibody or antigen-binding fragment thereof.
7. The method of any one of claims 1-6, wherein the measuring comprises measuring Prox-1 mRNA in the colon tissue.
8. The method of claim 7, wherein the measuring comprises in situ hybridization to measure Prox-1 mRNA in the colon sample.
9. The method of claim 7, wherein the measuring comprises steps of isolating mRNA from the colon tissue and measuring Prox-1 mRNA in the isolated mRNA. . . . .
10. The method according to any one of claims 1-9, wherein the measuring comprises quantitative polymerase chain reaction (PCR) to quantify Prox-1 mRNA in the colon tissue relative to Prox-1 mRNA in healthy colon tissue.
11. A method according to any one of claims 1-10, further comprising measuring expression of at least one gene selected from the group consisting of CD44, Encl, and 1D2 in the colon tissue, wherein elevated Prox-1 expression and elevated expression of the at least one gene in the colon tissue conelate with a pathological phenotype.
12. A method according to any one of claims 1-11, further comprising measuring activation of -catenin/TCF pathway in the colon tissue, wherein activation of the -catenin/TCF pathway and elevated Prox-1 expression in the colon tissue conelate with a pathological phenotype.
13. A method according to claim 12, wherein activation of the - catenin/TCF pathway is measured by at least one indicator in the colon tissue selected from the group consisting of: mutations in an APC gene; mutations in a -catenin gene; and nuclear localization of -catenin.
14. The method according to any one of claims 1-13, wherein the mammalian subject is a human.
15. A method according to claim 14, further comprising a step of administering to a human subject identified as having a pathological condition characterized by increased Prox-1 expression in colon tissue a composition comprising a Prox-1 inhibitor.
16. Use of a molecule that suppresses expression or activity of Prox-1 in the manufacture of a medicament for the treatment of colorectal cancer.
17. A method of inhibiting the growth of colorectal cancer cells in a mammalian subject comprising the step of: administering to the subject a composition comprising a molecule that suppresses expression or activity of Prox-1, thereby inhibiting the growth of colon carcinoma cells.
18. A method or use according to claim 16 or 17, wherein the molecule suppresses Prox-1 expression.
19. A method or use according to any one of claims 16-18, comprising a step of identifying a mammalian subject with a colon cancer characterized by increased Prox-1 expression, wherein the composition is administered after the identifying step.
20. A method or use according to any one of claims 16-19, wherein the cancer is selected from a colorectal adenoma and a colorectal carcinoma.
21. The method or use according to any one of claims 16-20, wherein the composition further comprises a pharmaceutically acceptable diluent, adjuvant, or carrier medium.
22. The method or use according to any one of claims 16-21, wherein the molecule comprises an antisense oligonucleotide that inhibits Prox-1 expression.
23. The method or use according to any one of claims 16-21, wherein the molecule comprises micro-RNA that inhibits Prox-1 expression.
24. The method or use according to any one of claims 16-21, wherein the molecule comprises short interfering RNA (siRNA) that inhibits Prox-1 expression.
25. The method or use of claim 24, wherein the siRNA comprises at least one nucleotide sequence set forth in SEQ JJ) NOS: 4, 5, 6, and 7.
26. The method or use according to any one of claims 16-21 , wherein the molecule comprises a zinc finger protein that inhibits Prox-1 expression.
27. The method or use according to any one of claims 16-21, wherein the molecule comprises a dominant negative form of Prox-1 protein, or an expression vector containing a nucleotide sequence encoding the dominant negative Prox-1 protein.
28. The method or use of claim 27, wherein the dominant negative form of Prox-1 protein has a disrupted DNA binding domain.
29. The method or use of claim 27, wherein the dominant negative form of Prox-1 protein has a disrupted transactivation domain.
30. The method or use according to any one of claims 16-21 , wherein the molecule comprises short hairpin RNA (shRNA) that inhibits Prox-1 expression.
31. The method according to any one of claims 17-30, wherein the composition is administered in an amount effective to suppress Prox-1 expression and increase Notch 1 signaling.
32. The use according to any one of claims 16-30, wherein the molecule is present in the composition in an amount effective to suppress Prox-1 expression and increase Notch-1 signaling.
33. The method according to any one of claims 17-31, wherein the composition is administered in and amount effective to increase 15-PDGH activity or decrease prostaglandin D2 synthase activity.
34. The method according to any one of claims 17-31, further comprising administering to the subject an inhibitor of the -catenin/TCF signaling pathway.
35. The use according to any one of claims 16-30, wherein the medicament further includes an inhibitor of the -catenin/TCF signaling pathway.
36. The method or use of claim 34 or 35, wherein the inhibitor of the -catenin TCF signahng pathway is dominant negative form of TCF-4.
37. The method or use of claim 34 or 35, wherein the inhibitor of the -catenin/TCF signaling pathway targets TCF-4, β-catenin, or c-myc.
38. The method according to any one of claims 17-31, further comprising admimstering to the subject a COX-2 inhibitor.
39. The use according to any one of claims 16-30, wherein the medicament further includes a COX-2 inhibitor.
40. The method or use of claim 38 or 39, wherein the COX-2 inhibitor is a non-steroid anti-inflammatory drug.
41. The method according to any one of claims 17-31, further comprising administering to the subject a Notch signaling pathway agonist.
42. The use according to any one of claims 16-30, wherein the medicament further includes a Notch signahng pathway antagonist.
43. The method or use according to claim 41 or 42, wherein the Notch signaling pathway agonist is a Notch ligand.
44. The method or use of claim 43, wherein the Notch ligand is
Jaggedl, Jagged2, Deltal, Delta3, Delta4, or Senate.
45. The method or use of claim 41 or 42, wherein the Notch signahng pathway agonists are Notch targets Heyl, Hey2, or Hesl.
46. A method of inhibiting Prox-1 function in a mammalian subject having a disease characterized by Prox-1 overexpression in cells, comprising the step of administering to said mammalian subject a composition, said composition comprising a compound effective to inhibit Prox-1 function in cells.
47. Use of an inhibitor of Prox-1 function in mammalian cells for the manufacture of a medicament for inhibiting Prox- 1 function.
48. A method of screening for a Prox-1 modulator, comprising steps of: contacting a test molecule with Prox-1 protein, or a nucleic acid comprising a nucleotide sequence that encodes Prox-1 protein, under conditions which permit the interaction of the test molecule with the Prox-1 protein or nucleic acid; and measuring interaction between the test molecule and Prox-1 protein or nucleic acid, wherein a test molecule that binds the Prox-1 protein or nucleic acid is identified as a Prox-1 modulator.
49. The method of claim 48, wherein the test molecule comprises a protein, a carbohydrate, a lipid, or a nucleic acid.
50. The method of claim 48, wherein the test molecule comprises a member of a chemical library.
51. The method of any one of claims 48-50, comprising measuring the binding between the test molecule and the DNA binding domain of Prox-1.
52. A method of screening for modulators of binding between a DNA and Prox-1 protein comprising steps of: a) contacting a DNA with a Prox-1 protein in the presence and in the absence of a putative modulator compound; b) detecting binding between the DNA and the Prox-1 protein in the presence and absence ofthe putative modulator compound; and c) identifying a modulator compound based on a decrease or increase in binding between the DNA and the Prox-1 protein in the presence of the putative modulator compound, as compared to binding in the absence of the putative modulator compound.
53. A method of screening for modulators of binding between a DNA and Prox-1 protein comprising steps of: a) contacting a DNA with a Prox-1 protein in the presence and in the absence of a putative modulator compound; b) detecting binding between the DNA and the Prox-1 protein in the presence and absence of the putative modulator compound; and c) identifying a modulator compound based on a decrease or increase in differentiation in the presence of the putative modulator compound, as compared to differentiation in the absence of the putative modulator compound.
54. A method according to any one of claims 48-53, further comprising steps of: contacting a cell from a colorectal cancer or colorectal cancer cell line with the Prox-1 modulator; and selecting a Prox-1 modulator that inhibits growth of the cell.
55. A method according to claim 54, further comprising: formulating a composition comprising the selected Prox-1 modulator and a pharmaceutically acceptable carrier; administering the composition to a mammalian subject having a colorectal cancer; and monitoring the mammalian subject for growth, metastasis, shrinkage, or disappearance of the colorectal cancer.
56. A small interfering RNA (siRNA) molecule that comprises a sense region and an antisense region, wherein said antisense region comprises sequence complementary to a nucleotide sequence encoding Prox-1 set forth as SEQ ID NO: 2, or a fragment thereob and wherein the sense region comprises sequence complementary to the antisense region, or a fragment thereob
57. The siRNA molecule of claim 56, wherein said siRNA molecule comprises two nucleic acid fragments, wherein one fragment comprises the sense region and the second fragment comprises the antisense region.
58. The siRNA molecule of claim 57, wherein said sense region comprises a 3 '-terminal overhang relative to the antisense region.
59. The siRNA molecule of claim 57 or 58, wherein the antisense region comprises a 3'-terminal overhang relative to the sense region.
60. The siRNA molecule of claim 59, wherein said 3'-terminal overhangs each comprise 1-5 nucleotides.
61. The siRNA molecule of claim 59, wherein said antisense region 3'-terminal nucleotide overhang is complementary to RNA encoding Prox-1.
62. The siRNA molecule according to any one of claims 56-61, wherein said complementary sequences are 18-100 nucleotides in length.
63. The siRNA molecule according to any one of claims 56-61, wherein said complementary sequences are 18-30 nucleotides in length.
64. The siRNA molecule according to any one of claims 56-61, wherein said complementary sequences are 21-23 nucleotides in length.
65. The siRNA molecule according to any one of claims 56-61, wherein said antisense region comprises sequence complementary to sequence having any of SEQ ID NOs. 4 and 6.
66. The siRNA molecule according to any one of claims 56-61, wherein said antisense region comprises sequence having any of SEQ TJ) NOs. 5 and 7.
67. The use of an siRNA molecule according to any one of claims 56-66 in the manufacture of a medicament for the treatment of colorectal cancer.
68. The use according to claim 16, wherein the molecule comprises a compound comprising a nucleic acid 8 to 50 nucleotides in length, wherein said compound specifically hybridizes with a polynucleotide encoding Prox-1, or hybridizes to the complement of the polynucleotide, and inhibits the expression of Prox-1 when inkoduced into a cell that expresses Prox-1.
69. The use of claim 68, wherein the compound is an antisense oligonucleotide.
70. The use of claim 69, wherein the antisense oligonucleotide has a sequence complementary to a fragment of SEQ DD NO: 1.
71 The use of claim 70, wherein the fragment of SEQ ID NO: 1 comprises a promoter or other control region, an exon, an intron, or an exon-intron boundary.
72. The use of claim 70, wherein the fragment of SEQ ID NO: 1 comprises an exon-intron splice junction.
73. The use of claim 70, wherein the fragment of SEQ ID NO: 1 comprises a region within 50-200 bases of an exon-intron splice junction.
74. The method or use according to any one of claims 16-21, wherein the molecule comprises an inhibitor of DNA methyltransferases, thereby inhibiting Prox-1 expression.
75. The method or use according to claim 74, wherein the inhibitor of DNA methyltransferases is 5-aza-2' -deoxycytidme.
76. The method according to any one of claims 22-31 , further comprising admimstering to the subject an inhibitor of DNA methyltransferases.
77. The use according to any one of claims 22-30, wherein the medicament further includes an inhibitor of DNA methyltransferases.
78. The method or use of claim 76 or 77, wherein the inhibitor of DNA methyltransferases is 5-aza-2'-deoxycytidine.
PCT/EP2004/008819 2003-08-08 2004-08-06 Materials and methods for colorectal cancer screening, diagnosis, and therapy WO2005014854A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP04763854A EP1654383A1 (en) 2003-08-08 2004-08-06 Materials and methods for colorectal cancer screening, diagnosis, and therapy
US10/567,630 US8029984B2 (en) 2003-08-08 2004-08-06 Materials and methods for colorectal cancer screening, diagnosis and therapy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US49422103P 2003-08-08 2003-08-08
US60/494,221 2003-08-08

Publications (1)

Publication Number Publication Date
WO2005014854A1 true WO2005014854A1 (en) 2005-02-17

Family

ID=34135330

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2004/008819 WO2005014854A1 (en) 2003-08-08 2004-08-06 Materials and methods for colorectal cancer screening, diagnosis, and therapy

Country Status (3)

Country Link
US (1) US8029984B2 (en)
EP (1) EP1654383A1 (en)
WO (1) WO2005014854A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006092062A1 (en) * 2005-03-04 2006-09-08 The Hospital For Sick Children Methods for cancer prognosis
WO2008048680A2 (en) * 2006-10-19 2008-04-24 Maine Medical Center Research Institute Notch 2 signaling as a breast cancer suppressor pathway
EP3705137A4 (en) * 2017-11-03 2021-06-23 Korea Institute of Science and Technology Composition for regulating expression of prox1 or expression of telomerase reverse transcriptase comprising activity regulator or method for screening telomerase reverse transcriptase regulator

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7642091B2 (en) 2005-02-24 2010-01-05 Jau-Nan Lee Human trophoblast stem cells and use thereof
US8105777B1 (en) 2008-02-13 2012-01-31 Nederlands Kanker Instituut Methods for diagnosis and/or prognosis of colon cancer
CA2715518A1 (en) * 2009-09-23 2011-03-23 Wilson Roa Microrna expression profiles associated with lung cancer
WO2011128886A1 (en) 2010-04-12 2011-10-20 Ramot At Tel Aviv University Ltd. A micro-rna for cancer diagnosis, prognosis and therapy
US9574173B2 (en) * 2010-11-15 2017-02-21 Accelerated Biosciences Corp. Generation of neural stem cells from human trophoblast stem cells
WO2012135824A2 (en) * 2011-03-31 2012-10-04 H. Lee Moffitt Cancer Center And Research Institute, Inc. Method of screening for colon cancer using biomarkers
WO2013092960A1 (en) * 2011-12-22 2013-06-27 Fundacion Gaiker Methods and kits for the diagnosis of colorectal cancer
CA2865404C (en) 2012-02-24 2019-08-27 Stem Centrx, Inc. Dll3-binding antibodies and drug conjugates thereof to treat cancer
EP3591036B1 (en) 2012-11-30 2023-03-22 Accelerated BioSciences Corp. Methods of differentiating stem cells by modulating mir-124
PL2958944T3 (en) 2013-02-22 2019-09-30 Abbvie Stemcentrx Llc Antidll3-antibody-pbd conjugates and uses thereof
PE20160674A1 (en) 2013-08-28 2016-07-21 Stemcentrx Inc METHODS OF CONJUGATION OF SITE-SPECIFIC ANTIBODIES AND COMPOSITIONS
WO2015127407A1 (en) 2014-02-21 2015-08-27 Stemcentrx, Inc. Anti-dll3 antibodies and drug conjugates for use in melanoma
GB2548059B (en) 2014-11-26 2020-09-02 Accelerated Biosciences Corp Induced hepatocytes and uses thereof
JP7398226B2 (en) 2019-09-03 2023-12-14 浜松ホトニクス株式会社 Methods for assessing the risk of developing colorectal cancer

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003027285A1 (en) * 2001-09-27 2003-04-03 Bionomics Limited Dna sequences for human angiogenesis genes
US20030087807A1 (en) * 1999-12-02 2003-05-08 Neurosciences Research Foundation, Inc. Methods for identifying compounds for motion sickness, vertigo and other disorders related to balance and the perception of gravity

Family Cites Families (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5116742A (en) * 1986-12-03 1992-05-26 University Patents, Inc. RNA ribozyme restriction endoribonucleases and methods
US4987071A (en) * 1986-12-03 1991-01-22 University Patents, Inc. RNA ribozyme polymerases, dephosphorylases, restriction endoribonucleases and methods
US5254678A (en) * 1987-12-15 1993-10-19 Gene Shears Pty. Limited Ribozymes
US4932207A (en) 1988-12-28 1990-06-12 Sundstrand Corporation Segmented seal plate for a turbine engine
US5225337A (en) * 1989-09-25 1993-07-06 Innovir Laboratories, Inc. Ribozyme compositions and methods for use
ATE139258T1 (en) * 1990-01-12 1996-06-15 Cell Genesys Inc GENERATION OF XENOGENE ANTIBODIES
US6004924A (en) * 1991-12-11 1999-12-21 Imperial Cancer Research Technology, Ltd. Protein sequences of serrate gene products
US5786158A (en) * 1992-04-30 1998-07-28 Yale University Therapeutic and diagnostic methods and compositions based on notch proteins and nucleic acids
JPH08500481A (en) 1992-05-11 1996-01-23 リボザイム・ファーマシューティカルズ・インコーポレーテッド Methods and agents for inhibiting viral replication
SG48760A1 (en) 1992-07-24 2003-03-18 Abgenix Inc Generation of xenogenetic antibodies
US5750652A (en) * 1994-01-21 1998-05-12 Yale University Deltex proteins
US7625697B2 (en) * 1994-06-17 2009-12-01 The Board Of Trustees Of The Leland Stanford Junior University Methods for constructing subarrays and subarrays made thereby
EP0822830B1 (en) 1995-04-27 2008-04-02 Amgen Fremont Inc. Human anti-IL-8 antibodies, derived from immunized xenomice
EP0823941A4 (en) 1995-04-28 2001-09-19 Abgenix Inc Human antibodies derived from immunized xenomice
ES2334953T3 (en) * 1995-06-28 2010-03-17 Imperial Cancer Research Technology Limited SEQUENCES OF NUCLEOTIDES AND PROTEINS OF DELTA GENES OF VERTEBRATES AND METHODS BASED ON THE SAME.
US5780300A (en) * 1995-09-29 1998-07-14 Yale University Manipulation of non-terminally differentiated cells using the notch pathway
CA2256481A1 (en) * 1996-05-31 1997-12-04 The National American Red Cross Therapeutic and diagnostic methods and compositions based on jagged/notch proteins and nucleic acids
JP3922726B2 (en) * 1996-07-16 2007-05-30 旭化成株式会社 Differentiation inhibitor
US6121045A (en) * 1997-04-04 2000-09-19 Millennium Biotherapeutics, Inc. Human Delta3 nucleic acid molecules
US6436650B1 (en) * 1997-07-23 2002-08-20 Yale University Activated forms of notch and methods based thereon
JP2002523018A (en) 1998-07-27 2002-07-30 アムジエン・インコーポレーテツド Delta-related polypeptide
US20030224516A1 (en) * 2002-06-03 2003-12-04 Isis Pharmaceuticals Inc. Antisense modulation of prox-1 expression
EP1147189A2 (en) 1998-12-04 2001-10-24 Immusol, Inc. Ribozyme therapy for the treatment and/or prevention of restenosis
US20020192665A1 (en) 1999-06-01 2002-12-19 Zoghbi Huda Y. Compositions and methods for the therapeutic use of an atonal-associated sequence for a gastrointestinal condition
AU6913100A (en) 1999-08-19 2001-03-13 Chiron Corporation Notch receptor ligands and uses thereof
US6380362B1 (en) * 1999-12-23 2002-04-30 Genesis Research & Development Corporation Ltd. Polynucleotides, polypeptides expressed by the polynucleotides and methods for their use
AU2001292750B2 (en) 2000-09-22 2005-07-14 Genentech, Inc. Notch receptor agonists and uses
AU2003217966A1 (en) * 2002-03-07 2003-10-08 Licentia, Ltd. Lymphatic and blood endothelial cell genes
US20050271636A1 (en) * 2002-08-09 2005-12-08 St. Jude Children's Research Hospital, Inc. Diagnostic and therapeutic uses for prox 1
US20070110744A1 (en) * 2003-09-08 2007-05-17 Kari Alitalo Lymphatic and blood endothelial cell genes

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030087807A1 (en) * 1999-12-02 2003-05-08 Neurosciences Research Foundation, Inc. Methods for identifying compounds for motion sickness, vertigo and other disorders related to balance and the perception of gravity
WO2003027285A1 (en) * 2001-09-27 2003-04-03 Bionomics Limited Dna sequences for human angiogenesis genes

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
BRUMMELKAMP T.R. ET AL.: "A system for stable expression of short interfering RNAs in mammalian cells", SCIENCE, AMERICAN ASSOCIATION FOR THE ADVANCEMENT OF SCIENCE,, US, vol. 296, no. 5567, 2002, pages 550 - 553, XP002225638, ISSN: 0036-8075 *
HONG Y.-K. ET AL.: "Prox1 is a master control gene in the program specifying lymphatic endothelial cell fate.", DEVELOPMENTAL DYNAMICS, vol. 225, no. 3, November 2002 (2002-11-01), pages 351 - 357, XP009040935, ISSN: 1058-8388 *
LOHELA M. ET AL.: "Lymphangiogenic growth factors, receptors and therapies.", THROMBOSIS AND HAEMOSTASIS, vol. 90, no. 2, August 2003 (2003-08-01), pages 167 - 184, XP009040757, ISSN: 0340-6245 *
PETROVA T.V. ET AL.: "Effects of lymphatic transcription factor Prox-1 on cell cycle progression", JOURNAL OF SUBMICROSCOPIC CYTOLOGY AND PATHOLOGY, vol. 32, no. 3, July 2000 (2000-07-01), & XITH INTERNATIONAL VASCULAR BIOLOGY MEETING; GENEVA, SWITZERLAND; SEPTEMBER 05-09, 2000, pages 406, XP009040762, ISSN: 1122-9497 *
PETROVA T.V. ET AL.: "Lymphatic endothelial reprogramming of vascular endothelial cells by the Prox-1 homeobox transcription factor", EMBO (EUROPEAN MOLECULAR BIOLOGY ORGANIZATION) JOURNAL, vol. 21, no. 17, September 2002 (2002-09-01), pages 4593 - 4599, XP002309905, ISSN: 0261-4189 *
WIGLE J.T. ET AL.: "An essential role for Prox1 in the induction of the lymphatic endothelial cell phenotype", EMBO (EUROPEAN MOLECULAR BIOLOGY ORGANIZATION) JOURNAL, vol. 21, no. 7, 2 April 2002 (2002-04-02), pages 1505 - 1513, XP002309907, ISSN: 0261-4189 *
WIGLE J.T. ET AL.: "Prox1 function is required for the development of the murine lymphatic system", CELL, vol. 98, no. 6, 17 September 1999 (1999-09-17), pages 769 - 778, XP002309908, ISSN: 0092-8674 *
XIA H. ET AL.: "siRNA-mediated gene silencing in vitro and in vivo", NATURE BIOTECHNOLOGY, NATURE PUBLISHING, vol. 20, no. 10, October 2002 (2002-10-01), pages 1006 - 1010, XP002251054, ISSN: 1087-0156 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006092062A1 (en) * 2005-03-04 2006-09-08 The Hospital For Sick Children Methods for cancer prognosis
WO2008048680A2 (en) * 2006-10-19 2008-04-24 Maine Medical Center Research Institute Notch 2 signaling as a breast cancer suppressor pathway
WO2008048680A3 (en) * 2006-10-19 2009-04-16 Maine Medical Ct Res Inst Notch 2 signaling as a breast cancer suppressor pathway
EP3705137A4 (en) * 2017-11-03 2021-06-23 Korea Institute of Science and Technology Composition for regulating expression of prox1 or expression of telomerase reverse transcriptase comprising activity regulator or method for screening telomerase reverse transcriptase regulator

Also Published As

Publication number Publication date
US8029984B2 (en) 2011-10-04
EP1654383A1 (en) 2006-05-10
US20070026405A1 (en) 2007-02-01

Similar Documents

Publication Publication Date Title
JP6527565B2 (en) Method for treating hair loss disorders
Li Survivin study: what is the next wave?
US8029984B2 (en) Materials and methods for colorectal cancer screening, diagnosis and therapy
Joesting et al. Identification of SFRP1 as a candidate mediator of stromal-to-epithelial signaling in prostate cancer
US10655130B2 (en) Modulation of breast cancer growth by modulation of XBP1 activity
EP2859120B1 (en) Method for the diagnosis and prognosis of lung cancer metastasis
US9850543B2 (en) Biomarkers associated with BRM inhibition
JP2004159640A5 (en) Methods and compositions for predicting, diagnosing, prognosing, preventing and treating malignant tumors
JP2010525301A (en) Methods for diagnosis, classification and treatment of endometrial and precancer
WO2003058201A2 (en) Methods for identifying marker genes for cancer
JP2008514209A (en) Cancer marker
EP1900827A2 (en) Methods and compositions for the prediction, diagnosis, prognosis, prevention and treatment of malignant neoplasia
US20080113344A1 (en) Methods and Compositions for the Response Prediction of Malignant Neoplasia to Treatment
Fu et al. Foxp1 is indispensable for ductal morphogenesis and controls the exit of mammary stem cells from quiescence
EP2972372B1 (en) Markers associated with wnt inhibitors
Jiwani et al. Suppressor of fused controls cerebellum granule cell proliferation by suppressing Fgf8 and spatially regulating Gli proteins
US20140113006A1 (en) Methods of determining whether the wnt signaling pathway is upregulated in a tumor
US20050026194A1 (en) Gene amplification and overexpression in cancer
US11473149B2 (en) Breast cancer susceptibility gene GT198 and uses thereof
EP3268025B1 (en) Targeting gdf6 and bmp signaling for anti-melanoma therapy
US20040126788A1 (en) Fibulin-5 and uses thereof
US20150148411A1 (en) Compositions and methods for diagnosis and therapy of disorders related to alterations of myh9
Hasnis et al. Regulation of HDAC2-PDX1 by RNF125 defines pancreatic cancer development
Kadamb et al. Lineage commitment pathways epigenetically oppose oncogenic Gαq/11-YAP signaling in dormant disseminated uveal melanoma
AU2010355523A1 (en) MLK4 gene, a new diagnostic and prognostic marker in cancers

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2004763854

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2004763854

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2007026405

Country of ref document: US

Ref document number: 10567630

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 10567630

Country of ref document: US