WO2004037993A2 - Methodes permettant de produire une bibliotheque et methodes permettant de selectionner des polynucleotides d'interet - Google Patents

Methodes permettant de produire une bibliotheque et methodes permettant de selectionner des polynucleotides d'interet Download PDF

Info

Publication number
WO2004037993A2
WO2004037993A2 PCT/US2003/033557 US0333557W WO2004037993A2 WO 2004037993 A2 WO2004037993 A2 WO 2004037993A2 US 0333557 W US0333557 W US 0333557W WO 2004037993 A2 WO2004037993 A2 WO 2004037993A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
polynucleotide
seq
nucleotides
nucleic acid
Prior art date
Application number
PCT/US2003/033557
Other languages
English (en)
Other versions
WO2004037993A3 (fr
Inventor
Maurice Zauderer
Ernest S. Smith
Mark Paris
Original Assignee
University Of Rochester
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
Priority claimed from US10/277,161 external-priority patent/US20030194696A1/en
Application filed by University Of Rochester filed Critical University Of Rochester
Priority to AU2003294234A priority Critical patent/AU2003294234A1/en
Publication of WO2004037993A2 publication Critical patent/WO2004037993A2/fr
Publication of WO2004037993A3 publication Critical patent/WO2004037993A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/023Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a poxvirus

Definitions

  • the present invention relates to a high efficiency method of introducing DNA into poxvirus, a method of producing libraries in poxvirus, and methods of isolating polynucleotides of interest based on cell nonviability or screening or selection methods,
  • Toxic Sequences Several approaches have been employed for the identification and isolation of cell proliferation genes such as oncogenes and tumor suppressor genes.
  • Traditional approaches include detection of cytogenetic abnormalities in tumor cells, kindred analysis of familial forms of cancer, and loss of heterozygosity analysis in tumor cells.
  • Each of these classical genetic approaches is limited in the type of gene which can be isolated or in the extensive time and labor required.
  • a faster approach would be to identify disease genes using in vitro techniques.
  • a major technical hmitation to the cloning of many disease genes is their negative or toxic effect on cell proliferation when present in multiple copies, such as when carried on a vector.
  • revertants that have lost certain malignancy traits
  • a cytotoxic agent which kills dividing cells.
  • Revertants that have lost the ability to rapidly divide are thus selected.
  • revertant lines typically are difficult to identify and separate from the majority of rapidly growing transformed parental cells, hi addition, the method may preclude the isolation of certain classes of revertants.
  • the selection procedure may itself induce epigenetic or cytogenetic changes, thus further complicating the identification of genes responsible for the revertant phenotype.
  • Zarbl et al. developed an alternative assay for the selection of revertant tumor cells (Zarbl et al., Environmental Health Perspectives 93:83-89 (1991)). This selection protocol is based on the prolonged retention of a fluorescent molecule within the mitochondria of a number of transformed cells relative to non-transformed cells. However, the approach is limited to particular transformation mechanisms because the prolonged dye retention phenotype is neither essential nor sufficient for cell transformation.
  • candidates are identified based on a presumed (or identifiable) biochemical function or an abnormal pattern of expression. These candidates are then tested further for involvement in cancer. Such tests include mutation detection in primary cancers or cell lines, experiments using somatic cells (for example, to determine the effect of ectopic expression), or experiments in transgenic mice or knockout mice containing inactivated genes.
  • a more recent method for identifying cell proliferation genes involves the isolation of variants of transformed cells to identify a cell proliferation promoting activity. See U.S. Patent 5,998,136.
  • This selection system comprises the creation of growth arrested tumor cell lines or cells which undergo apoptosis by, for example, the expression of a gene encoding a growth suppressor or apoptosis-inducing gene product under the control of an inducible promoter, and selection of revertants that allow the cells to survive. Induction of the suppressor. or apoptosis-inducing product causes suppression of tumor cell growth and/or cell death. Growth-proficient revertants cells are identified by virtue of their continued proliferation.
  • Another approach is a method of selection subtraction by tagging a clone in an expression library, cloning the tagged clone into a vector, delivering the tagged clone to a target cell, and comparing tags before and after selection whereby toxic genes and the attached tags disappear. See WO 99/47643.
  • This method comprises use of a cloning vector encoding a recombinant immunoglobulin molecule (rAb) that is specific for a particular hapten and expressed on the cell surface.
  • rAb recombinant immunoglobulin molecule
  • Cells receiving the vector express the rAb early after transfection, and are separated from the non-recipient cells by the ability to bind the cognate hapten conjugated to a solid surface, such as beads.
  • This method does not distinguish recipients expressing a gene or cD ⁇ A of interest, e.g., a negative or toxic variant, from the remaining recipients.
  • Animal model systems such as the fruit fly and the worm are often used in gene identification because of ease of manipulation of the genome and ability to screen for mutants. While these systems have their limitations, large numbers of developmental mutations have been identified in those organisms either by monitoring the phenotypic effects of mutations or by screening for expression of reporter genes incorporated into developmentally regulated genes.
  • ES cells are totipotent cells isolated from the inner cell mass of the blastocyst. Methods are well known for obtaining ES cells, incorporating genetic material into ES cells, and promotion of differentiation of ES cells. ES cells may be caused to differentiate in vitro or the cells may be incorporated into a developing blastocyst in which the ES cells will contribute to all differentiated tissues of the resulting animal. Vectors for transforming ES cells and suitable genes for use as reporters and selectors are also well known.
  • entrapment strategies also have been employed to identify developmentally regulated genes.
  • One type of entrapment vector is called a "promoter trap,” which consists of a reporter gene sequence lacking a promoter. Its integration is detected when the reporter is integrated "in-frame” into an exon.
  • a "gene trap” vector targets the more prevalent introns of the eucaryotic genome.
  • the latter vector consists of a splice- acceptor site upstream from a reporter gene. Integration of the reporter into an intron results in a fusion transcript containing RNA from the endogenous gene and from the reporter gene sequence.
  • Gene trap vectors may be made more efficient by incorporation of an internal ribosomal entry site (IRES) such as that derived from the 5' non- translated region of encephalomyocarditis virus (EMCN). Placement of a IRES site between the splice acceptor and the reporter gene of a gene trap vector means the reporter gene product need not be translated as a fusion product with the endogenous gene product, thereby increasing the Mkelihood that integration of the vector will result in expression of the reporter gene product.
  • IRES internal ribosomal entry site
  • EMCN encephalomyocarditis virus
  • Gossler, A., et al. Science 244:A63-A65 (1989) describe the use of enhancer trap gene trap vectors for use in identifying developmentally regulated genes.
  • the gene trap vector consists of the mouse En-2 splice acceptor upstream from lacZ (reporter) and a selector gene (hBa-neo). This and other current methods requires elaborate screening procedures for linking a mutation to a particular spacial/ temporal scheme or event whereby the mutation is detected in the relevant tissue.
  • the method makes use of known genes whose expression is restricted to specific tissue, tissues or specialized cells ("restricted expression") to facilitate identification and manipulation of new genes and their associated transcription control elements which have similar patterns of expression.
  • the method comprises (i) transforming a eucaryotic cell with a D ⁇ A sequence encoding a first indicator component under the control of a promoter having restricted expression; (ii) fransforming the cell of (i) or a descendent of the cell of step (i), by operably integrating into the cell's genome D ⁇ A lacking a promoter but which comprises a sequence encoding a second indicator component; (iii) producing tissue or specialized cells from the cell of (ii); and (iv) monitoring the tissue or specialized cells of (iii) for a detectable indicator resulting from both the first and second indicator components.
  • Expression Libraries A basic tool in the field of recombinant genetics is the conversion of poly(A) + mR A to double-stranded (ds) cD ⁇ A, which then can be inserted into a cloning vector and expressed in an appropriate host cell.
  • ds double-stranded
  • a method common to many cDNA cloning strategies involves the construction of a "cDNA library" which is a collection of cDNA clones derived from the poly(A) + mRNA derived from a cell of the organism of interest.
  • a mammalian cell may contain up to 30,000 different mRNA sequences, and the number of clones required to obtain low-abundance mRNAs, for example, may be much greater.
  • Methods of constructing genomic eukaryotic DNA libraries in different expression vectors including bacteriophage lambda, cosmids, and viral vectors, are known. Some commonly used methods are described, for example, in Maniatis et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, publisher, Cold Spring Harbor, N.Y. (1982).
  • genomic cDNA library Once a genomic cDNA library has been constructed and expressed in host cells, it is necessary to isolate from the thousands of host cells the particular cell or cells which contain the particular gene of interest.
  • Many different methods of isolating target genes from cDNA libraries have been utilized, with varying success. These include, for example, the use of nucleic acid probes, which are labeled mRNA fragments having nucleic acid sequences complementary to the DNA sequence of the target gene.
  • this method is applied to cDNA clones of abundant mRNAs in transformed bacterial hosts, colonies hybridizing strongly to the probe are likely to contain the target DNA sequences. The identity of the clone then may be proven, for example, by in situ hybridization/selection (Goldberg et al, Methods Enzymol.
  • mammalian expression libraries to isolate cDNAs encoding mammalian proteins such as those described above would offer several advantages.
  • the protein expressed in a mammalian host cell should be functional and should undergo any normal posttranslational modification.
  • a protein ordinarily transported through the intracellular membrane system to the cell surface should undergo the complete transport process.
  • a mammalian expression system also would allow the study of intracellular transport mechanisms and of the mechanism that insert and anchor cell surface proteins to membranes.
  • use of a mammalian system would make it possible to isolate polynucleotides based on functional expression of mammalian RNA or protein.
  • COS mammalian host cell
  • SN40 simian virus strain 40
  • Any foreign D ⁇ A cloned on a vector containing the SN40 origin of replication will replicate because SN40 T antigen is present in COS cells.
  • the foreign D ⁇ A will replicate transiently, independently of the cellular D ⁇ A.
  • Lymphokine bioassays are very sensitive ((Wong, G. G., et al, Science 225:810-815 (1985); Lee, F. et al, Proc. Natl. Acad. Sci. USA 53:2061-2065 (1986); Yokota, T. et al, Proc. Natl. Acad. Sci. USA 53:5894- 5898 (1986); Yang, Y. et al, Cell 47:3-10 (1986)) and the mRNAs are typically both abundant and short (Wong, G. G.
  • Oshima et al. used plaque hybridization to screen a phage lambda gtl 1 cDNA library for the gene encoding human placental beta-glucuronidase. Oshima et al, Proceedings of the National Academy of Sciences, U.S. A. 84:685-689 (1987). The identity of isolated cDNA clones was verified by immunoprecipitation of the protein expressed by COS-7 cells transfected with cloned inserts using the SV40 late promoter.
  • This method for cloning cDNA encoding a cell surface antigen comprises preparing a cDNA library; introducing this cDNA library into eukaryotic mammalian cells; culturing the cells under conditions allowing expression of the cell surface antigen; exposing the cells to a first antibody or antibodies directed against the cell surface antigen, thereby allowing the formation of a cell surface antigen-first antibody complex; subsequently exposing the cells to a substrate coated with a second antibody directed against the first antibody, thereby causing cells expressing the cell surface antigen to adhere to the substrate via the formation of a cell surface antigen-first antibody-second antibody complex; and separating adherent from non- adherent cells.
  • this method is limited to the isolation and cloning of proteins which are expressed and transported to the cell surface, whose expression does not adversely affect cell viability, and for which specific antibody has been isolated.
  • Poxvirus Vectors are used extensively as expression vehicles for protein and antigen expression in eukaryotic cells. The ease of cloning and propagating vaccinia in a variety of host cells has led to the widespread use of poxvirus vectors for expression of foreign protein and as vaccine delivery vehicles (Moss, B., Science 252:1662-1667 (1991).
  • a foreign protein coding sequence is introduced into the poxvirus genome by homologous recombination.
  • a previously isolated foreign DNA is cloned in a transfer plasmid behind a vaccinia promoter flanked by sequences homologous to a region in vaccinia which is non-essential for viral replication.
  • the transfer plasmid is introduced into vaccinia virus-infected cells to allow the transfer plasmid and vaccinia virus genome to recombine in vivo via homologous recombination.
  • the foreign DNA is transferred to the viral genome.
  • homologous recombination is efficient for transferring previously isolated foreign DNA of relatively small size into vaccinia virus, the method is much less efficient for transferring large inserts, for constructing libraries, and for transferring foreign DNA which is deleterious to bacteria.
  • the vaccinia WR genome was modified by removing the Notl site in the Hindlll F fragment and reintroducing a Notl site proximal to the thymidine kinase gene such that insertion of a sequence at this locus disrupts the thymidine kinase gene, allowing isolation of chimeric genomes via use of drug selection (Merchlinsky, M. et al, Virology 190:522-526 (1992)).
  • the direct ligation vector vNotl/tk allows one to efficiently clone and propagate previously isolated DNA inserts at least 26 Mlobase pairs in length (Merchlinsky, M. et al, Virology 190:522-526 (1992)). Although large DNA fragments are efficiently cloned into the genome, proteins encoded by the DNA insert will only be expressed at the low level corresponding to the thymidine kinase gene, a relatively weakly expressed early class gene in vaccinia. In addition, the DNA will be inserted in both orientations at the Notl site.
  • a method of high efficiency cloning using a linear DNA virus vector such as vaccinia virus vector, comprising tri-molecular recombination.
  • a method of producing a library using a linear DNA virus vector such as vaccinia virus vector there is provided a method of producing a library using a linear DNA virus vector such as vaccinia virus vector.
  • a method of cloning a polynucleotide which negatively affects cell viability there is provided.
  • kits for producing a library using tri-molecular recombination there is provided a kit for producing an antisense expression library comprising a linear DNA viral genome such as vaccinia virus or two fragments thereof, and two vectors for producing a transfer plasmid containing a polynucleotide insert in each of two orientations.
  • the invention provides a kit for producing a protein expression library comprising a linear DNA genome such as vaccinia virus or two fragments thereof, and three vectors for producing a transfer plasmid containing a polynucleotide insert in each of three translation reading frames.
  • FIG. 1 Nucleotide Sequence of p7.5/tk (SEQ ID NO:l) and pEL/tk
  • SEQ ID NO:3 The nucleotide sequence of the promoter and beginning of the thymidine kinase gene for v7.5/tk and vEL/tk.
  • the partial thymidine kinase amino acid sequence is also shown (SEQ ID NO:2).
  • FIG. 2 Southern Blot Analysis of Viral Genomes ⁇ 7.5/tk and pEL/tk.
  • the viruses v7.5/tk and vEL/tk were used to infect a well of a 6 well dish of BSC-1 cells at high multiplicity of infection (moi) and after 48 hours the cells were harvested and the DNA was isolated using DNAzol (Gibco).
  • the final DNA product was resuspended in 50 microliters of TE 8.0 and 2.5 microliters were digested with Hindlll, HindHI and Apal, or Hindlll and Notl, electrophoresed through a 1.0% agarose gel, and transferred to Nytran (Schleicher and Schuell) using a Turboblotter (Schleicher and Schuell).
  • the samples were probed with p7.5/tk (A) or pEL/tk (B) labeled with 32 P using Random Primer DNA Labeling Kit (Bio-Rad) in QuickHyb (Stratagene).
  • the lower portion of the figure denotes a map of the Hindlll J fragment with the positions of the Hindi ⁇ , Notl, and Apal sites illustrated.
  • the leftmost 0.5 kilobase fragment has electrophoresed off the bottom of the gel.
  • FIG. 3 Restriction Enzyme Analysis of Virus Genomes Using CHEF
  • BSC-1 cells were infected at high multiplicity of infection (moi) by vaccinia WR, vEL/tk, v7.5/tk, or vNotl tk. After 24 hours the cells were harvested and formed into agarose plugs. The plugs were equilibrated in the appropriate restriction enzyme buffer and ImM PMSF for 16 hours at room temperature, incubated with restriction enzyme buffer, lOOng/ml Bovine Serum Albumin and 50 units Notl or Apal for two hours at 37 °C (Notl) or room temperature (Apal) and electrophoresed in a 1.0% agarose gel on a Bio- Rad CHEFII apparatus for 15 hours at 6 V/cm with a switching time of 15 seconds.
  • moi multiplicity of infection
  • the leftmost sample contains lambda DNA
  • the second sample contains undigested vaccinia DNA
  • the remainder of the samples contains the DNA samples described above each well digested with Apal or Notl where vEL refers to vEL/tk and v7.5 refers to v7.5/tk.
  • vEL refers to vEL/tk
  • v7.5 refers to v7.5/tk.
  • the lower portion of the figure is a schematic map showing the location of the Notl and Apal sites in each virus.
  • FIG. 4 Analysis ofv7.5/tk and vEL/tk by PCR.
  • One well of a 6 well dish of BSC-1 cells was infected with v7.5/tk, vEL/tk, vNotl/tk, vpNotl, vNotl lacZ/tk, or wild type vaccinia WR at high multiplicity of infection (moi) and after 48 hours the cells were harvested, and the DNA was isolated using DNAzol (Gibco). The final DNA product was resuspended in 50 microliters of TE (lOmM TrisHCl, pH8.0. ImM EDTA) and used in a PCR with primers MM407 and MM408.
  • TE lOmM TrisHCl, pH8.0. ImM EDTA
  • the primers are separated by 518 nucleotides in vaccinia WR and yield a fragment containing the N terminus of the thymidine kinase gene.
  • the products were electrophoresed through a 2% agarose gel.
  • the leftmost sample contains phiX 174 HaeHI digestion products; all others ""-' contain the PCR product using primers MM407 and MM408 with the DNA sample indicated above the well.
  • FIG. 5 Promoter strength of recombinant viruses.
  • the units of ⁇ -ghi activity were determined as described by Miller (Mamon, H., et al., Cold Spring Harb. Symp. Quant. Biol. 56:251-263 (1991)) as adapted for 96-well plates.
  • the A 4 05 values were determined on a microplate reader (Dynatech MR3000) and the ⁇ -glu activity was determined by comparison to ⁇ -glu (Clontech) standards analyzed in the same assay.
  • FIG. 6 Plaque assay on vEL/tk.
  • Ten-fold dilutions of vEL/tk were incubated with Hutk " cells (top to bottom) for one hour at 37 °C in 1ml of E- MEM (Gibco) with 10% Fetal Bovine Serum for one hour, the media was replaced with 3ml of E-MEM with 5% methyl cellulose (Sigma M-0387), 5% Fetal Bovine Serum and HAT supplement (Gibco), 25 or 125mM bromodeoxyuridine, or no drug, incubated for 48 hours at 37 °C, and stained with 0.5% Crystal Violet (Sigma C 0775), 20% ethanol, 7.5% formaldehyde.
  • FIG. 7 Schematic of the Tri-Molecular Recombination Method.
  • FIG. 8 Modifications in the nucleotide sequence of the p7.5/tk vaccinia transfer plasmid.
  • Four new vectors, p7.5/ATG0/tk (SEQ ID NO:4), p7.5/ ATGl/tk (SEQ ID NO:5), p7.5/ATG2/tk (SEQ ID NO:6), and ⁇ 7.5/ATG3/tk (SEQ ID NO:7) have been derived as described in the text from the p7.5/tk vaccinia transfer plasmid.
  • Each vector includes unique BamHI, Smal, Pstl, and Sail sites for cloning DNA inserts that employ either their own endogenous translation initiation site (in vector p7.5/ ATGO/tk) or make use of a vector translation initiation site in any one of the three possible reading frames (p7.5/ATGl/tk, p7.5/ATG3/tk, and p7.5/ATG4/tk).
  • FIG. 9 Schematic of a direct selection method using CTL.
  • FIG. 10 Schematic of the strategy to identify shared tumor antigen.
  • FIG. 11 CML selected recombinant vaccinia cDNA clones stimulate tumor specific CTL.
  • A CML Selected vaccinia clones were assayed for the ability, following infection of B/C.N, to stimulate tumor specific CTL to secrete interferon gamma (IFN ⁇ ). The amount of cytokine was measured by ELISA, and is represented as OD490 (Jothy, S., et al., Am. J. Pathol. 143:250- 257 (1993)). An OD490 of 1.4 is approximately equal to 4 ng/ml of IFN ⁇ , and an OD490 of 0.65 is approximately equal to 1 ngml of IFN ⁇ .
  • FIG. 12 The tumor antigen is encoded by a ribosomal protein L3 gene. Sequence of H2.16 and rpL3 from amino acid position 45 to 56.
  • A The amino acid (in single letter code) (SEQ ID NO:8) and nucleotide sequence (SEQ ID NO:9) of cDNA clone ⁇ pL3 (GenBank Accession no. Y00225).
  • B A single nucleotide substitution at C170T of the H2.16 tumor cDNA (SEQ ID NO: 10) is the only sequence change relative to the published L3 ribosomal allele. This substitution results in a T54I amino acid substitution in the protein (SEQ ID NO: 11).
  • A CML assay to identify the peptide recognized by tumor specific CTL.
  • Target cells were labeled with 51 Cr (Schrewe, H., et al., Mol. Cell Biol. 10:2738-2748 (1990)). During the 51 Cr incubation samples of B/C.N cells were incubated with l ⁇ M peptide L3 8 -s 6 (I54), 100 ⁇ M L3 8 . 5 6 (T54) or lOO ⁇ M peptide L3 5- 54 (I54).
  • Target cells were incubated with the indicated ratios of tumor specific Cytotoxic T Lymphocytes for 4 hours at 37 °C and percentage specific lysis was determined.
  • FIG. 14 Analysis of L3 expressed by each cell line.
  • A Sau3AI map of published rpL3 and H2.16. Shown above is the Sau3AI restriction map for the published ribosomal protein L3 gene (Top), and for H2.16 (Bottom). Digestion of cDNA for the published L3 sequence generates fragments of 200, 355, 348, 289, and 84bp. The pattern for H2.16 is identical except for an extra Sau3AI site at position 168 caused by the C170T. This results in a 168bp digestion product in place of the 200bp fragment.
  • B The BCA tumors express both L3 alleles.
  • RT-PCR products generated from each cell line or from vH2.16 were generated using L3 specific primers and then digested with Sau3AI, and resolved on a 3% agarose gel for 2 hours at 80 volts.
  • C The Immunogenic L3 allele is expressed at greatly reduced levels in B/C.N, BCB13, and Thymus.
  • L3 specific RT-PCR products from each indicated sample were generated using a 32 P end labeled 5 prime PCR primer. No PCR product was observed when RNA for each sample was used as template for PCR without cDNA synthesis, indicating that no sample was contaminated with genomic DNA.
  • PCR products were gel purified to ensure purity, digested with Sau3AI, and resolved on a 3% agarose gel for 15 hours at 60 volts. No PCR product was observed in a control PCR sample that had no template added to it. This result has been reproduced a total of 3 times.
  • Immunization with H2.16 induces tumor specific CTL.
  • Balb/c mice (2/group) were immunized by subcutaneous injection with 5X10 6 pfu of vH2.16, or control vector v7.5/tk. Seven days later splenocytes were harvested and restimulated with peptide L3 8 - 5 6(I54) (Pardoll, D. M. and Topalian, S. L., Curr. O ⁇ in.Immunol. 10:588-594 (1998)). Five days following the second restimulation the lymphocytes were tested in a chromium release assay as described in Figure 11. The L3 48 -5 6 (I54) peptide was used at a 1 micromolar concentration, and the L3 48 .
  • mice Female Balb/cByJ mice were immunized as indicated (Southwood, S., et al., J. Immunol. 160:3363-3373 (1998)). The mice were challenged by SC injection with 200,000 viable BCA 34 tumor cells into the abdominal wall. Data are from day 35 post challenge. These data are representative of 4 independent experiments.
  • FIG. 16 Influenza-specific cytolytic activity of CD4+ CD45RA+ human T cells stimulated in the presence of IL12 and IL18.
  • Na ⁇ ve human CD4+ CD45RA+ T cells were isolated from PBL of an HLA-A2+ normal donor and stimulated in vitro with autologous dendritic cells pulsed with heat- inactivated influenza virus.
  • the dendritic cells were derived from PBMC by culture with GM-CSF+IL-4 (1000 U/ml each) for 7 days.
  • DC were pulsed with heat-inactivated influenza virus (1000 HAU) and transferred to monocyte conditioned medium for 3 more days to induce maturation prior to T cell stimulation.
  • rhIL-2 (20 U/ml
  • rhIL-12 (20 U/ml, R&D Systems
  • rhIL-18 (10 ng/ml, R&D Systems
  • rhlFN- ⁇ (1 ng/ml
  • mouse anti-human IL-4 50 mg/ml, Pharmingen
  • Cells were restimulated after 7 days using identical conditions with fresh autologous DC pulsed with virus. Cytotoxic activity was assayed at day 14 in a 4 hr 51 Cr release assay using autologous monocytes +/- heat-inactivated virus or K562 control targets.
  • FIG. 17. CD4+ cytotoxic T cell response.
  • Na ⁇ ve CD4+ mouse T lymphocytes from heterozygous DO 11.10 transgenic females were cultured for 9 days in the presence of BALB/c bone marrow-derived mature dendritic cells pulsed with OVA 323-339 (10 mM).
  • Recombinant murine cytokines were purchased from R&D Systems and used at the same concentrations as indicated above for the human cytokines.
  • Rat ant-mouse IL-4 11B11, Pharmingen was used at 50 mg/ml.
  • B/c.N (H-2 d ) targets were incubated 72 hours with imlFN- ⁇ (1000 U/ml) to induce expression of class II MHC molecules prior to a 4 hr 51 Cr release assay.
  • the four panels demonstrate that OVA (323-339) specific cytotoxic cells are efficiently induced only in the presence of all 4 cytokines and anti-IL-4 antibody. As expected for this OVA (323-339) class II MHC restricted response, all the T cells recovered were CD4 positive.
  • FIG. 18 Gene isolation in solution. Schematic of a method for selection of longer length cDNA from single strand circles rescued from a phagemid library. DNA fragments identified through RDA or Modified Differential Display are employed to select more full length cDNA.
  • FIG. 19 An example of ADCC during a 4 hour incubation of normal
  • FIG. 20 Tolerance Induction.
  • DBA/2 (H-2 d ) mice were immunized with 10 7 C57B1/6 (H-2 b ) spleen cells intraperitoneally and, in addition, were injected with either saline or 0.5 mg monoclonal anti-CD40 ligand antibody (MR1B, anti-CD 154) administered both at the time of immunization and two days later.
  • MR1B, anti-CD 154 monoclonal anti-CD40 ligand antibody
  • spleen cells from these mice were removed and stimulated in vitro with either C57B1/6 or control allogeneic C3H (H-2 k ) spleen cells that had been irradiated (20 Gy).
  • C57B1/6 and C3H specific cytolytic responses were assayed at various effector :target ratios by 5I Cr release assay from specific labeled targets, in this case, either C3H or C57B1/6 dendritic cells pulsed with syngeneic spleen cell lysates.
  • FIG. 21 Attenuation of poxvirus-mediated cytopathic effects.
  • FIG. 22 Differentiation of mesenchymal stem cells following infection with vaccinia virus recombmants expressing BMP-2 or Sox-9 chondrocyte differentiation factors.
  • Mouse mesenchymal stem cells were harvested from embryonic day 11.5 mice. Control cells were plated at low density to avoid aggregation-mediated spontaneous differentiation. Bone Morphogenic Protein 2 (BMP-2) was used at a concentration of (50ng/ml).
  • BMP-2 Bone Morphogenic Protein 2
  • the graphs show a scaled composite of two independent realtime-PCR experiments performed on reverse transcribed total RNA.
  • the data shows the induction of cBfal, aggrecan, and collagen type ⁇ , key markers of chondrogenesis either by treatment with low levels of BMP-2 or through infection with vaccinia virus recombinants containing BMP-2 (wB) or SOX-9 (wS).
  • FIG. 23 Schematic of "points of convergence.”
  • B, C, and G represent proteins encoded by target polynucleotides (upstream genes), which are isolated in the methods of the invention by detecting the expression of reporter gene products such as endogenous proteins ("baits").
  • FIG 24 Human C35 genomic locus. Using the sequence of the human
  • C35 transcript the human C35 genomic locus was identified by BLAST analysis against the public human genome sequence database. This entire C35 gene (composed of four exons and three introns) is contained within a 1401 nucleotide region between 37,795,754 and 37,797,155 on chromosome 17.
  • the ERBB2/HER2 gene terminates 505 nucleotides from the 3' end of C35 while the GRB7 gene begins 7402 nucleotides from the 5' start of C35 transcription.
  • FIG 25 Distribution of Predicted Transcription Factor Binding Sites
  • the X axis represents nucleotide positions 706 to 1107 of SEQ ID NO:77 (number 1 corresponds to nucleotide 706 of SEQ TD NO:77, and number 402 corresponds to nucleotide 1107 of SEQ ID NO:77), while the Y axis shows the number of predicted binding sites at a given nucleotide position.
  • FIG 26 Testing of C35 Promoter By Luciferase Assay. Luciferase constructs were transfected into H16N2 and 21MT2 cell lines. Luciferase assay was performed 36 hours after transfection.
  • the invention provides a method of selecting a target polynucleotide, comprising: (a) introducing into a population of host cells a library of insert polynucleotides; wherein at least one of the insert polynucleotides comprises the target polynucleotide; and wherein expression of the target polynucleotide directly or indirectly promotes host cell death; (b) culturing said host cells; and (c) collecting insert polynucleotides from those host cells which undergo cell death.
  • the method further comprises: (d) introducing the collected polynucleotides into a population of host cells, wherein expression of the target polynucleotide directly or indirectly promotes host cell death; (e) culturing said host cells; and (f) collecting insert polynucleotides from those host cells which undergo cell death.
  • the method further comprises repeating steps
  • the method further comprises purifying the collected polynucleotides.
  • the host cells are adherent to a solid support.
  • expression of the target polynucleotide indirectly promotes cell death upon exposure of the host cells to an agent.
  • the agent comprises a member selected from the group consisting of: a physical agent, a chemical agent, and a biological agent.
  • the physical agent is selected from the group consisting of: radiation, UV radiation, gamma radiation, infrared radiation, visible light, increased temperature, and decreased temperature.
  • the chemical agent is selected from the group consisting of: a chemotherapeutic agent, a cytotoxic agent, and a DNA damaging agent.
  • the biological agent is selected from the group consisting of an antisense construct, an infectious agent, a therapeutic agent, an antibody, a cytotoxic T-lymphocyte (CTL), a ligand, a hapten, an epitope, and a receptor.
  • CTL cytotoxic T-lymphocyte
  • the biological agent is conjugated to a toxin.
  • the biological agent effects cell death by a process selected from the group consisting of: CTL-induced cytotoxicity, antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity.
  • the biological agent comprises a cytotoxic T lymphocyte (CTL), wherein said CTL expresses surface CD4, wherein said target polynucleotide encodes a polypeptide, and wherein said polypeptide is processed and presented in association with a class II major histocompatibility molecule (MHC).
  • CTL cytotoxic T lymphocyte
  • MHC major histocompatibility molecule
  • expression of said target polynucleotide effects a cellular process selected from the group consisting of cellular differentiation, growth regulation, cellular proliferation, apoptosis, and hormonal response.
  • cell death is the result of apoptosis.
  • apoptosis is induced through expression of a apoptosis-related gene product which directly promotes apoptosis.
  • apoptosis is induced through expression of an apoptosis-related gene product which indirectly promotes apoptosis.
  • the apoptosis-related gene product comprises a death domain containing receptor expressed on the surface of said host cells, and wherein said host cells are contacted with a ligand for said death domain containing receptor.
  • those cells which have undergone apoptosis are released from said substrate.
  • the released host cells, or contents thereof are collected by removing the liquid medium in which said host cells are cultured.
  • those host cells which have undergone apoptosis are fully or partially lysed, thereby releasing their cytoplasmic contents into the liquid medium in which said host cells are cultured.
  • the released host cell contents are collected by removing the liquid medium in which said host cells are cultured.
  • the cell death is the result of a cytotoxic T- lymphocyte induced lytic event.
  • the target polynucleotide encodes a target epitope for a cytotoxic T lymphocyte (CTL).
  • CTL a CD4+ CTL.
  • the target epitope is expressed on the surface of said host cells in the context of a native MHC molecule expressed on said host cell, and wherein said host cells are contacted with CTLs which are restricted for said MHC molecule and specific for said target epitope.
  • the MHC molecule is selected from the group consisting of a class I MHC molecule and a class II MHC molecule.
  • the MHC molecule is a class II MHC molecule.
  • the target polynucleotide is fused to a polynucleotide encoding Ii-80 fragment of the class II MHC molecule invariant chain.
  • those cells which have undergone a CTL- mediated lytic event are released from said substrate.
  • the released host cells, or contents thereof are collected by removing the liquid medium in which said host cells are cultured.
  • those host cells which have undergone a CTL- mediated lytic event are fully or partially lysed, thereby releasing their cytoplasmic contents into the liquid medium in which said host cells are cultured.
  • the released host cell contents are collected by removing the liquid medium in which said host cells are cultured.
  • the cell death is the result of expression of a suicide gene product.
  • the suicide gene product is selected from the group consisting of a diphtheria toxin A chain polypeptide, a Pseudomonas exotoxin A chain polypeptide, a ricin A chain polypeptide, an abrin A chain polypeptide, a modeccin A chain polypeptide, and an alpha-sarcin polypeptide.
  • the host cells are progenitor cells comprising a suicide gene operably associated with a tissue-restricted promoter; wherein expression of said target polynucleotide directly or indirectly induces transcription of said tissue-restricted promoter, resulting in expression of said suicide gene; and wherein expression of said suicide gene promotes death of those progenitor cells harboring said target polynucleotide.
  • the host cell is a RAW cell, and wherein said suicide gene is operably associated with the TRAP promoter.
  • the target polynucleotide directly or indirectly regulates osteoclast differentiation.
  • the suicide gene encodes the Diphtheria toxin
  • the tissue-restricted promoter is identified by gene expression profiling of said host cells under different conditions in microarrays of ordered cDNA libraries.
  • those host cells expressing said suicide gene product are released from said substrate.
  • the released host cells, or contents thereof are collected by removing the liquid medium in which said host cells are cultured.
  • those host cells expressing said suicide gene product are fully or partially lysed, thereby releasing their cytoplasmic contents into the liquid medium in which said host cells are cultured.
  • the released host cell contents are collected by removing the liquid medium in which said host cells are cultured.
  • cell death occurs within a period selected from the group consisting of: 48 hours after expression of said insert polynucleotide, 24 hours after expression of said insert polynucleotide, and 12 hours after expression of said insert polynucleotide.
  • said library of polynucleotides is constructed in a eukaryotic virus vector.
  • the eukaryotic virus vector is an animal virus vector.
  • the eukaryotic virus vector is a plant virus vector.
  • the eukaryotic virus vector is capable of producing infectious viral particles in cells selected from the group consisting of insect cells, plant cells, and mammalian cells. [0120] In further embodiments, the eukaryotic virus vector is attenuated. [0121] In further embodiments, the eukaryotic virus vector is capable of producing infectious viral particles in mammalian cells.
  • the attenuation is by genetic mutation.
  • the attenuation is by reversible inhibition of virus replication.
  • the naturally-occurring genome of said eukaryotic virus vector is DNA.
  • the naturally-occurring genome of said eukaryotic virus vector is linear, double-stranded DNA.
  • the eukaryotic virus vector is selected from the group consisting of an adenoviras vector, a herpesvirus vector and a poxvirus vector.
  • the eukaryotic virus vector is a poxvirus vector.
  • the poxvirus vector is selected from the group consisting of an orthopoxvirus vector, an avipoxvirus vector, a capripoxvirus vector, a leporipoxvirus vector, and a suipoxvirus vector.
  • the poxvirus vector is an orthopoxvirus vector selected from the group consisting of a vaccinia virus vector and a raccoon poxvirus vector.
  • the orthopoxvirus vector is a vaccinia virus vector.
  • the invention provides a method of selecting a target polynucleotide, comprising: (a) introducing into a population of host cells a library of insert polynucleotides; wherein said library is constructed in a linear DNA virus vector; wherein at least one of said insert polynucleotides comprises said target polynucleotide; and wherein expression of said target polynucleotide directly or indirectly prevents death of a host cell comprising said target polynucleotide; (b) culturing said host cells; and (c) collecting insert polynucleotides from those host cells which do not undergo cell death.
  • the method further comprises: (d) introducing said collected polynucleotides into a population of host cells, and wherein expression of said target polynucleotide directly or indirectly prevents death of a host cell comprising said target polynucleotide; (e) culturing said host cells; and (f) collecting insert polynucleotides from those host cells which do not undergo cell death.
  • the method further comprises repeating steps
  • the method further comprises purifying said collected polynucleotides. .
  • the invention provides a method of selecting a target polynucleotide, comprising; (a) introducing into a population of host cells a library of insert polynucleotides; wherein said library is constructed in a linear DNA virus vector; wherein at least one of said insert polynucleotides comprises said target polynucleotide; wherein exposure of said host cells to an agent promotes cell death; and wherein expression of said target polynucleotide directly or indirectly prevents death of a host cell comprising said target polynucleotide; (b) culturing said host cells; (c) exposing said host cells to said agent; and (d) collecting insert polynucleotides from those host cells which do not undergo cell death.
  • the method further comprises: (e) introducing said collected polynucleotides into a population of host cells, wherein exposure of said host cells to an agent promotes cell death; and wherein expression of said target polynucleotide directly or indirectly prevents death of a host cell comprising said target polynucleotide; (f) culturing said host cells; (g) exposing said host cells to said agent; and (h) collecting insert polynucleotides from those host cells which do not undergo cell death.
  • the method further comprised repeating steps
  • the method further comprises purifying said collected polynucleotides.
  • said cell death is the result of a cellular effect selected from the group consisting of cell lysis, expression of a suicide gene product, a cytotoxic T-lymphocyte induced lytic event, apoptosis, loss of viability, loss of membrane integrity, loss of structural stability, cell disruption, disruption of cytoskeletal elements, inability to maintain membrane potential, arrest of cell cycle, inability to generate energy, growth arrest, cytotoxic effects, cytostatic effects, genotoxic effects, and growth suppressive effects.
  • cell death occurs within a period selected from the group consisting of: 48 hours after expression of said insert polynucleotide, 24 hours after expression of said insert polynucleotide, and 12 hours after expression of said insert polynucleotide.
  • the host cells are adherent to a solid support.
  • the agent is a member selected from the group consisting of: a physical agent, a chemical agent, and a biological agent.
  • the physical agent is selected from the group consisting of: radiation, UV radiation, gamma radiation, infrared radiation, visible light, increased temperature, and decreased temperature.
  • the chemical agent is selected from the group consisting of: a chemotherapeutic agent, a cytotoxic agent, and a DNA damaging agent.
  • the biological agent is selected from the group consisting of an antisense construct, an infectious agent, a therapeutic agent, an antibody, a cytotoxic T-lymphocyte (CTL), a ligand, a hapten, an epitope, and a receptor.
  • CTL cytotoxic T-lymphocyte
  • the biological agent is selected from the group consisting of: an infectious agent, a therapeutic agent, an antibody, a ligand, a hapten, an epitope, and a receptor; and wherein said biological agent is conjugated to a toxin.
  • the biological agent effects cell death by a process selected from the group consisting of: CTL-induced cytotoxicity, antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity.
  • the biological agent comprises a cytotoxic T lymphocyte (CTL), wherein said CTL expresses surface CD4, wherein said target polynucleotide encodes a polypeptide, and wherein said polypeptide is processed and presented in association with a class II major histocompatibility molecule (MHC).
  • CTL cytotoxic T lymphocyte
  • MHC major histocompatibility molecule
  • expression of said target polynucleotide effects a cellular process selected from the group consisting of cellular differentiation, growth regulation, cellular proliferation, apoptosis, and hormonal response.
  • the cell death is the result of apoptosis.
  • apoptosis is induced through expression of an apoptosis-related gene product which directly promotes apoptosis.
  • apoptosis is induced through expression of an apoptosis-related gene product which indirectly promotes apoptosis.
  • the apoptosis-related gene product comprises a death domain containing receptor expressed on the surface of said host cells, and wherein said host cells are contacted with a ligand for said death domain containing receptor.
  • those cells which have undergone apoptosis are released from said support.
  • the released host cells, or contents thereof are removed from said cells which do not undergo cell death.
  • those host cells which have undergone apoptosis are fully or partially lysed, thereby releasing their cytoplasmic contents into the liquid medium in which said host cells are cultured.
  • the released host cell contents are removed from said cells which do not undergo cell death.
  • cell death is the result of expression of a suicide gene product.
  • expression of said target polynucleotide directly or indirectly inhibits expression of a suicide gene encoding said suicide gene product.
  • the suicide gene product is selected from the group consisting of a diphtheria toxin A chain polypeptide, a Pseudomonas exotoxin A chain polypeptide, a ricin A chain polypeptide, an abrin A chain polypeptide, a modeccin A chain polypeptide, and an alpha-sarcin polypeptide.
  • the host cells are progenitor cells comprising a suicide gene operably associated with a tissue-restricted promoter; wherein exposure to said agent induces transcription from said tissue-restricted promoter, resulting in expression of said suicide gene; and wherein expression of said target polynucleotide directly or indirectly inhibits expression of said suicide gene, thereby preventing death of said progenitor cells comprising said target polynucleotide.
  • the host cell is a RAW cell, wherein said agent is the RANK ligand (RANKL), and wherein said tissue-restricted promoter is the TRAP promoter.
  • said agent is the RANK ligand (RANKL)
  • said tissue-restricted promoter is the TRAP promoter.
  • the target polynucleotide directly or indirectly regulates osteoclast differentiation.
  • the suicide gene encodes the Diphtheria toxin
  • the tissue-restricted promoter is identified by gene expression profiling of said host cells under different conditions in microarrays of ordered cDNA libraries .
  • the expression profiling compares gene expression under different conditions in host cells infected with a eukaryotic
  • said eukaryotic virus expression vector is the vector used to construct said library of polynucleotides.
  • the host cells are non-dividing cells comprising a suicide gene operably associated with a proliferation-specific promoter; wherein exposure to said agent induces transcription from said proliferation-specific promoter, resulting in expression of said suicide gene; and wherein expression of said target polynucleotide directly or indirectly inhibit sexpression of said suicide gene, thereby preventing death of said non- dividing cells comprising said target polynucleotide.
  • the proliferation-specific promoter is identified by gene expression profiling of said host cells under different conditions in microarrays of ordered cDNA libraries.
  • the expression profiling compares gene expression under different conditions in host cells infected with a eukaryotic viras expression vector, wherein said eukaryotic virus expression vector is the vector used to construct said library of polynucleotides.
  • those host cells expressing said suicide gene product are released from said support.
  • the released host cells, or contents thereof are removed from said cells which do not undergo cell death.
  • those host cells expressing said suicide gene product are fully or partially lysed, thereby releasing their cytoplasmic contents into the liquid medium in which said host cells are cultured.
  • the released host cell contents are removed from said cells which do not undergo cell death.
  • cell death occurs within a period selected from the group consisting of: 48 hours after expression of said insert polynucleotide, 24 hours after expression of said insert polynucleotide, and 12 hours after expression of said insert polynucleotide.
  • the invention provides a method of selecting a target polynucleotide, comprising: (a) introducing into a population of host cells a library of insert polynucleotides; wherein said library is constructed in a linear DNA virus vector; wherein at least one of said insert polynucleotides comprises the target polynucleotide; and wherein expression of said target polynucleotide directly or indirectly alters a phenotype in a cell comprising said target polynucleotide; (b) culturing said host cells; and (c) collecting insert polynucleotides from those host cells which exhibit said altered phenotype.
  • the method further comprises: (d) introducing said collected polynucleotides into a population of host cells, and wherein expression of said target polynucleotide directly or indirectly alters a phenotype of a host cell comprising said target polynucleotide; (e) culturing said host cells; and (f) collecting insert polynucleotides from those host cells which exhibit said altered phenotype.
  • the method further comprises repeating steps
  • method further comprises purifying said collected polynucleotides.
  • the altered phenotype is the expression of a reporter gene product.
  • the reporter gene product is selected from the group consisting of an epitope (e.g., an endogenous protein or epitope- containing fragment thereof), chloramphenicol acetyl transferase (CAT), green fluorescent protein (GFP), blue fluorescent protein (BFP), yellow fluorescent protein (YFP), red fluorescent protein (RFP), luciferase and ⁇ -galactosidase.
  • expression of the reporter gene product is detected by FACS.
  • the cells are selected by cell sorting.
  • the polynucleotide encoding the reporter gene product is under the control of a promoter and the target polynucleotide encodes an upstream gene (e.g. upstream regulator) of the promoter.
  • the target polynucleotide may directly or indirectly activate (transactivate, induce, stimulate, upregulate, enhance, increase, etc.) or suppress (decrease) expression of the promoter.
  • a reporter gene product that is an epitope (e.g. endogenous protein or fragment)
  • the polynucleotide encoding the epitope e.g.
  • endogenous protein or fragment may be under the control of its endogenous promoter, and the target polynucleotide directly or indirectly activates (transactivates, induces, stimulates, upregulates, enhances, increases, etc.) or suppresses (decreases) expression of the promoter and/or expression of the epitope (e.g. endogenous protein or fragment).
  • target polynucleotide directly or indirectly activates (transactivates, induces, stimulates, upregulates, enhances, increases, etc.) or suppresses (decreases) expression of the promoter and/or expression of the epitope (e.g. endogenous protein or fragment).
  • the polynucleotide encoding the epitope may be endogenous or may be exogenous (i.e., recombinant; the host cell is engineered with the polynucleotide encoding the epitope or both).
  • Epitopes e.g. endogenous proteins or fragments
  • the endogenous protein is full length.
  • Epitopes include those in Examples 28-29, or elesewhere herein.
  • the reporter gene product such as an epitope
  • the antibodies may be introduced into the cell by pinocytic vesicle delivery, HIV-TAT mediated delivery, peptide-based delivery or lipid-based delivery.
  • the antibody may be labeled or may be unlabeled.
  • the antibody may be detected by a secondary antibody.
  • the antibody or secondary antibody is labeled with a fluorescent label.
  • the antibody is detected by FACS, and the cells are selected by cell sorting.
  • the reporter gene product such as green fluorescent protein (GFP), blue fluorescent protein (BFP), yellow fluorescent protein (YFP), red fluorescent protein (RFP), or luciferase is encoded by a polynucleotide under the control of a promoter such as, for example, a promoter involved in cancer, inflammatory disease, atherosclerosis, differentiation or musculoskeletal disorders.
  • the promoter may be, for example, those in Examples 7, or 26-29, or elsewhere herein.
  • expression of said target polynucleotide indirectly promotes expression of said reporter gene product in said host cells upon exposure of said host cells to an agent.
  • the agent is a member selected from the group consisting of: a physical agent, a chemical agent, and a biological agent.
  • the physical agent is selected from the group consisting of: radiation, UV radiation, gamma radiation, infrared radiation, visible light, increased temperature, and decreased temperature.
  • the chemical agent is selected from the group consisting of: a chemotherapeutic agent, a cytotoxic agent, and a DNA damaging agent.
  • the biological agent is selected from the group consisting of an antisense construct, an infectious agent, a therapeutic agent, an antibody, a cytotoxic T-lymphocyte (CTL), a ligand, a hapten, an epitope, and a receptor.
  • CTL cytotoxic T-lymphocyte
  • the biological agent is selected from the group consisting of: an infectious agent, a therapeutic agent, an antibody, a ligand, a hapten, an epitope, and a receptor; and wherein said biological agent is conjugated to a toxin.
  • the biological agent is selected from the group consisting of: an infectious agent, a therapeutic agent, an antibody, a ligand, a hapten, an epitope, and a receptor; and wherein said biological agent is labeled.
  • the biological agent effects cell death by a process selected from the group consisting of: CTL-induced cytotoxicity, antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity.
  • the biological agent comprises a cytotoxic T lymphocyte (CTL), wherein said CTL expresses surface CD4, wherein said target polynucleotide encodes a polypeptide, and wherein said polypeptide is processed and presented in association with a class II major histocompatibility molecule (MHC).
  • CTL cytotoxic T lymphocyte
  • MHC major histocompatibility molecule
  • expression of said target polynucleotide effects a cellular process selected from the group consisting of cellular differentiation, growth regulation, cellular proliferation, apoptosis, and hormonal response.
  • the host cells are progenitor cells comprising a selectable gene product operably associated with a tissue-restricted promoter; wherein expression and secretion of a product encoded by said target polynucleotide directly or indirectly induces transcription of said tissue- restricted promoter, resulting in expression of said selectable gene product.
  • the host cell is a RAW cell, and wherein said selectable gene product is operably associated with the TRAP promoter.
  • the target polynucleotide directly or indirectly regulates osteoclast differentiation.
  • the tissue-restricted promoter is identified by gene expression profiling of said host cells under different conditions in microarrays of ordered cDNA libraries.
  • the host cells are non-dividing cells comprising a selectable gene product operably associated with a proliferation- specific promoter; wherein expression and secretion of a product encoded by said target polynucleotide directly or indirectly induces transcription of said proliferation-specific promoter, resulting in expression of said selectable gene product.
  • the proliferation-specific promoter is identified by gene expression profiling of said host cells under different conditions in microarrays of ordered cDNA libraries.
  • the invention provides a method of selecting a target polynucleotide, comprising: (a) dividing host cells comprising a library of insert polynucleotides into pools; wherein said library is constructed in a linear DNA virus vector; wherein at least one of said insert polynucleotides comprises the target polynucleotide; and wherein expression of said target polynucleotide and secretion of the product of said target polynucleotide directly or indirectly alters a phenotype of an indicator cell; (b) culturing said host cell pools in the presence of indicator cells; (c) collecting insert polynucleotides from those host cell pools in which said indicator cells exhibit an altered phenotype.
  • the method further comprises: (d) introducing said collected polynucleotides into host cells; (e) dividing the host cells of (d) into pools; wherein expression of said target polynucleotide directly or indirectly alters a phenotype of said indicator cells; (f) culturing said host cell pools in the presence of indicator cells; (g) collecting insert polynucleotides from those host cell pools in which said indicator cells exhibit an altered phenotype.
  • the method further comprises repeating steps
  • the method further comprises purifying said collected polynucleotides.
  • the altered phenotype is the expression of a reporter gene product.
  • the reporter gene product is selected from the group consisting of an epitope, chloramphenicol acetyl transferase (CAT), green fluorescent protein (GFP), blue fluorescent protein (BFP), yellow fluorescent protein (YFP), red fluorescent protein (RFP), luciferase and ⁇ - galactosidase.
  • CAT chloramphenicol acetyl transferase
  • GFP green fluorescent protein
  • BFP blue fluorescent protein
  • YFP yellow fluorescent protein
  • RFP red fluorescent protein
  • luciferase luciferase and ⁇ - galactosidase.
  • the indicator cells are selected from the group consisting of: tumor cells, metastatic tumor cells, primary cells, transformed primary cells, immortalized primary cells, dividing cells, non dividing cells, terminally differentiated cells, pluripotent stem cells, committed progenitor cells, uncommitted stem cells, progenitor cells, muscle cells, epithelial cells, nervous system cells, circulatory system cells, respiratory system cells, endocrine cells, endocrine-associated cells, skeletal system cells, connective tissue cells, musculoskeletal cells, chondrocytes, osteoblasts, osteoclasts, myocytes, fully differentiated blood cells, fully differentiated epidermal cells, neurons, glial cells, kidney cells, liver cells, muscle cell progemtors, epithelial cell progenitors, nervous system cell progenitors, circulatory system cell progenitors, respiratory system cell progenitors, endocrine cell progenitors, endocrine-associated cell progenitors, skeletal system cell progenitors,
  • the target polynucleotide alters a phenotype of said indicator cells upon exposure of said host cells to an agent.
  • the agent is selected from the group consisting of: a physical agent, a chemical agent, and a biological agent.
  • the physical agent is selected from the group consisting of: radiation, UV radiation, gamma radiation, infrared radiation, visible light, increased temperature, and decreased temperature.
  • the chemical agent is selected from the group consisting of: a chemotherapeutic agent, a cytotoxic agent, and a DNA damaging agent.
  • the biological agent is selected from the group consisting of an antisense construct, an infectious agent, a' therapeutic agent, an antibody, a cytotoxic T-lymphocyte (CTL), a ligand, a hapten, an epitope, and a receptor.
  • CTL cytotoxic T-lymphocyte
  • the biological agent is selected from the group consisting of: an infectious agent, a therapeutic agent, an antibody, a ligand, a hapten, an epitope, and a receptor; and wherein said biological agent is conjugated to a toxin.
  • the biological agent is selected from the group consisting of: an infectious agent, a therapeutic agent, an antibody, a ligand, a hapten, an epitope, and a receptor; and wherein said biological agent is labeled.
  • the biological agent effects cell death by a process selected from the group consisting of: CTL-induced cytotoxicity, antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity.
  • the biological agent comprises a cytotoxic T lymphocyte (CTL), wherein said CTL expresses surface CD4, wherein said target polynucleotide encodes a polypeptide, and wherein said polypeptide is processed and presented in association with a class II major histocompatibility molecule (MHC).
  • CTL cytotoxic T lymphocyte
  • MHC major histocompatibility molecule
  • expression of said target polynucleotide effects a cellular process selected from the group consisting of cellular differentiation, growth regulation, cellular proliferation, apoptosis, and hormonal response.
  • the indicator cells are progenitor cells comprising a selectable gene product operably associated with a tissue- restricted promoter; wherein expression of said target polynucleotide directly or indirectly induces transcription of said tissue-restricted promoter, resulting in expression of said selectable gene product.
  • the indicator cell is a RAW cell, and wherein the marker gene is operably associated with the TRAP promoter.
  • the target polynucleotide directly or indirectly regulates osteoclast differentiation in said indicator cells.
  • the indicator cells are non-dividing cells comprising a marker gene operably associated with a proliferation-specific promoter; wherein expression of said target polynucleotide in said host cells directly or indirectly induces transcription of said proliferation-specific promoter, resulting in expression of said marker gene.
  • the invention provides a method of selecting a target polynucleotide encoding a secreted product, comprising: (a) introducing into a population of host cells a library of insert polynucleotides; wherein at least one of said insert polynucleotides comprises said target polynucleotide; and wherein expression of said target polynucleotide directly or indirectly alters a phenotype in a population of indicator cells; (b) culturing said pools of host cells in the presence of said indicator cells; and (c) collecting insert polynucleotides from those host cell pools in the presence of which said selectable gene product is expressed.
  • the method further comprises: (d) dividing said collected insert polynucleotides into sub-pools; (e) introducing said polynucleotide sub-pools into sub-pools of host cells, wherein expression of said target polynucleotide and secretion of said product directly or indirectly promotes expression of a selectable gene product in a population of indicator cells; (f) culturing said host cell sub-pools in the presence of said indicator cells; and (g) collecting insert polynucleotides from those sub-pools of host cells in the presence of which said selectable gene product is expressed.
  • the invention provides a method of selecting a target polynucleotide encoding a secreted product, comprising:
  • the method further comprises: (e) dividing said collected insert polynucleotides into sub-pools; (f) introducing said polynucleotide sub-pools into sub-pools of host cells, wherein expression of said target polynucleotide and secretion of said product directly or indirectly inhibits expression of a selectable gene product in a population of indicator cells, said indicator cells expressing said selectable gene product upon exposure to an agent; (g) culturing said host cell sub-pools in the presence of said indicator cells; (h) exposing said indicator cells to said agent; and (i) collecting insert polynucleotides from those sub-pools of host cells in the presence of which said selectable gene product is not expressed.
  • the method further comprised repeating steps
  • the method further comprises purifying said collected polynucleotides. »
  • the library of insert polynucleotides is an antisense library.
  • the agent is a member selected from the group consisting of: a physical agent, a chemical agent, and a biological agent.
  • the physical agent is selected from the group consisting of: radiation, UV radiation, gamma radiation, infrared radiation, visible light, increased temperature, and decreased temperature.
  • the chemical agent is selected from the group consisting of: a chemotherapeutic agent, a cytotoxic agent, and a DNA damaging agent.
  • the biological agent is selected from the group consisting of an antisense construct, an infectious agent, a therapeutic agent, an antibody, a cytotoxic T-lymphocyte (CTL), a ligand, a hapten, an epitope, and a receptor.
  • CTL cytotoxic T-lymphocyte
  • the biological agent is selected from the group consisting of: an infectious agent, a therapeutic agent, an antibody, a ligand, a hapten, an epitope, and a receptor; and wherein said biological agent is conjugated to a toxin.
  • the biological agent effects cell death by a process selected from the group consisting of: CTL-induced cytotoxicity, antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity.
  • the biological agent comprises a cytotoxic T lymphocyte (CTL), wherein said CTL expresses surface CD4, wherein said target polynucleotide encodes a polypeptide, and wherein said polypeptide is processed and presented in association with a class II major histocompatibility molecule (MHC).
  • CTL cytotoxic T lymphocyte
  • MHC major histocompatibility molecule
  • expression of said target polynucleotide effects a cellular process selected from the group consisting of cellular differentiation, growth regulation, cellular proliferation, apoptosis, and hormonal response.
  • the indicator cells are progenitor cells comprising a selectable gene product operably associated with a tissue- restricted promoter; wherein expression and secretion of a product encoded by said target polynucleotide directly or indirectly inhibits transcription of said tissue-restricted promoter, thereby blocking expression of said selectable gene product.
  • the indicator cell is a RAW cell, wherein said agent is the RANK Ligand (RANKL), and wherein said tissue-restricted promoter is the TRAP promoter.
  • the target polynucleotides expressing a secreted product which directly or indirectly regulates osteoclast differentiation in indicator cells.
  • the tissue-restricted promoter is identified by gene expression profiling of said indicator cells under different conditions in microarrays of ordered cDNA libraries.
  • the indicator cells are non-dividing cells comprising a selectable gene product operably associated with a proliferation- specific promoter; and wherein expression and secretion of a product encoded by said target polynucleotide directly or indirectly inhibits transcription of said proliferation-specific promoter, thereby blocking expression of said selectable gene product.
  • the proliferation-specific promoter is identified by gene expression profiling of said indicator cells under different conditions in microarrays of ordered cDNA libraries.
  • the cell death may be the result of a cellular effect selected from the group consisting of cell lysis, expression of a suicide gene product, a cytotoxic T-lymphocyte induced lytic event, apoptosis, loss of viability, loss of membrane integrity, loss of structural stability, cell disruption, disruption of cytoskeletal elements, inability to maintain membrane potential, arrest of cell cycle, inability to generate energy, growth arrest, cytotoxic effects, cytostatic effects, genotoxic effects, and growth suppressive effects.
  • a cellular effect selected from the group consisting of cell lysis, expression of a suicide gene product, a cytotoxic T-lymphocyte induced lytic event, apoptosis, loss of viability, loss of membrane integrity, loss of structural stability, cell disruption, disruption of cytoskeletal elements, inability to maintain membrane potential, arrest of cell cycle, inability to generate energy, growth arrest, cytotoxic effects, cytostatic effects, genotoxic effects, and growth suppressive effects.
  • the population of host cells is selected from the group consisting of: tumor cells, metastatic tumor cells, primary cells, transformed primary cells, immortalized primary cells, dividing cells, non dividing cells, terminally differentiated cells, pluripotent stem cells, committed progenitor cells, uncommitted stem cells, progenitor cells, muscle cells, epithelial cells, nervous system cells, circulatory system cells, respiratory system cells, endocrine cells, endocrine-associated cells, skeletal system cells, connective tissue cells, musculoskeletal cells, chondrocytes, osteoblasts, osteoclasts, myocytes, fully differentiated blood cells, fully differentiated epidermal cells, neurons, glial cells, kidney cells, liver cells, muscle cell progemtors, epithelial cell progenitors, nervous system cell progenitors, circulatory system cell progenitors, respiratory system cell progenitors, endocrine cell progenitors, endocrine-associated cell progenitors
  • the solid support is selected from the group consisting of: tissue culture plastic, glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, magnetite, soluble material, partially soluble material, insoluble material, magnetic material, and nonmagnetic material.
  • the solid support has a structure selected from the group consisting of: spherical, beadlike, bead, cylindrical, test tube-like, tube-like, tube, rod-like, rod, flat, sheetlike, sheet, test strip, strip-like, strip, bead, microbead, well, plate, tissue culture plate, petri plate, microplate, microtiter plate, flask, stick, vial, and paddle.
  • the library of insert polynucleotides is selected from the group consisting of: a cDNA library, a genomic library, a combinatorial polynucleotide library, a library of natural polynucleotides, a library of artificial polynucleotides, a library of polynucleotides endogenous to the host cells, a library of polynucleotides exogenous to the host cells, an antisense library, and any combination thereof.
  • the library of insert polynucleotides is constructed in said eukaryotic virus vector by a method comprising: (a) providing host cells comprising a linear DNA . virus genome which has been cleaved to produce a first viral fragment and a second viral fragment, wherein said first fragment is nonhomologous with said second fragment; (b) providing a population of transfer plasmids comprising said insert polynucleotides in operable association with a vector transcriptional control region, a 5' flanking region, and a 3' flanking region; wherein said 5' flanking region is homologous to said first viral fragment and said 3' flanking region is homologous to said second viral fragment; and wherein said transfer plasmids are capable of homologous recombination with said first and second viral fragments such that a viable viras genome is formed; (c) introducing said transfer plasmids into said host cells under conditions wherein a transfer plasmids into said host cells under conditions wherein a transfer
  • the first and second viral fragments are produced by infecting said host cells with a virus comprising said linear DNA virus genome, and wherein said virus genome is cleaved in vivo.
  • the first and second viral fragments are produced by cleaving an isolated linear DNA viras genome in vitro, and wherein said first and second viral fragments are introduced into said host cells.
  • the virus genome comprises a first recognition site for a first restriction endonuclease; and wherein said first and second viral fragments are produced by digesting said viral genome with said first restriction endonuclease, and isolating said first and second viral fragments.
  • the viras genome further comprises a second recognition site for a second restriction endonuclease; and wherein said first and second viral fragments are produced by digesting said viral genome with said first restriction endonuclease and said second restriction endonuclease, and isolating said first and second viral fragments.
  • the first and second recognition sites are physically arranged in said genome such that the region extending between said first and second viral fragments is not essential for viras infectivity.
  • the modified viras genome is packaged in an infectious viral particle.
  • the modified viras genome is defective in an essential gene and said host cell comprises a complementing copy of said essential gene.
  • the complementing copy of said essential gene is operably associated with an inducible promoter.
  • the inducible promoter is selected from the group consisting of: a differentiation- induced promoter, a cell type-restricted promoter, a tissue-restricted promoter, a temporally-regulated promoter, a spatially-regulated promoter, a proliferation-induced promoter, a cell-cycle specific promoter.
  • the linear DNA virus genome is a herpes virus genome.
  • the linear DNA viras genome is an adenoviras genome.
  • the linear DNA virus genome is a poxvirus genome.
  • the poxvirus genome is a vaccinia viras genome.
  • the transfer plasmid and said first and second viral fragments are introduced into a host cell comprising a helper viras, wherein said host cell is non-permissive for the production of infectious virus particles of said helper viras.
  • the helper virus is an avipoxvirus.
  • the helper viras is a fowlpox virus.
  • the 5' and 3' flanking regions of said transfer plasmids are capable of homologous recombination with a vaccinia virus thymidine kinase gene.
  • the 5' and 3' flanking regions of said transfer plasmids are capable of homologous recombination with a vaccinia virus Hindlll J fragment.
  • the transfer plasmid comprises an insert polynucleotide operably associated with a promoter selected from the group consisting of a vaccinia viras p7.5 promoter, a synthetic early/late promoter, and a vaccinia virus MH5 early/late promoter.
  • a promoter selected from the group consisting of a vaccinia viras p7.5 promoter, a synthetic early/late promoter, and a vaccinia virus MH5 early/late promoter.
  • the transfer plasmid comprises the sequence shown in SEQ ID NO: .
  • the library of polynucleotides is constructed in a eukaryotic virus vector.
  • the host cells are infected with said library at an MOI selected from the group consisting of: from about 1 to about 10, about 1 to about 5, 1 to about 2, about
  • the eukaryotic viras vector is an animal virus vector.
  • said eukaryotic viras vector is a plant viras vector.
  • the eukaryotic viras vector is capable of producing infectious viral particles in cells selected from the group consisting of insect cells, plant cells, and mammalian cells.
  • the eukaryotic viras vector is attenuated.
  • the eukaryotic viras vector is capable of producing infectious viral particles in mammalian cells.
  • the attenuation is by genetic mutation.
  • the attenuation is by reversible inhibition of viras replication.
  • the natmally-occurring genome of said eukaryotic virus vector is DNA.
  • the naturally-occurring genome of said eukaryotic virus vector is linear, double- stranded DNA.
  • the eukaryotic viras vector is selected from the group consisting of an adenoviras vector, a herpesvirus vector and a poxvirus vector.
  • the eukaryotic virus vector is a poxviras vector.
  • the poxviras vector is selected from the group consisting of an orthopoxvirus
  • the poxvirus vector is an orthopoxviras vector selected from the group consisting of a vaccinia virus vector and a raccoon poxvirus vector.
  • the orthopoxvirus vector is a vaccinia virus vector.
  • the host cells are permissive for the production of infectious viral particles of said viras.
  • the vaccinia virus is attenuated.
  • the attenuation is by genetic mutation.
  • the attenuation is by reversible inhibition of virus replication.
  • the vaccinia viras vector is derived from strain MVA.
  • the vaccinia viras vector is derived from strain D4R.
  • the insert polynucleotide is in operable associated with a transcriptional control sequence.
  • the transcriptional control sequence functions in the cytoplasm of a poxviras- infected cell.
  • the transcriptional control sequence comprises a promoter.
  • the promoter is constitutive.
  • the promoter is a vaccinia viras p7.5 promoter.
  • the promoter is a T7 promoter.
  • the vector comprises the sequence shown in SEQ ID NO: .
  • the promoter is a synthetic early/late promoter.
  • the vector comprises the sequence shown in SEQ ID NO: .
  • the transcriptional control sequence comprises a transcriptional termination region.
  • Poxviras includes any member of the family Poxviridae, including the subfamililes Chordopoxviridae (vertebrate poxviruses) and Entomopoxviridae (insect poxviruses). See, for example, B. Moss in: Virology, B.N. Fields, D.M. Knipe et al, Eds., Raven Press, p. 2080 (1990).
  • chordopoxviruses comprise, inter alia, the following genera: Orthopoxvirus (e.g., vaccinia); Avipoxvirus (e.g., fowlpox); Capripoxvirus (e.g, sheeppox) Leporipoxvirus (e.g., rabbit (Shope) fibroma, and myxoma); and Suipoxvirus (e.g., swinepox).
  • Orthopoxvirus e.g., vaccinia
  • Avipoxvirus e.g., fowlpox
  • Capripoxvirus e.g, sheeppox
  • Leporipoxvirus e.g., rabbit (Shope) fibroma, and myxoma
  • Suipoxvirus e.g., swinepox
  • the entomopoxviruses comprise three genera: A, B and C.
  • orthopoxviruses are preferred.
  • Vaccinia viras is the prototype orthopoxvirus, and has been developed and is well- characterized as a vector for the expression of heterologous proteins.
  • vaccinia virus vectors particularly those that have been developed to perform trimolecular recombination, are preferred.
  • other orthopoxviruses, in particular, raccoon poxviras have also been developed as vectors and in some applications, have superior qualities.
  • Vaccinia virus like other members of the pox viras group, is distinguished by its large size and complexity.
  • the DNA of vaccinia virus is similarly large and complex.
  • Vaccinia DNA is about 180 kilodaltons in size, for instance, compared with a DNA size of only 3.6 megadaltons for simian viras 40 (SV40).
  • the DNA molecule of vaccinia is double-stranded and terminally crosslinked so that a single stranded circle is formed upon denaturation of the DNA.
  • Vaccinia DNA has been physically mapped using a number of different restriction enzymes and a number of such maps are presented in an article by Panicali et al, J.
  • Vaccinia a eukaryotic virus, reproduces entirely within the cytoplasm of a host cell. It is a lytic viras, i.e. a virus, the replication of which in a cell results in lysis of the cell. The virus is considered non-oncogenic. The virus has been used for approximately 200 years in vaccines for inoculation against smallpox and the medical profession is well acquainted with the properties of the viras when used in a vaccine.
  • the cross-linked double strands of the DNA are characterized by inverted terminal repeats each approximately 8.6 megadaltons in length, representing about 10 kilobasepairs (kbp). Since the central portions of the DNA of all pox viruses are similar, while the terminal portions of the viruses differ more strongly, the responsibility of the central portion for functions common to all the viruses, such as replication, is suggested, whereas the terminal portions appear responsible for other characteristics such as pathogenicity, host range, etc.
  • the portion of the naturally- occurring DNA which is rearranged, removed, or disrupted by the introduction of exogenous DNA thereinto must be non-essential to the viability and stability of the vaccinia virus.
  • Such non-essential portions of the genome have been found to be present in the WR strain of vaccinia virus, for instance within the region present within the L-variant but deleted from the S-variant or within the Hind IH F-fragment of the genome. See, for example, Paoletti et al, U.S. 5,972,597.
  • Attenuated and Defective Viral Vectors Such as Vaccinia Virus.
  • a limitation of wild type vaccinia virus as an expression vector for lethality based selection is that the viras has cytopathic effects in many mammalian cells. The kinetics of these effects is very dependent on the host cells. For many tissue culture lines, cytopathic effects that result in release from a monolayer are not significant until well after 48 to 72 hours infection. This allows a 2 to 3 day time frame for high level expression of recombinant genes and selection of a non-viable or non-adherent phenotype independent of the intrinsic cytopathic effects of the vector.
  • vaccinia virus mutants have been characterized. These may be fully defective mutants, i.e., the production of infectious viras particles requires helper virus, or they may be conditional mutants, e.g., temperature sensitive mutants. Conditional mutants are particularly preferred, in that the virus-infected host cells can be maintained in a non-permissive environment, e.g., at a non-permissive temperature, during the period where host gene expression is required, and then shifted to a permissive environment, e.g., a permissive temperature, to allow viras particles to be produced.
  • a permissive environment e.g., a permissive temperature
  • a fully infectious viras may be "attenuated" by chemical inhibitors which reversibly block viras replication at defined points in the infection cycle.
  • Chemical inhibitors include, but are not limited to hydroxyurea and 5- fluorodeoxyuridine. Virus-infected host cells are maintained in the chemical inhibitor during the period where host gene expression is required, and then the chemical inhibitor is removed to allow viras particles to be produced.
  • Modified Vaccinia Ankara is a highly attenuated strain of vaccinia viras that was derived during over 570 passages in primary chick embryo fibroblasts (Mayr, A. et al, Infection 3:6-14 (1975)). The recovered virus deleted approximately 15% of the wild type vaccima DNA which profoundly affects the host range restriction of the viras. MVA cannot replicate or replicates very inefficiently in most mammalian cell lines. A unique feature of the host range restriction is that the block in non-permissive cells occurs at a relatively late stage of the replication cycle. Expression of viral late genes is relatively unimpaired but virion morphogenesis is interrupted (Suter, G.
  • MVA Proc Natl Acad Sci USA 5P:10847-10851 (1992); Carroll, M.W. and Moss, B., Virology 235:198-211 (1997)).
  • the high levels of viral protein synthesis even in non-permissive host cells make MVA an especially safe and efficient expression vector.
  • MVA cannot complete the infectious cycle in most mammalian cells, in order to recover infectious virus for multiple cycles of selection it will be necessary to complement the MVA deficiency by coinfection or superinfection with a helper virus that is itself deficient and that can be subsequently separated from infectious MVA recombinants by differential expansion at low MOI in MVA permissive host cells.
  • vaccinia viras that are deficient in an essential early gene have been shown to have greatly reduced inhibitory effects on host cell protein synthesis.
  • Attenuated poxviruses which lack defined essential early genes have also been described. See, e.g., U.S. Patent No. 5,766,882, by Falkner, et al.
  • essential early genes which may be rendered defective include, but are not limited to the vaccinia viras 17L, F18R, D13L, D6R, A8L, J1R, E7L, F11L, E4L, I1L, J3R, J4R, H7R, and A6R genes.
  • a preferred essential early gene to render defective is the D4R gene, which encodes a uracil DNA glycosylase enzyme.
  • Vaccinia viruses defective in defined essential genes are easily propagated in complementing cell lines which provides the essential gene product.
  • complementation refers to a restoration of a lost function in trans by another source, such as a host cell, transgenic animal or helper virus. The loss of function is caused by loss by the defective virus of the gene product responsible for the function.
  • a defective poxviras is a non-viable form of a parental poxvirus, and is a form that can become viable in the presence of complementation.
  • the host cell, transgenic animal or helper virus contains the sequence encoding the lost gene product, or "complementation element.”
  • the complementation element should be expressible and stably integrated in the host cell, fransgenic animal or helper virus, and preferably would be subject to little or no risk for recombination with the genome of the defective poxvirus.
  • Viruses produced in the complementing cell line are capable of infecting non-complementing cells, and further are capable of high-level expression of early gene products. However, in the absence of the essential gene product, host shut-off, DNA replication, packaging, and production of infectious virus particles do not take place.
  • selection of desired insert polynucleotides expressed in a complex library constructed in vaccima virus is accomplished through coupling induction of expression of the complementation element to expression of the insert polynucleotide. Since the complementation element is only expressed in those host cells expressing the insert polynucleotide, only those host cells will produce infectious viras which is easily recovered.
  • inactivation of the library constructed in a virus vector is carried out by treating a sample of the library constructed in a virus vector with 4'-aminomethyl-trioxsalen (psoralen) and then exposing the virus vector to ultraviolet (UN) light.
  • psoralen 4'-aminomethyl-trioxsalen
  • UN ultraviolet
  • Psoralen and UN inactivation of viruses is well known to those of ordinary skill in the art. See, e.g., Tsung, K., et al, J. Virol. 70:165-111 (1996), which is incorporated herein by reference in its entirety.
  • Psoralen treatment typically comprises incubating a cell-free sample of the virus vector with a concentration of psoralen ranging from about 0.1 ⁇ g/ml to about 20 ⁇ g/ml, preferably about 1 ⁇ g/ml to about 17.5 ⁇ g/ml, about 2.5 ⁇ g ml to about 15 ⁇ g/ml, about 5 ⁇ g/ml to about 12.5 ⁇ g/ml, about 7.5 ⁇ g/ml to about 12.5 ⁇ g/ml, or about 9 ⁇ g/ml to about 11 ⁇ g/ml.
  • the concentration of psoralen may be about 0.1 ⁇ g/ml, 0.5 ⁇ g/ml, 1 ⁇ g/ml, 2 ⁇ g/ml, 3 ⁇ g/ml, 4 ⁇ g/ml, 5 ⁇ g/ml, 6 ⁇ g/ml, 7 ⁇ g/ml, 8 ⁇ g/ml, 9 ⁇ g/ml, 10 ⁇ g/ml, 11 ⁇ g/ml, 12 ⁇ g/ml, 13 ⁇ g/ml, 14 ⁇ g/ml, 15 ⁇ g/ml, 16 ⁇ g/ml, 17 ⁇ g/ml, 18 ⁇ g/ml, 19 ⁇ g/ml, or 20 ⁇ g/ml.
  • the concentration of psoralen is about 10 ⁇ g/ml.
  • the term "about” takes into account that measurements of time, chemical concentration, temperature, pH, and other factors typically measured in a laboratory or production facility are never exact, and may vary by a given amount based on the type of measurement and the instrumentation used to make the measurement.
  • the incubation with psoralen is typically carried out for a period of time prior to UV exposure.
  • This time period preferably ranges from about one minute to about 20 minutes prior to the UV exposure.
  • the time period ranges from about 2 minutes to about 19 minutes, from about 3 minutes to about 18 minutes, from about 4 minutes to about 17 minutes, from about 5 minutes to about 16 minutes, from about 6 minutes to about 15 minutes, from about 7 minutes to about 14 minutes, from about 8 minutes to about 13 minutes, or from about 9 minutes to about 12 minutes.
  • the incubation time may be about 1 minute, about 2 minutes, about three minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, about 10 minutes, about 11 minutes, about 12 minutes, about 13 minutes, about 14 minutes, about 15 minutes, about 16 minutes, about 17 minutes, about 18 minutes, about 19 minutes, or about 20 minutes. More preferably, the incubation is carried out for 10 minutes prior to the UV exposure.
  • the psoralen-treated viruses are then exposed to UV light.
  • the UV may be of any wavelength, but is preferably long-wave UV light, e.g., about
  • Exposure to UV is carried out for a time period ranging from about
  • the time period ranges from about
  • the incubation time may be about 0.1 minute, about 0.5 minute, about 1 minute, about 2 minutes, about three minutes, about 4 minutes, about 5 minutes, about 6 minutes, about 7 minutes, about 8 minutes, about 9 minutes, about 10 minutes, about 11 minutes, about 12 minutes, about 13 minutes, about 14 minutes, about 15 minutes, about 16 minutes, about 17 minutes, about 18 minutes, about 19 minutes, or about 20 minutes.
  • the viras vector is exposed to UV light for a period of about 5 minutes.
  • the terms "vaccima virus” and "poxviras" are used interchangeably herein.
  • the preferred embodiments relating to vaccinia virus may be modified in ways apparent to one of ordinary skill in the art for use with any poxviras vector. In the direct selection method, vectors other than poxviras or vaccinia virus may be used.
  • Vaccinia viras vectors are not currently used to identify previously unknown genes of interest from a complex population of clones, such as a cDNA or other library, because a high efficiency, high titer-producing method of cloning does not exist for vaccinia.
  • the standard methods of cloning in vaccinia viras are in vivo homologous recombination and in vitro direct ligation. Using homologous recombination, the cloning efficiency is in the range of approximately 0.1% or less, and although the cloning efficiency using direct ligation is higher, the resulting titer is relatively low.
  • the use of vaccinia viras vector has been limited to the cloning of previously isolated DNA for the purposes of protein expression and vaccine development.
  • Tri-molecular recombination is a novel, high efficiency, high titer- producing method for cloning in vaccima virus.
  • the present inventor has achieved cloning efficiencies of at least 90%, and titers at least 30-fold, at 100-fold, i.e., at least 2 orders of magnitude higher, than those obtained by direct ligation.
  • the invention provides a method of high efficiency cloning using a vaccinia virus vector, comprising tri-molecular recombination.
  • tri-molecular recombination or a “tri-molecular recombination method” is meant a method of producing a vaccinia virus genome which contains insert polynucleotide (e.g. insert DNA), comprising introducing two nonhomologous fragments of a vaccinia viras genome and a transfer vector or transfer polynucleotide (e.g. transfer DNA) containing insert polynucleotide (e.g. DNA) into a recipient cell, and allowing the three polynucleotide (e.g. DNA) molecules to recombine in vivo.
  • a single vaccinia virus genome molecule is produced which comprises each of the two vaccinia genome fragments and the insert polynucleotide (e.g. DNA).
  • the tri-molecular recombination method comprises: (a) providing a recipient cell which comprises packaging function; (b) introducing into the cell (i) transfer vector or transfer polynucleotide (e.g. DNA), and (ii) two fragments of vaccinia viras genome; and (c) culturing the cells under conditions such that the transfer polynucleotide (e.g. DNA) and the two fragments undergo trimolecular recombination.
  • transfer vector or transfer polynucleotide e.g. DNA
  • cloning is meant the use of in vitro and in vivo recombination techniques to insert one or a plurality of polynucleotide sequences into a vector.
  • in vitro and in vivo recombination techniques to insert one or a plurality of polynucleotide sequences into a vector.
  • it is often necessary to employ methods for generating polynucleotide fragments, for joining the fragments to vector molecules, for introducing the composite polynucleotide molecule into a host cell in which it can replicate, and for selecting the clone having a polynucleotide insert, for example by virtue of a resistance marker such as thymidine kinase " (tk ⁇ ), from among the cells containing either no vector or vector alone without an insert.
  • a resistance marker such as thymidine kinase " (tk ⁇ )
  • cloning efficiency or “efficiency of cloning” is meant the ratio of recombinant virus to total virus produced during tri-molecular recombination. As shown in Example 2, the efficiency may be calculated by dividing the titer of recombinant virus by the titer of total virus and multiplying by 100%. For example, the titer is determined by plaque assay of crude virus stock on appropriate cells either with selection (e.g., for recombinant viras) or without selection (e.g., for recombinant viras plus wild type viras). Methods of selection are well-known in the art and include BrdU resistance due to disruption of the tk gene as described herein and other well-known methods. See Examples 1 and 2.
  • high efficiency cloning is meant a cloning efficiency of at least
  • a cloning efficiency of at least 2%, 2.264%, 3%, 3.5%, 3.571%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, or 99%.
  • a number of selection systems may be used, including but not limited to the thymidine kinase such as herpes simplex virus thymidine kinase (Wigler, et al, Cell 11:223 (1977)), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, Proc. Natl. Acad. Sci. USA 48:2026 (1962)), and adenine phosphoribosyltransferase (Lowy, et al, Cell 22:817 (1980)) genes which can be employed in tk " , hgprt " or aprf cells, respectively.
  • thymidine kinase such as herpes simplex virus thymidine kinase (Wigler, et al, Cell 11:223 (1977)), hypoxanthine-guanine phosphoribosyltransferase (Szy
  • antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler, et al, Natl. Acad. Sci. USA 77:3561 (1980); O ⁇ are, et al, Proc. Natl. Acad. Sci. USA 75:1527 (1981)); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl Acad. Sci. USA 75:2072 (1981)); neo, which confers resistance to the aminoglycoside G-418 (Colberre- Garapin, et al, J. Mol. Biol. 150:1 (1981)); and hygro, which confers resistance to hygromycin (Santerre, et al, Gene 30: 147 (1984)).
  • the two fragments or arms of the vaccinia virus genome contain all the genes necessary for viral replication and for production of infectious viral particles. See the Examples for examples of structures of vaccinia viras vector useful in the trimolecular recombination method. Preferred structures are presented in Examples 1 and 2. See Falkner et al, U.S. 5,770,212 for guidance concerning essential regions for vaccinia replication.
  • virus-encoded function also referred to as "packaging function,” includes an RNA polymerase that recognizes the transfected vaccinia DNA as a template and initiates transcription and, ultimately, replication of the transfected DNA. See Dorner, et al. U.S. 5,445,953.
  • the recipient cell preferably contains packaging function.
  • the packaging function may be provided by helper viras, i.e., a viras that, together with the transfected naked genomic DNA, provides appropriate proteins and factors necessary for replication and assembly of progeny virus.
  • the helper virus may be a closely related viras, for instance, a poxviras of the same poxvirus subfamily as vaccima, whether from the same or a different genus. In such a case it is advantageous to select a helper virus which provides an RNA polymerase that recognizes the transfected DNA as a template and thereby serves to initiate transcription and, ultimately, replication of the transfected DNA.
  • heterologous viras to package the modified DNA molecule, such as the use of temperature sensitive (ts) vaccinia, fowlpox, or ectromelia (mouse pox) virus as a helper for vaccinia viras constructs, minimizes recombination events between the helper viras genome and the transfected genome which take place when homologous sequences of closely related viruses are present in one cell. See Fenner & Comben, Virology 5:530 (1958); Fenner, Virology 5:499 (1959).
  • the necessary packaging function in the recipient cell is supplied by a genetic element other than a helper viras, such as a plasmid or refroviras or other expression vector suitable for expressing the required helper viras function. See Dorner , et al. U.S. 5,445,953.
  • the two vaccinia genomic fragments are unable to ligate with each other, i.e., they do not contain compatible cohesive ends or alternatively, they have been treated with a dephosphorylating enzyme. Further, the two fragments are nonhomologous, such that they are unable to recombine with one another.
  • insert polynucleotide e.g. "insert DNA”
  • polynucleotides are also referred to herein as nucleic acid.
  • An insert polynucleotide may be DNA.
  • a polynucleotide (e.g. DNA) segment may be naturally occurring, non naturally occurring, synthetic, or a combination thereof.
  • a polynucleotide (e.g. DNA) segment may be any length. It may be endogenous or exogenous to the vector, such as vaccinia virus, or it may be endogenous or exogenous to the host cell, it may be a previously known sequence or an unknown sequence.
  • Insert polynucleotides may also be referred to herein by the term “candidate polynucleotides” or “test polynucleotides” or other equivalents, especially in reference to selection methods of the invention.
  • transfer plasmid is meant a vector containing insert polynucleotide (e.g. DNA) positioned between a 5' flanking region and a 3' flanking region.
  • the 5' flanking region shares homology with one of the vaccinia genome fragments
  • the 3' flanking region shares homology with the other of the vaccima genome fragments.
  • the transfer plasmid contains a suitable promoter, such as a strong, constitutive vaccinia promoter, upstream of the insert DNA.
  • vector means a polynucleotide (e.g. DNA) construct containing a polynucleotide (e.g.
  • DNA sequence which is operably linked to a suitable control sequence capable of effecting the expression of the polynucleotide (e.g. DNA) in a suitable host.
  • control sequences include a promoter to effect transcription, an optional operator sequence to control such transcription, a sequence encoding suitable mRNA ribosome binding sites, sequences which control the termination of transcription and translation, and other confrol sequences as are well known in the art.
  • the vector may be a plasmid, a phage particle, or simply a potential genomic insert. Once transformed into a suitable host, the vector may replicate and function independently of the host genome, or may in some instances, integrate into the genome itself. In the present specification, "plasmid” and “vector” are sometimes used interchangeably.
  • Typical expression vectors for mammalian cell culture expression are based on pRK5 (EP 307,247), pSV16B (WO 91/08291) and pVL1392 (Pharmingen).
  • transfer plasmid is not limited to a vector or a plasmid. Any polynucleotide (e.g. DNA) segment in circular or linear or other suitable form may act as a vehicle for transferring the polynucleotide (e.g. DNA) insert to the vaccinia vector in the tri-molecular recombination method.
  • Other suitable "plasmids” include lambda phage, polynucleotide (e.g. DNA) fragments, etc., as described herein or otherwise known in the art.
  • a plurality of plasmids maybe a "primary library" such as those described herein for lambda.
  • the insert polynucleotides are operably associated with T7 or poxvirus expression control sequences, more preferably, strong constitutive poxvirus promoters.
  • Nucleic acid is "operably associated" when it is placed into a functional relationship with another nucleic acid sequence.
  • This can be a gene and a regulatory sequence(s) which are connected in such a way as to permit gene expression when the appropriate molecules (e.g., transcriptional activator proteins) are bound to the regulatory sequences(s).
  • DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • "operably associated" means that the polynucleotide (e.g.
  • DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • control sequences polynucleotide (e.g. DNA) sequences necessary for the expression of an operably associated coding sequence in a particular host organism.
  • the confrol sequences that are suitable for prokaryotes include a promoter, optionally an operator sequence, and a ribosome binding site.
  • Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.
  • the control sequence is a transcriptional control sequence, it is one which functions in the cytoplasm of a poxviras-infected cell.
  • Poxviras transcriptional control sequences comprise a promoter and a transcription termination signal. Gene expression in poxviruses is temporally regulated, and promoters for early, intermediate, and late genes possess different structures. Certain poxviras genes are expressed constitutively (i.e., early to late), and promoters for these "early-late" genes bear hybrid structures. Synthetic early-late promoters have also been developed. See Hammond J.M., et al, J. Virol. Methods 66:135-13 (1997); Chakrabarti S., et al, Biotechniques 23:1094-1097 (1997). In the present invention, any poxvirus promoter may be used, but use of early, late, or constitutive promoters may be desirable based on the host cell and/or selection scheme chosen. Typically, the use of constitutive promoters is preferred.
  • Examples of early promoters include the 7.5-kD promoter (also a late promoter), the DNA pol promoter, the tk promoter, the RNA pol promoter, the 19-kD promoter, the 22-kD promoter, the 42-kD promoter, the 37-kD promoter, the 87-kD promoter, the H3' promoter, the H6 promoter, the DI promoter, the D4- promoter, the D5 promoter, the D9 promoter, the D12 promoter, the 13 promoter, the Ml promoter, and the N2 promoter.
  • Example of late promoters include the 7.5-kD promoter, the MIL promoter, the 37-kD promoter, the 11 -kD promotor, the 11L promoter, the 12L promoter, the 13L promoter, the 15L promoter, the 17L promoter, the 28- kD promoter, the H1L promoter, the H3L promoter, the H5L promoter, the H6L promoter, the H8L promoter, the DHL promoter, the D12L promotor, the D13L promoter, the AIL promoter, the A2L promoter, the A3L promoter, and the P4b promoter.
  • Preferred constitutive promoters for use in the present invention include the synthetic early-late promoters described by Hammond and Chakrabarti, the MH-5 early-late promoter, and the 7.5-kD or "p7.5" promoter. Examples utilizing these promoters are disclosed herein.
  • the insert polynucleotides may be operably associated with T7 phage promoter, which is active in cells in which T7 RNA polymerase is expressed, for the regulation of the expression of insert polynucleotides.
  • the host cell is stably transfected with a polynucleotide encoding the T7 RNA polymerase under the control of a mammalian promoter as described prevuously (O. Elroy-Stefn, B. Moss. 1990 Proc. Natl. Acad. Sci. USA 87: 6743-6747).
  • T7 RNA polymerase may also be provided by co-infection with a T7 RNA polymerase recombinant helper viras, such as fowlpox viras (P. Britton, P. Green, S. Kottier, K.L. Mawditt, Z. Penzes, D. Cavanagh, M.A. Skinner. 1996 J. General Virology 11: 963-967).
  • helper viras such as fowlpox viras (P. Britton, P. Green, S. Kottier, K.L. Mawditt, Z. Penzes, D. Cavanagh, M.A. Skinner. 1996 J. General Virology 11: 963-967).
  • the T7 polymerase system adapted to vaccinia gives higher levels of expression relative to native vaccinia promoters. (Eckert D. and Merchlinsky M. J Gen Virol 80 ( Pt 6): 1463-9 (1999); E
  • CPE cytopathic effect
  • this time frame may not be sufficient for certain selection methods, especially where higher MOIs are used, and further, the time before the onset of CPE may be shorter in a desired cell line.
  • viras vectors particularly poxviras vectors such as vaccinia viras, with attenuated cytopathic effects so that, wherever necessary, the time frame of selection can be extended.
  • recipient cell or "host cell” or “cell” is meant a cell or plurality of cells into which polynucleotides (e.g. DNA) is introduced for the tri-molecular recombination method or a direct or indirect selection method described below, preferably a eukaryotic cell or cell line, preferably an animal, vertebrate, mammalian, rodent, mouse, primate, or human cell or cell line. Additional examples of host cells are disclosed herein. The host cells may also comprise packaging function.
  • Polynucleotides e.g.
  • DNA may be introduced into host cells by any method known to those of skill, for example, by lipofection (such as with anionic liposomes (see, e.g., Feigner et al, Proc. Natl. Acad. Sci. U.S.A. 84:1 '413 (1987)) or cationic liposomes (see, e.g., Brigham, K.L. et al. Am. J Med Sci. 2P5(4):278-2821 (1989); U.S. Patent No.
  • lipofection such as with anionic liposomes (see, e.g., Feigner et al, Proc. Natl. Acad. Sci. U.S.A. 84:1 '413 (1987)) or cationic liposomes (see, e.g., Brigham, K.L. et al. Am. J Med Sci. 2P5(4):278-2821 (1989); U.S. Patent No.
  • the nucleic acid can be complexed with a cationic liposome, such as DOTMA:DOPE, DOTMA, DOPE, DC-cholesterol, DOTAP, Transfectam ® (Promega), Tfx ® (Promega), LipoTAXI TM (Stratagene), PerFect Lipid TM (Invitrogen), SuperFect (Qiagen).
  • a cationic liposome such as DOTMA:DOPE, DOTMA, DOPE, DC-cholesterol, DOTAP, Transfectam ® (Promega), Tfx ® (Promega), LipoTAXI TM (Stratagene), PerFect Lipid TM (Invitrogen), SuperFect (Qiagen).
  • a cationic liposome such as DOTMA:DOPE, DOTMA, DOPE, DC-cholesterol, DOTAP, Transfectam ® (Promega), Tfx ® (Prom
  • Trimolecular recombination has been used by the present inventors to construct cDNA libraries in vaccinia virus with titers of the order of 10? pfu. There are several factors that limit the complexity of these cDNA or other libraries. These include: the size of the primary cDNA library or other library that can be constructed in a plasmid vector, and the labor involved in the purification of large quantities (hundreds of micrograms) of vaccinia viras, poxvirus, or other vector DNA. Modifications of trimolecular recombination that would allow for vector (e.g. vaccinia or other poxvirus DNA) recombination with transfer polynucleotides (e.g.
  • Lambda phage vectors have several advantages over plasmid vectors for construction of cDNA libraries or other libraries. Plasmid libraries (e.g. cDNA or other DNA insert) or linear polynucleotide (e.g. DNA) libraries are introduced into bacterial cells by chemical/heat shock transformation, or by elecfroporation. Bacterial cells are often transformed preferentially by smaller plasmids, resulting in a potential loss of representation of longer polynucleotides (e.g., cDNA or other insert DNA) in a library.
  • Plasmid libraries e.g. cDNA or other DNA insert
  • linear polynucleotide e.g. DNA
  • transformation is a relatively inefficient process for introducing foreign polynucleotides (e.g., cDNA or other insert DNA) into a cell requiring the use of expensive commercially prepared competent bacteria in order to construct a library (e.g. a cDNA library).
  • foreign polynucleotides e.g., cDNA or other insert DNA
  • lambda phage vectors can tolerate polynucleotide inserts
  • Lambda vectors are packaged into virions in vitro using high efficiency commercially available packaging extracts so that the recombinant viral (lambda) genomes can be introduced into bacterial cells by infection. This results in primary libraries with higher titers and better representation of large polynucleotide (e.g. cDNA or DNA) inserts than is commonly obtained in plasmid libraries.
  • the lambda vector must be modified to include vaccima virus DNA sequences that allow for homologous recombination with the vaccinia virus DNA.
  • the vaccinia viras Hindlll J fragment (vaccinia tk gene) contained in plasmid p7.5/ATG0/tk can be excised using HindlH and SnaBI (3 kb of vaccinia DNA sequence), and subcloned into the Hindlfl/SnaBI sites of pT7Blue3 (Novagen cat no.
  • the vaccinia tk gene can be excised from this vector with Sad and SnaBI and inserted into the Sacl/Smal sites of Lambda Zap Express (Stratagene) to create lambdaNtk.
  • the lambdaNtk vector will contain unique ⁇ otI, BamHI, Smal, and Sail sites for insertion of cD ⁇ A downstream of the vaccinia 7.5k promoter.
  • Libraries e.g., cD ⁇ A or other D ⁇ A libraries
  • D ⁇ A from a library (e.g. cD ⁇ A or other library) constructed in lambdaNtk, or, any similar bacteriophage that includes polynucleotide (e.g. cD ⁇ A inserts or other insert D ⁇ A) with flanking vaccinia D ⁇ A sequences to promote homologous recombination, can be employed to generate cD ⁇ A (or other insert D ⁇ A or polynucleotide) recombinant vaccima viras.
  • Methods are well known in the art for excising a plasmid from the lambda genome by coinfection with a helper phage (ExAssist phage, Stratagene cat no. 211203).
  • Mass excision from a lambda based library creates an equivalent library (e.g. cD ⁇ A or other library) in a plasmid vector.
  • Plasmids excised from, for example, the lambdaNtk polynucleotide (e.g. cD ⁇ A) library will contain the vaccinia tk sequences flanking the polynucleotide (e.g. cD ⁇ A or other D ⁇ A) inserts.
  • This plasmid D ⁇ A can then be used to construct vaccinia recombmants by trimolecular recombination.
  • Another embodiment of this method is to purify the lambda D ⁇ A directly from the initial lambdaNtk library, and to transfect this recombinant viral (lambda) D ⁇ A or fragments thereof together with the two large vaccinia viras D ⁇ A fragments for trimolecular recombination.
  • Cells e.g. packaging or host cells
  • a restriction endonuclease that recognizes a unique site introduced into the vaccinia virus genome.
  • the restriction endonuclease will digest the vaccinia DNA, generating "arms" that can only be repaired, i.e., rejoined, by trimolecular recombination.
  • restriction endonucleases include the bacterial enzymes Notl and Apal, the Yeast endonuclease VDE (R. Hirata, et al, J.
  • Confluent monolayers of these cells will be induced with doxycycline and then infected with v7.5/tk (unique Notl site in tk gene), and transfected with polynucleotide (e.g. cDNA or DNA) recombinant transfer plasmid or transfer polynucleotide (e.g. transfer DNA) or lambda phage or phagemid DNA.
  • polynucleotide e.g. cDNA or DNA
  • transfer polynucleotide e.g. transfer DNA
  • lambda phage or phagemid DNA e.g. transfer DNA
  • Digestion of host cell chromosomal DNA by Notl is not expected to prevent production of infectious viral recombinants because the host cells are not required to proliferate during viral replication and virion assembly.
  • a vaccinia strain in another embodiment of this method to generate vaccinia arms in vivo, is constructed that contains a unique endonuclease site in the tk gene or other sequence and the cDNA encoding the endonuclease under the control of the T7 bacteriophage promoter at another site in the vaccinia genome. Infection of cells that express the T7 RNA polymerase would result in expression of the endonuclease, and subsequent digestion of the vaccima DNA by the endonuclease.
  • the endonuclease is Notl.
  • the v7.5/tk strain of vaccinia is modified by insertion of a cassette containing the cDNA encoding Notl with expression controlled by the T7 promoter into the Hindi ⁇ C or F region (Coupar, E.H.B. et al, Gene 68:1-10 (1988); Flexner, C. et al, Nature 330:259-262 (1987)), generating v7.5/tk/T7NotI.
  • a cell line is stably transfected with the cDNA encoding the T7 RNA polymerase under the control of a mammalian promoter as described (O. Elroy-Stein, B.
  • the T7 RNA polymerase can be provided by co-infection with a T7 RNA polymerase recombinant helper viras, such as fowlpox virus (P. Britton, et al, J. General Virology 77:963-967 (1996)).
  • trimolecular recombination employing these various sfrategies for generation of large vaccinia DNA fragments in vivo is that digestion of the vaccinia DNA may but does not need to precede recombination. It suffices that only recombinant viras escapes destraction by digestion. This contrasts with trimolecular recombination employing transfection of vaccinia DNA digested in vitro where, of necessity, vaccinia DNA fragments are created prior to recombination. It is possible that the opportunity for bimolecular recombination prior to digestion will yield a greater frequency of recombinants than can be obtained through trimolecular recombination following digestion.
  • the trimolecular recombination method is used in the production of a library, preferably an expression library.
  • a cDNA library is prepared by extracting total RNA, mRNA, size-fractionated RNA, etc. from a cell, synthesizing a series of complementary double-stranded cDNA fragments from the RNA and introducing these cDNA fragments into cells in tissue culture, by, for example, first cloning the fragments into a vector, preferably a poxviras vector.
  • the cells are maintained under conditions which allow them to express the protein, or alternatively only the transcript, encoded by the cDNA.
  • the tri-molecular recombination method is used to produce a genomic library or a combinatorial library.
  • the methods of the invention are applicable to a wide variety of libraries including mixtures of cDNAs cloned in sense or anti-sense orientation, mixtures of fragments of genomic DNA or cDNA, fragments of individual cDNAs and mixtures of artificial sequences.
  • Libraries include combinatorial libraries made up of natural and/or artificial sequences.
  • the library is an expression library.
  • Libraries may comprise a plurality of insert polynucleotides (e.g. DNAs).
  • the insert polynucleotides e.g. DNAs
  • the insert polynucleotides may comprise any DNA or other polynucleotide segment as described above.
  • cDNA complementary or copy DNA produced from an
  • RNA template by the action of RNA-dependent DNA polymerase (reverse transcriptase).
  • a "cDNA clone” means a duplex DNA sequence complementary to an RNA molecule of interest, carried in a cloning vector.
  • the cDNA may be from mRNA, a portion of a mRNA, a pre-mRNA including introns, or an intron.
  • the cDNA may be from viral RNA.
  • Molecular cloning methods for ds cDNA have been reviewed, for example, by Williams, "The Preparation and Screening of a cDNA Clone Bank," in Williamson, ed., Genetic Engineering, Vol. 1, p.
  • cDNA library is meant a collection of recombinant DNA molecules containing cDNA inserts which together comprise all or a portion of the genome of an organism.
  • a cDNA library may be prepared by art- recognized methods described, for example, in Maniatis et al, Molecular Cloning: A Laboratory Manual, supra.
  • RNA is first isolated from the cells of an organism from whose genome it is desired to clone a particular gene.
  • Preferred for the purposes of the present invention are animal cells, including insect cells, mammalian cells, and particularly human cells, or cell lines derived from these organisms.
  • RNA can be isolated from a tumor cell derived from an animal tumor, and preferably from a human tumor.
  • a library may be prepared from, for example, a human adrenal tumor, but any tumor may be used.
  • expression library is meant a recombinant vector in which insert polynucleotide (e.g. DNA) is cloned and which allows expression of at least RNA, and preferably also expression of protein encoded by the insert polynucleotide (e.g. DNA).
  • the insert polynucleotide may be cloned in two vectors for producing a transfer plasmid containing a polynucleotide (e.g. DNA) insert in each of two orientations.
  • the invention also provides a kit for producing a transcription expression library comprising poxviras and two transfer plasmids for inserting polynucleotide (e.g. DNA) in each orientation.
  • the invention provides a kit for producing a protein expression library comprising a vaccinia virus genome or two fragments thereof, and three vectors for producing a transfer plasmid containing a polynucleotide (e.g. DNA) insert in each of three translation reading frames.
  • vaccima viras now may be used to identify previously unknown genes of interest from a complex population of clones, such as a cDNA or other library. Further, its ability to replicate in nondividing cells and its potential to express proteins at a high level in nondividing cells allows vaccinia viras to be used for cloning and isolating sequences that positively or negatively affect growth, differentiation, or cell viability.
  • the invention provides a method of producing a library in nondividing cells comprising the trimolecular recombination method.
  • Nondividing cells include terminally differentiated cells.
  • Nondividing cells include musculoskeletal cells such as, for example, chondrocytes, osteoblast, osteoclasts, and myocytes.
  • Other examples of nondividing cells are fully differentiated blood and epidermal cells, neurons, glial cells, kidney and liver cells.
  • the library is produced in a cell line which efficiently produces recombinant vector, such as BSC-1 or HeLa cells for libraries cloned in vaccinia viras, and the progeny recombinant vector population is transferred to a second population of cells in which the direct or indirect selection method is used.
  • the library may be amplified by several passages in the efficient cell line before the direct or indirect selection method is used in the cell line in which the insert polynucleotide is functionally selected.
  • the invention provides a method of isolating an insert polynucleotide in a cell which is dead or which has ceased dividing.
  • the present inventor has used the tri-molecular recombination method to select polynucleotides encoding a target epitope recognized by particular cytotoxic T lymphocytes (CTLs).
  • CTLs cytotoxic T lymphocytes
  • monolayer of calls was infected with a complex library produced using tri-molecular recombination, the monolayer was incubated under conditions allowing expression of the target epitope, and epitope-specific
  • CTLs were added .to the cell monolayer. After CTL-mediated induction of lysis in cells expressing the target epitope, vaccinia viras DNA was recovered from cells that had become nonadherent or that had lysed.
  • Herpesvirus A method has been described for the generation of helper viras free stocks of recombinant, infectious Herpes Simplex Virus amplicons (T.A. Stavropoulos, and CA. Strathdee. J. Virology 72:7137-7143 (1998)).
  • a polynucleotide library e.g. cDNA or other library constructed in such a plasmid amplicon vector could be packaged into a library of infectious amplicon particles using this method.
  • Such a library could be employed in the indirect or direct selection methods (e.g. lethality/adherence based selection methods) of this invention.
  • a library e.g.
  • cDNA or other library constructed in a herpes amplicon vector could be used to infect a monolayer of target cells. Then, for example, tumor specific CTL added to this infected monolayer would induce lysis of cells that express polynucleotides (e.g. cDNA or other DNA) encoding the target antigen. Amplicons recombinant for such polynucleotides (e.g. cDNA or other DNA) might then be recovered from non-viable cells released from the monolayer. However, since in the absence of helper virus the amplicons are replication defective and are not packaged into infectious particles, the amplicon vector recovered from these selected cells could not be employed directly to infect fresh target cells for another cycle of selection.
  • polynucleotides e.g. cDNA or other DNA
  • Amplicon plasmid DNA could then be re-extracted from bacteria following antibiotic selection and packaged into infectious viral particles by cofransfection of amplicon DNA and packaging defective HSV genomic DNA into packaging cells.
  • the infectious amplicon particles harvested could then be used to infect a fresh population of target cells for another round of selection.
  • a more rapid means of recovering infectious amplicons would be, for example, to fransfect cells employed as targets for CTL selection with the packaging defective HSV genomic DNA. This would, however, severely restrict the functional host range for selection to a few highly transfectable cell lines, i.e. Cos 7 and 293 cells.
  • Adenoviras Methods have been described for the production of recombinant Adenoviras (Miyake, S. et al, Proc. Natl Acad. Sci. USA P3:1320-1324 (1996); He, T.C. et al, Proc. Natl. Acad. Sci. USA 95:2509- 2514 (1998)). Although current methods yield relatively low viral titers and construction of a representative (e.g. cDNA) library in adenoviras has not been reported, it is possible that a cDNA library or other library could be constructed in an Adenoviras vector using either of these methods.
  • Insertion of cDNA or other insert polynucleotide (e.g. DNA) into the E4 region of Adenoviras results in a replication competent recombinant viras.
  • a library could be employed in the direct (e.g. lethality/adherence based) and indirect selection methods of this invention.
  • a tumor cDNA library constructed in an adenoviras vector could be used to infect a monolayer of target (e.g. host) cells.
  • Tumor specific CTL added to this infected monolayer would induce lysis of cells that express cDNA recombinants encoding the target antigen.
  • Adenoviras recombinants for the cDNA encoding the target antigen recognized by CTL could then be recovered from cells released from the monolayer as described for selection from a vaccinia virus cDNA library. The virus recovered from the selected cells is infectious and could be employed for additional cycles of selection.
  • Vaccinia Virus hi comparison with either herpesviras or adenoviras, there are significant advantages to vaccinia viras based vectors for the construction and manipulation of libraries, preferably representative and other cDNA libraries.
  • libraries preferably representative and other cDNA libraries.
  • the titers that have been reported for primary adenoviral constructs are relatively low compared to what can be achieved through trimolecular recombination with vaccima virus.
  • Vaccinia viras because it replicates in the cytoplasm of the host cell, may undergo more frequent recombination events with transfer DNA than other mammalian DNA viruses like adenoviras and herpesvirus, which replicate in the nucleus and are presumed to require translocation of transfer plasmids to the nucleus for recombination.
  • the frequency of transfer plasmid recombination events in vaccima virus infected cells may be further enhanced because vaccinia viras, but not adenoviras or herpesvirus, replicates plasmids in the cytoplasm in a sequence independent manner (M. Merchlinsky, B. Moss. Cancer Cells 6 ⁇ :87 ⁇ 93 (1988)).
  • the invention provides a method of selecting target polynucleotides, directly or indirectly, based on a phenotype such as nonadherence or nonviability.
  • the invention provides a method of directly selecting a target polynucleotide comprising: (a) providing a plurality of cells which comprise vector comprising insert polynucleotides, and (b) culturing the cells under conditions such that the insert polynucleotides are expressed and wherein a cell expressing a target polynucleotide becomes nonadherent, (c) collecting or removing the nonadherent cell or cell contents, thereby directly selecting the target polynucleotide.
  • polynucleotides may be selected based on a phenotype such as adherence or viability, or another phenotype, as is further described herein.
  • the invention provides a method of selecting target polynucleotides based on any altered phenotype.
  • altered phenotype or “changed physiology” or other grammatical equivalents herein is meant that the phenotype of the cell is altered in some way, preferably in some detectable and/or measurable way.
  • a strength of the present invention is the wide variety of cell types and potential phenotypic changes which may be tested using the present methods. Accordingly, as described above, any phenotypic change which may be observed, detected, or measured may be the basis of the screening methods herein.
  • Suitable phenotypic changes include, but are not limited to: gross physical changes such as changes in cell morphology, cell growth, cell viability, adhesion to substrates or other cells, and cellular density; changes in the expression of one or more RNAs, proteins, lipids, hormones, cytokines, or other molecules; changes in the equilibrium state (i.e.
  • RNAs, proteins, lipids, hormones, cytokines, or other molecules changes in the localization of one or more RNAs, proteins, lipids, hormones, cytokines, or other molecules; changes in the bioactivity or specific activity of one or more RNAs, proteins, lipids, hormones, cytokines, receptors, or other molecules; changes in the secretion of ions, cytokines, hormones, growth factors, or other molecules; alterations in cellular membrane potentials, polarization, integrity or transport; changes in infectivity, susceptibility, latency, adhesion, and uptake of viruses and bacterial pathogens; etc.
  • target polynucleotide can change the phenotype of the cell in some detectable and/or measurable way. See, e.g., below, and Examples 24, and 26-29.
  • An altered phenotype may be detected by any method known in the art, such as expression of a marker (e.g. indicator or reporter) gene.
  • a marker e.g. indicator or reporter
  • An indicator/marker reporter gene includes an epitope, (e.g., an antigen TAG or an endogenous protein or epitope-containing fragment thereof), an enzyme (such as one that generates a novel or chromogenic product), a selectable marker, or a fluorescent molecule, such as green fluorescent protein (GFP), blue fluorescent protein (BFP), yellow fluorescent protein (YFP), red fluorescent protein (RFP), luciferase, ⁇ -galactosidase, and CAT, and others disclosed herein or known in the art.
  • GFP green fluorescent protein
  • BFP blue fluorescent protein
  • YFP yellow fluorescent protein
  • RFP red fluorescent protein
  • epitops e.g., antigen TAGs or endogenous proteins
  • preferred embodiments utilize cell surface antigens.
  • the epitope is preferably any detectable peptide which is not generally found on the cytoplasmic membrane, although in some instances, if the epitope is one normally found on the cells, increases may be detected, although this is generally not preferred.
  • the epitope may be intracellular (e.g. cytoplasmic) and may still be detectable by methods known in the art and /or disclosed herein (see, e.g., Example 29).
  • An altered phenotype may also be detected by FACS as described herein, by optical properties, such as inherent optical properties, or those resulting from dyes, etc., and other detection methods, such as binding of an antibody, ligand, etc., as described herein.
  • Such an antibody, ligand, etc. may be labeled or may be conjugated to a toxin.
  • Many other methods for detecting an altered phenotype are well known in the art, and may also be described herein.
  • the antibody is labeled with a fluorescent marker such as FITC and is detected by FACS. Cells may thus be sorted by FACS to speed selection of larger polynucleotides.
  • a target polynucleotide may alter a phenotype of a cell comprising said target polynucleotide.
  • a target polynucleotide may alter a phenotype of a second cell (i.e. an indicator cell) as described elsewhere herein.
  • the phenotype of a second cell it may be preferable to divide the host cells into pools which also comprise the indicator cells. Then insert polynucleotides are collected from those pools in which an altered phenotype of an indicator cell is detected.
  • the selection methods include one or a combination of the modifications, uses, and other embodiments described herein and also provide for the exclusion of any one or more modifications, uses and other embodiments.
  • the invention provides a method for selection based on immunological methods which include antibody-based methods, but which exclude CD8+ CTL-based methods.
  • the method may include selection of DNA inserts in dividing cells, but exclude selection in tumor cells.
  • the selection method may include mechanisms based on nonadherence, but exclude nonadherence due to lysis.
  • the selection method may include direct selection, but exclude CTL-mediated selection.
  • the plurality of cells comprise a library.
  • the "vector comprising insert polynucleotides” is preferably a plurality of insert polynucleotides which represent a library, and which is cloned in a vector.
  • the plurality of cells may contain a single type of insert polynucleotide cloned in a vector, i.e., the plurality of cells represents a single "clone.”
  • at least one of the insert polynucleotides is a target polynucleotide, i.e., a polynucleotide of interest, which is selectable by at least one direct selection method of the invention.
  • insert polynucleotides may also be referred to herein as “candidate polynucleotides” or other grammatical equivalents.
  • the library may be any type of library such as cDNA, genomic DNA, combinatorial, etc., as described elsewhere herein or otherwise known in the art.
  • the insert polynucleotides may be natural or synthetic or a mixture of natural and synthetic products. They may be generated, for example, in vivo or in vitro, enzymatically, or chemically.
  • the insert polynucleotides may be from any source such as a cell line, a biological sample, a patient sample, or from any of the cells described below as examples of host cells, etc.
  • the insert polynucleotides are cloned in vaccinia viras.
  • the insert polynucleotide is one or a plurality of products of tri-molecular recombination, including progeny thereof, preferably infectious progeny.
  • the methods herein are applicable to a wide variety of expression libraries including mixtures of cDNAs cloned in sense or anti-sense orientation, mixtures of fragments of genomic DNA or cDNA or both, fragments of individual cDNAs and mixtures of artificial sequences.
  • the insert polynucleotide e.g. DNA
  • the insert polynucleotides are operably associated with strong constitutive expression control sequences, preferably strong constitutive poxviras promoters such as the 7.5K vaccinia viras promoter or a strong synthetic early/late (E/L) promoter.
  • host cell or "recipient cell” or “library cell” is meant a cell or plurality of cells into which DNA is introduced for the tri-molecular recombination method or a direct or indirect selection method described below.
  • the host cells adhere to a solid support.
  • normally nonadherent cells and cell types may be made to adhere to a solid support by use of a molecule such as a protein, an antibody, or poly L-lysine on the surface of the solid support. Other means for attaching cells to a solid support are well known in the art.
  • the host cells may also comprise packaging function.
  • Host cells are eukaryotic cells or cell lines, preferably animal, vertebrate, mammalian, or human cells or cell lines.
  • Host cells may be cells derived from primary tumors, cells derived from metastatic tumors, primary cells, cells which have lost contact inhibition, transformed primary cells, immortalized primary cells, cells which may undergo apoptosis, and cell lines derived therefrom.
  • Cells and cells lines for use as host or recipient or library cells according to the present invention include those disclosed in scientific literature such as American Type Culture Collection publications including American Type Culture Collection Catalogue of Cell Lines and Hybridomas, 7th ed., ATCC, Rockville, MD (1992) and subsequent editions, which list deposited cell lines as well as culture conditions and additional references.
  • host cells include the monkey kidney cell line, designated "COS,” including COS cell clone M6.
  • COS cells are those that have been fransformed by SV40 DNA containing a functional early gene region but a defective origin of viral DNA replication.
  • murine "WOP" cells which are NIH 3T3 cells transfected with polyoma origin deletion DNA.
  • host cells for use in the disclosed methods are monkey kidney CVI line transformed by SV40 (COS-7, ATCC CRL 165 1); human embryonic kidney line (293, Graham et al. J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary- cells-DHFR (CHO, Urlaub and Chasin, Proc. Natl. Acad. Sci. (USA) 77:4216, (1980); mouse sertoli cells (TM4, Mather, Biol. Reprod.
  • monkey kidney cells CVI ATCC CCL 70); african green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3 A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL 51); TRI cells (Mather et al, Annals N. Y. Acad.
  • Suitable cell types for use in the invention will vary with the cellular phenotype to be modulated. Suitable cells include, but are not limited to, mammalian cells, including animal (rodents, including mice, rats, hamsters and gerbils), primates, and human cells, particularly including tumor cells of all types, including breast, skin, lung, cervix, colorectal, leukemia, brain, etc.
  • the choice of cells or cell lines is not limited to those described herein, and may be any cell or cell line. As indicated below, the choice depends on the system under study, or the particular polynucleotide which is desired to be isolated. For example, to select or isolate an epitope recognized by a human CD8 + CTL, it is preferable to use a host cell which expresses human class I MHC molecules, and to select or isolate an epitope recognized by a human CD4 CTL, it is preferable to use a host cell which expresses or can be induced to express human class II MHC molecules, to allow the CTL to recognize the encoded epitope in association with the appropriate MHC molecules.
  • breast cancer cell lines such as 21NT, 21PT, 21MT-1, AND 21MT-2. Band et al, Cancer Res. 50:1351-1 (1990).
  • a growth suppressive polynucleotide Once a growth suppressive polynucleotide is isolated, it may be tested in non fransformed controls, such as normal breast epithelial cell fine H16N2, to determine whether its growth suppressive activity is specific for tumor cells.
  • Cells include dividing cells, non dividing cells, terminally differentiated cells, pluripotent stem cells, committed progenitor cells and uncommitted stem cells.
  • Cells and cell types also include muscle cells such as cardiac muscle cells, skeletal muscle cells and smooth muscle cells; epithelial cells such as squamous epithelial cells, including endothelial cells, cuboid epithelial cells and columnar epithelial cells; nervous tissue cells such as neurons and neuroglia. Also included are progenitor cells of each of these cells.
  • Cells that can be used in the selection method of the present invention also include nervous system cells such as neurons, including cortical neurons, inter neurons, central effector neurons, peripheral effector neurons and bipolar neurons; and neuroglia, including Schwann cells, oligodendrocytes, astrocytes, microglia and ependyma. Also included are progemtor cells of each of these cells.
  • nervous system cells such as neurons, including cortical neurons, inter neurons, central effector neurons, peripheral effector neurons and bipolar neurons
  • neuroglia including Schwann cells, oligodendrocytes, astrocytes, microglia and ependyma.
  • progemtor cells of each of these cells are also included.
  • endocrine and endocrine-associated cells may also be used such cells as pituitary gland cells including epithelial cells, pituicytes, neuroglia, agranular chromophobes, granular chromophils (acidophils and basopbils); adrenal gland cells including epinephrine-secreting cells, non- epinephrine-secreting cells, medullary cells, cortical cells (cells of the glomerulosa, fasciculata and reticularis); thyroid gland cells including epithelial cells (principal and parafollicular); parathyroid gland cells including epithelial cells (chief cells and oxyphils); pancreas cells including cells of the islets of Langerhans (alpha, beta and delta cells); pineal gland cells including parenchymal cells and neuroglial cells; thymus cells including parafollulicular cells; cells of the testes including seminiferous tubule cells, interstitial
  • muscle cells such as myofibrils, intrafusal fibers and extrafusal fibers; skeletal system cells such as osteoblasts, osteocytes, osteoclasts and their progenitor cells. Also included are progenitor cells of each of these cells.
  • Circulatory system cells are also included such as heart cells
  • myocardial cells cells of the blood and lymph including erythropoietin- sensitive stem cells, erythrocytes, leukocytes (such as eosinophils, basophils and neutrophils (granular cells) and lymphocytes and monocytes (agranular cells)), thrombocytes, tissue macrophages (histiocytes), organ-specific phagocytes (such as Kupffer cells, alveolar macrophages and microglia), B- lymphocytes, T-lymphocytes (such as cytotoxic T cells, helper T cells and suppressor T cells), megaloblasts, monoblasts, myeloblasts, lymphoblasts, proerythroblasts, megakaryoblasts, promonocytes, promyelocytes, prolymphocytes, early normoblasts, megakaryocytes, intermediate normoblasts, metamyelocytes (such as juvenile metamyelocytes, segmented metamyelocytes and polymorphonucle
  • Respiratory system cells are also included such as capillary endothelial cells and alveolar cells; as are urinary system cells such as nephrons, capillary endothelial cells, granular cells, tubule endothelial cells and podocytes; digestive system such as simple columnar epithelial cells, mucosal cells, acinar cells, parietal cells, chief cells, zymogen cells, peptic cells, enterochromaffin cells, goblet cells, Argentaffen cells and G cells; and sensory cells such as auditory system cells (hair cells); olfactory system cells such as olfactory receptor cells and columnar epithelial cells; equihbrium vestibular apparatus cells including hair cells and supporting cells; visual system cells including pigment cells, epithelial cells, photoreceptor neurons (rods and cones), ganglion cells, amacrine cells, bipolar cells and horizontal cells are also included. Also included are progenitor cells of each of these cells.
  • mesenchymal cells stromal cells, fibroblasts, hair cells/follicles, adipose (fat) cells
  • cells of simple epithelial tissues squamous epithelium, cuboidal epithelium, columnar epithelium, ciliated columnar epithelium and pseudostratified ciliated columnar epithelium
  • cells of stratified epithelial tissues stratified squamous epithelium (keratinized and non-keratinized), stratified cuboidal epithelium and transitional epithelium
  • goblet cells endothelial cells of the mesentery, endothelial cells of the small intestine, endothelial cells of the large intestine, endothelial cells of the vasculature capillaries, endothelial cells of the microvasculature, endothelial cells of the arteries, endothelial cells of the arterioles, endothelial cells of
  • progenitor cells of each of these cells are also included.
  • Cells useful in the inventions herein also include progenitor cells of all the cells above and elsewhere herein.
  • a “plurality” or “population” or “library” of cells is meant at least two cells, with at least about 10 3 , 10 4 ,10 5 ,10 6 ,10 7 ,10 8 , 10 9 being particularly preferred.
  • the plurality or population can contain a mixture of different cell types from either primary or secondary cultures although samples containing only a single cell type are preferred, for example, the population can be from a cell line, including tumor cell lines.
  • the cells may be in any cell phase, either synchronous or not, including M, GI, S, and G2.
  • solid support any support capable of binding a cell, which may be in any of various forms, as is known in the art.
  • Well-known supports include tissue culture plastic, glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite.
  • the nature of the carrier can be either soluble to some extent or insoluble for the purposes of the present invention.
  • the support material may have virtually any possible structural configuration as long " as the coupled molecule is capable of binding to a cell.
  • the support configuration may be spherical, as in a bead, or cylindrical, as in the inside surface of a test tube, or the external surface of a rod.
  • the surface may be flat such as a sheet, test strip, etc.
  • Preferred supports include polystyrene beads.
  • the support configuration may include a tube, bead, microbead, well, plate, tissue culture plate, petri plate, microplate, microtiter plate, flask, stick, strip, vial, paddle, etc., etc.
  • a sohd support may be magnetic or non-magnetic. Those skilled in the art will know many other suitable carriers for binding cells, or will be able to readily ascertain the same.
  • Cells may be directly or indirectly bound to a sohd support, as is well known in the art.
  • cells may be indirectly attached via one or more molecules such as protein, antibody, receptor, ligand,. poly L-lysine, or may be indirectly attached via other cells.
  • cells are attached to a solid support via antibody.
  • the cells are attached to a magnetic solid support, such as magnetic beads, via antibody.
  • tissue culture or "cell culture” or “culture” or “culturing” is meant the maintenance or growth of animal tissue or cells in vitro under conditions that allow preservation of cell architecture, preservation of cell function, further differentiation, or all three, as well as permitting expression of insert polynucleotides.
  • Primary tissue cells are those taken directly from tissue, i.e., a population of cells of the same kind performing the same function in an organism. Treating such tissue cells with the proteolytic enzyme trypsin, for example, dissociates them into individual primary tissue cells that grow or maintain cell architecture when seeded onto culture plates. Cell cultures arising from multiplication of primary cells in tissue culture are called “secondary cell cultures.” Most secondary cells divide a finite number of times and then die. A few secondary cells, however, may pass through this "crisis period,” after which they are able to multiply indefinitely to form a continuous "cell line.”
  • a recipient cell containing a target polynucleotide may become
  • nonadherent or “nonviable” by any mechanism, which may include lysis, inability to adhere, loss of viability, loss of membrane integrity, loss of structural stability, disruption of cytoskeletal elements, inability to maintain membrane potential, arrest of cell cycle, inability to generate energy, etc.
  • cells containing target polynucleotides may be collected, i.e., recovered or separated from remaining cells, by any physical means such as aspiration, washing, filtration, centrifugation, cell sorting, fluorescence activated cell sorting (FACS), etc.
  • Collected cells or polynucleotides may also be referred to herein as “recovered” cells or polynucleotides, or as “isolated” cells or polynucleotides.
  • cells containing target polynucleotides may lyse and thereby release recombinant vaccinia into the culture media or may become nonadherent and therefore lift away from the solid support.
  • released recombinant vaccinia and/or nonadherent cells are separated from adherent cells by aspiration or washing of the liquid medium, e.g. culture medium.
  • a recipient cell containing a target polynucleotide may become "adherent" or "viable” by any mechanism.
  • host cells which do not contain a target polynucleotide undergo lysis, are unable to adhere, are not viable in culture, lose membrane integrity, lose stractural stability, become disrupted in their cytoskeletal elements, are unable to maintain membrane potential, are arrested in cell cycle, are unable to generate energy, etc.
  • Cells containing a target polynucleotide fail to lyse, do adhere, remain viable, retain or gain membrane integrity, retain or gain structural stability, retain or gain cytoskeletal elements, maintain or gain membrane potential, are not growth cycle arrested, or generate energy, etc.
  • cells not containing target polynucleotides, or their contents may be removed from those containing target polynucleotides by any physical means of separation such as aspiration, washing, filtration, centrifugation, cell sorting, fluorescence activated cell sorting (FACS), etc.
  • FACS fluorescence activated cell sorting
  • cells which do not contain a target polynucleotide may be removed from the cells which do contain a target polynucleotide by washing or aspiration. The remaining cells containing target polynucleotides are then collected.
  • the methods of the invention are useful to enrich for target polynucleotides from a plurahty of test polynucleotides, such as a complex library including cDNA and other libraries.
  • a single round of direct or indirect selection may not necessarily result in isolation of a pure set of target polynucleotides; the mixture obtained after a first round may be enriched for the desired polynucleotides but may also be contaminated with non-target insert sequences.
  • selection of cytotoxic and cytostatic target polynucleotides may require or benefit by several rounds of selection, which thus increase the proportion of cells containing target polynucleotides.
  • the material obtained after the first round is used to infect a second population of cells and the resulting sublibrary is subjected to a second round of selection.
  • the invention provides a method of producing a subpopulation of target polynucleotides comprising the direct or indirect selection method, and further comprising: isolating target polynucleotides from the selected (e.g. nonviable) cells, introducing the target polynucleotides into a second plurality of host cells, and separating the selected cells (e.g. non-viable or contents thereof).
  • the method comprises additional rounds of infection of new host cells and direct or indirect selection.
  • the method provides multiple rounds of enrichment to produce subpopulations, or sublibraries, which comprise a progressively decreasing ratio of nontarget polynucleotides, i.e., decreased background.
  • those polynucleotides which have been recovered are "isolated," i.e., they are substantially removed from their native environment and are largely separated from polynucleotides in the library which do not encode antigen-specific immunoglobulin subunit polypeptides.
  • isolated polynucleotides i.e. insert polynucleotides contained in a vector are considered isolated for the purposes of the present invention.
  • a mixture of target polynucleotides is also considered to be “isolated.”
  • isolated polynucleotides include those maintained in heterologous host cells or purified (partially or substantially) DNA molecules in solution.
  • a polynucleotide contained in a virus vector is "isolated” after it has been recovered and plaque purified, and a polynucleotide contained in a plasmid vector is isolated after it has been expanded from a single bacterial colony.
  • a polynucleotide contained in a clone that is a member of a mixed library and that has not been isolated from other clones of the library is not “isolated” for the purposes of this invention.
  • a phenotype may be effected by more than one polynucleotide
  • several suitable polynucleotides e.g., two, three, four, five, ten, 100 or more polynucleotides, may be recovered from the first step of this embodiment, each of which may be a target a polynucleotide. It is contemplated that each different polynucleotide recovered from the library may be separately isolated. However, these polynucleotides maybe isolated as a group of polynucleotides which effect the same or similar phenotypes and these polynucleotides may be "isolated" together.
  • target polynucleotides may be purified (partially or substantially) from protein such as viral and or cellular proteins and other components, using well-known techniques such as those described in the examples (see, e.g., Example 2).
  • the present invention provides an alternative method of identification of polynucleotides (e.g. gene sequences) whose expression elicits growth suppression, apoptosis or any other phenotype selectable by the present invention. It is based on the isolation from an expression library of one or more cells expressing a target polynucleotide, such that expression of the target polynucleotide causes cell lysis or otherwise compromises cell integrity or cell viability.
  • the target polynucleotide may be toxic or may sensitize the cell to an agent.
  • the target polynucleotide may inhibit the ability of a cell to remain adherent, such as due to a cytoskeletal effect or an effect on ATP production.
  • the direct or indirect selection method exploits various cell-disrupting mechanisms to isolate polynucleotides of interest from a library.
  • Cell- disrupting mechanisms which may be used include the following: immune system-mediated disruption such as by CTLs, antibody-dependent cellular cytotoxicity (ADCC), and complement-dependent cytotoxicity (CDC); disruption by toxic sequences such as apoptosis-inducing polynucleotides, cell cycle arresters, tumor suppressor genes, dominant negative oncogene variants, cell proliferation inhibitors, and CDK inhibitors; negative regulation of essential genes by antisense expression; and induction of suicide gene expression.
  • immune system-mediated disruption such as by CTLs, antibody-dependent cellular cytotoxicity (ADCC), and complement-dependent cytotoxicity (CDC)
  • disruption by toxic sequences such as apoptosis-inducing polynucleotides, cell cycle arresters, tumor suppressor genes, dominant negative oncogene variants, cell proliferation inhibitors, and CDK inhibitors
  • This method has many applications such as isolation of growth suppressive molecules (DNA, RNA, polypeptides, or peptides) with specificity against certain cell types, and identification of new genes involved in negative growth regulation (i.e., tumor suppressive genes, genes involved in confrol of apoptosis, cellular senescence, sensitivity to chemical, physical or biological treatments, etc.), drag susceptibility, cell cycle regulation, cell differentiation, or transformation.
  • growth suppressive molecules DNA, RNA, polypeptides, or peptides
  • negative growth regulation i.e., tumor suppressive genes, genes involved in confrol of apoptosis, cellular senescence, sensitivity to chemical, physical or biological treatments, etc.
  • drag susceptibility i.e., cell cycle regulation, cell differentiation, or transformation.
  • examples of target polynucleotides are those that decrease growth rate or kill cells under normal conditions of growth or under specific conditions, including sensitization of cells to certain physical, chemical or biological treatments that have no or low effect on the majority of cells in, for example, a library-carrying cell population.
  • an indirect selection method may be used, wherein the host cells lyse or otherwise are compromised in cell integrity or cell viability, and wherein target polynucleotides are selected which allow such cells to remain intact.
  • Target polynucleotides may be cell proliferation genes which, when aberrantly expressed or regulated, may induce or otherwise be involved in the development of cell proliferative disorders.
  • cell proliferative disorders include, but are not limited to cancers, arteriosclerosis, psoriasis, viral disease, as well as inflammatory conditions such as arthritis or sepsis.
  • Cell proliferation genes include dominant transforming genes, such as oncogenes and other genes encoding products involved in the induction of cell growth and recessive cell proliferation genes, such as genes encoding tumor suppressors, genes involved in the induction of apoptosis or genes involved in viral growth.
  • Target polynucleotides include those that are growth suppressive for certain cell types (for example, tumor cells, viras infected cells, cells of certain tissue, etc.) but that have no or low effect on other cell types, such as the parent cell type. Isolation of clones that are specifically toxic for certain cell type may require additional selection of the isolated mixture of clones in the cell type of different origin in order to define those clones that have selective biological activity against the cells of choice.
  • cytotoxic and cytostatic target polynucleotides may require or benefit by several rounds of selection, which thus increase the proportion of cells containing target polynucleotides.
  • Isolation of target polynucleotides that are not cytotoxic or cytostatic by themselves, but which display growth suppressive activity under certain conditions of cell maintenance or treatment requires application of the appropriate selection.
  • selection is preferably noncytotoxic or low-cytotoxic by itself and should not lead to significant cell death or growth arrest in host cells which do not contain a target polynucleotide.
  • polynucleotides restoring the activity of the p53 pathway by blocking p53 inhibitors are expected to cause cell sensitization to DNA-damaging treatments since p53 plays a role of negative growth regulator that is activated under conditions of genotoxic stress.
  • the direct or indirect selection method is not limited to the isolation of cytotoxic or cytostatic elements. It can be used for isolation of genetic elements that induce any cell phenotype (i.e. expression of cell surface antigens, alterations in cell adhesion, cell size, etc.) that can be used as a selective trait to enrich or to exhaust a cell population.
  • the methods disclosed herein are used to select for modulators of cellular phenotypes.
  • Cellular phenotypes that may be selected for include, but are not limited to, cellular apoptosis, including cell cycle regulation, toxicity to small molecules, the expression of any number of moieties including receptors (particularly cell surface receptors), adhesion molecules, cytokine secretion, protein-protein interactions, transcriptional activation of particular promoters, etc.
  • FACS fluorescence-activated cell sorting
  • FACS Fluorescence activated cell sorting
  • flow cytometry is used to sort individual cells on the basis of optical properties, including fluorescence. It is useful for screening large populations of cells in a relatively short period of time. Rapid and inexpensive screens or selections such as by FACS would be of particular interest for identifying drug candidates such as modulators of cell cycle regulation.
  • cells containing target polynucleotides may be collected, for example, by magnetic beads. Such methods are described in more detail herein and are also well known in the art.
  • Target polynucleotides may modulate cell cycle regulation, by, for example, suppressing or activating a cell cycle checkpoint pathway, or ameliorating or inducing checkpoint defects.
  • host cells are sorted in a FACS machine by assaying cell parameters, including, but not limited to, cell viability, cell proliferation, and cell phase.
  • preferred cellular parameters or assays are cell viability assays, assays to determine whether cells are arrested at a particular cell cycle stage ("cell proliferation assays"), and assays to determine at which cell stage the cells have arrested (“cell phase assays").
  • the methods are used to evaluate cell cycle regulation.
  • Cells cycle through various stages of growth, starting with the M phase, where mitosis and cytoplasmic division (cytokinesis) occurs.
  • the M phase is followed by the GI phase, in which the cells resume a high rate of biosynthesis and growth.
  • the S phase begins with DNA synthesis, and ends when the DNA content of the nucleus has doubled.
  • the cell then enters G2 ' phase, which ends when mitosis starts, signaled by the appearance of condensed chromosomes. Terminally differentiated cells are arrested in the GI phase, and no longer undergo cell division.
  • preferred cellular parameters or assays are cell viability assays, assays to determine whether cells are arrested at a particular cell cycle stage ("cell proliferation assays”), and assays to determine at which cell stage the cells have arrested (“cell phase assays”).
  • cell proliferation assays assays to determine whether cells are arrested at a particular cell cycle stage
  • cell phase assays assays to determine at which cell stage the cells have arrested
  • an MVA vaccinia viras vector or other attenuated virus is used.
  • the methods outlined herein are performed on cells that are not arrested in the GI phase; that is, they are rapidly or uncontrollably growing and replicating, such as tumor cells.
  • insert polynucleotides are evaluated to target polynucleotides that alter cell cycle regulation, i.e. cause cells to arrest at cell cycle checkpoints, such as GI, although arresting in other phases such as S, G2 or M are also desirable.
  • insert polynucleotides are evaluated to find those that cause proliferation of a population of cells, i.e. that allow cells that are generally arrested in GI to start proliferating again; for example, peripheral blood cells, terminally differentiated cells, stem cells in culture, etc.
  • the direct or indirect selection method is useful for isolating the following polynucleotides: polynucleotides encoding epitopes and antigens; any toxic sequence, for example, apoptosis-inducing genes, genes that arrest cell cycle, tumor suppressor genes, dominant negative oncogene variants, inhibitors of cell proliferation, and CDK inhibitors; essential genes; and genes that induce expression ofa suicide gene constract (i.e., a suicide gene operably associated with a regulated promoter).
  • any toxic sequence for example, apoptosis-inducing genes, genes that arrest cell cycle, tumor suppressor genes, dominant negative oncogene variants, inhibitors of cell proliferation, and CDK inhibitors
  • essential genes and genes that induce expression of a suicide gene constract (i.e., a suicide gene operably associated with a regulated promoter).
  • genes that induce expression of a suicide gene include those that promote cell cycle progression and thereby induce a cell cycle specific suicide gene constract (i.e., operably associated with a cell-cycle specific promoter), those that promote differentiation and thereby induce a spatially or temporally regulated suicide gene constract (i.e., operably associated with a spatially-regulated or temporally-regulated promoter) or a tissue or cell specific suicide gene constract (i.e., operably associated with a tissue- or cell-specific promoter) (collectively, differentiation-specific promoters), those that interact with another protein in a two-hybrid transcription system and thereby induce expression of a suicide gene.
  • a cell cycle specific suicide gene constract i.e., operably associated with a cell-cycle specific promoter
  • those that promote differentiation and thereby induce a spatially or temporally regulated suicide gene constract i.e., operably associated with a spatially-regulated or temporally-regulated promoter
  • the invention provides a method of directly selecting a target polynucleotide comprising: (a) providing a plurality of cells which comprise
  • packaging function (i) transfer plasmid comprising a 5' flanking region, an insert polynucleotide, and a 3' flanking region, (iii) a first viras fragment comprising a sequence homologous to the 5' flanking region, and (iv) a second virus fragment comprising a sequence homologous to the 3' flanking region;
  • the method comprises contacting a plurality of cells with infectious progeny produced by tri-molecular recombination, culturing said cells under conditions such that the insert polynucleotides are expressed, and removing a nonviable cell expressing the target.
  • antigen-specific cytotoxic mechanisms such as cell, antibody, and complement- mediated cytotoxicity, may be used to isolate host cells expressing a protein of interest.
  • cytotoxic T cells CTL
  • antibody-toxin conjugates CTL
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-dependent cytotoxicity
  • the immune-based direct selection method may be used to identify target antigens in tumor cells, target antigens in other cells against which it is desirable to induce cell-mediated or humoral immunity, or any target epitope which is capable of inducing specific CTL or antibody.
  • the differential immunogenicity methods of the invention can be applied to identify immunogenic molecules of cells infected with viras, fungus or mycobacteria by tolerization of mice with uninfected cells followed by immunization with infected cells at different times after infection.
  • Isolated CTLs or antibodies can be employed to select polynucleotides (e.g. recombinants) that encode target antigens in a plasmid or viral expression library.
  • an expression library can be constructed with cDNA isolated from the infected cell in a vaccinia virus vector using tri-molecular recombination.
  • cDNA isolated from the infected cell in a vaccinia virus vector using tri-molecular recombination.
  • One use of this approach is to identify pathogen-encoded antigens and host cell antigens whose expression is altered during infection. Such antigens may be useful as a vaccine against infection by the pathogen.
  • Pathogens include all pathogenic agents known in the art. Pathogens include, but are not limited to: viral pathogens, such as human immunodeficiency viras (HIV), Epstein Barr viras, hepatitis viras, herpes virus, human papillomaviras, cytomegalovirus, respiratory syncytial virus; fungal pathogens, such as Candida albicans, Pneumocystis carnii; and mycobacterial pathogens, such as M. tuberculosis, M. avium.
  • viral pathogens such as human immunodeficiency viras (HIV), Epstein Barr viras, hepatitis viras, herpes virus, human papillomaviras, cytomegalovirus, respiratory syncytial virus
  • fungal pathogens such as Candida albicans, Pneumocystis carnii
  • mycobacterial pathogens such as M
  • Pathogens also include the bacteria Pseudomonas aeraginosa,
  • Mycobacterium tuberculosis Hemophilus influenzae, Staphylococcus aureus, Mycoplasma pneumoniae, Escherichia coli, Streptococcus pneumoniae, Neisseria gonorrhaea, Streptococcus viridans, Streptococcus pyogenes, Proteus mirabilis, Proteus vulgaris, Salmonella typhimurium, Shigella dysentereae, Clostridium difficile, and Klebsiella pneumoniae, and the fungi Candida albicans, Aspergillus flavus, Aspergillus fumagatus, and Histoplasmatus capsulatum.
  • Pathogens also include those listed elsewhere herein and otherwise known in the art.
  • Cytotoxic T Lymphocytes In this version of immune-system based selection, epitope-specific CTLs are used to select target polynucleotides expressing an epitope of interest.
  • target cells must be highly transfectable in order to take up cDNA for screening. Only two commonly employed cell lines have the requisite properties, Cos 7 and 293. However, since a fundamental property of T cells is that they are restricted to recognize peptide epitopes presented in association with MHC molecules, these African Green Monkey cells do not express the appropriate human MHC molecules. Although it is possible to co-transfect MHC genes, it is often unclear which of the six possible MHC class I or MHC class II alleles in a heterozygous individual are required for presentation of the target peptide epitope.
  • cDNA libraries in an infectious vector with a very broad host range, it is possible to employ as the target cell autologous fibroblasts, epithelial cells or EBV transformed B cells that naturally express the full complement of relevant MHC molecules.
  • alternatives to screening cDNA libraries such as the use of tandem HPLC/mass specfrometry to screen peptides eluted from tumor cells, have the limitation that very large numbers of tumor cells are required for peptide purification, quantities which are not normally available from patient samples.
  • human tumor-specific T cells are isolated by stimulation with either autologous tumor or autologous antigen presenting cells pulsed with tumor cell lysates and these T cells are employed to screen expression libraries generated from tumor cell DNA, RNA or cDNA to identify reactive target antigens.
  • tumor-specific or infection-specific CTLs generated in animals are used to screen expression libraries generated from tumor cell or infected cell DNA, RNA or cDNA to identify reactive target antigens.
  • animals tolerized with a non-tumorigenic or non-infected human cell line are immunized with tumor cells or infected cells derived from the non-tumorigenic or non-infected cell line.
  • tumor-specific or infection-specific CTLs generated in vitro are used to screen expression libraries.
  • the resulting CTLs which are tumor-specific or infection-specific and not cross-reactive with normal cells, can be used to screen expression libraries constructed from tumor cell- or infected cell-derived DNA, RNA or cDNA.
  • Clones so identified in the hbrary encode target antigens which are candidates for the immunogenic compositions and vaccines of the invention.
  • Improved and modified vaccinia virus vectors for efficient constraction of such DNA libraries using a "trimolecular recombination" approach are described to improve screening efficiency.
  • Animals such as normal or fransgenic mice, may be tolerized with normal cells or lysates thereof prior to immunizing with tumor cells or cells infected with a pathogen, or a lysate thereof.
  • Tolerance induction is preferred because the animal's immune response would otherwise be dominated by specificity for a large number of broadly expressed human proteins that are not specifically associated with tumor transformation or infection.
  • it is convenient to work with human tumors that are derived from an immortalized, non-tumorigenic human cell line by in vitro carcinogenesis or oncogene transformation. This provides a ready source of the normal control cells for an extended tolerization protocol in both neonatal and adult mice.
  • CTLs generated by this approach can be employed in a selection procedure (such as that described in Examples 3-4) to isolate recombinant clones (i.e. target polynucleotides) that encode the target antigens from a tumor cDNA library, for example, such as that constructed in vaccina viras by tri-molecular recombination (see Example 2).
  • recombinant clones i.e. target polynucleotides
  • a tumor cDNA library for example, such as that constructed in vaccina viras by tri-molecular recombination (see Example 2).
  • Antibody-based Selection In other versions of immune-based selection, the host cells are exposed to an antibody or plurality of antibodies directed against an epitope of interest. This results in formation of an antigen- antibody complex at the cell surface. Alternatively, the complexes are contacted with a second antibody or plurality of antibodies directed against the first antibody. The second antibody may be conjugated with a toxin, or alternatively, the first antibody is conjugated with a toxin.
  • complement-dependent cytotoxicity (CDC) or antibody- dependent cellular cytotocity (ADCC) is used to select target polynucleotides. See U.S. Pat. No. 5,500,362 for ADCC and CDC assays.
  • Such assays may be modified for use in the present selection method by, for example, omitting the 51 Cr labeling of cells, as will be apparent to one of ordinary skill in the art. See, e.g. Hellstrom et al, Proc. Natl Acad. Sci. USA 52:1499-1502 (1985).
  • the complement or cell mediators of ADCC may be added to the antigen- antibody complexes.
  • the antibody and cell mediators of ADCC may be combined prior to contacting the host cells.
  • Cells expressing the antigen are thus induced to undergo lysis or other mechanisms leading to nonadherence.
  • Nonviable cells, and the cell contents of lysed cells are separated from viable cells.
  • the antibody is labeled and the cells are separated by FACS.
  • the cells adhere to a magnetic solid support, such as a magnetic bead, via antibody on the surface of the solid support and are separated from nonadherent cells by the use of a magnet.
  • antibody As used herein, the term “antibody” (Ab) or “monoclonal antibody”
  • Mab is meant to include intact molecules as well as antibody fragments (such as, for example, Fab and F(ab') 2 fragments) which are capable of specifically binding to the protein encoded by a target polynucleotide. If ADCC- or complement-mediated cell killing is utilized in the direct or indirect selection method, the antibodies preferably include an Fc portion.
  • Mouse IgG 2a and IgG 3 and human IgGi and IgG 3 isotypes are most commonly associated with antibody-dependent cellular cytotoxicity and complement- dependent cytotoxicity.
  • antibodies can be, for example, polyclonal or monoclonal antibodies.
  • the present invention also includes chimeric, single chain and humanized antibodies, as well as Fab fragments, or the product of an Fab expression library. Various procedures known in the art can be used for the production of such antibodies and fragments.
  • the antibodies used according to the present invention may be directed against the antigen of interest or against a family of related antigens. A single antibody or a group of antibodies may be used, the choice depending upon the nature of the target antigen, its anticipated frequency, and other variables that will be apparent to those of skill. Target antigens expressed on the surfaces of host cells will form an antigen-antibody complex. Methods of preparing antibodies or fragments thereof for use according to the present invention are known to those of skill and/or disclosed in the scientific literature listed below.
  • Receptors and Ligands may also be used to directly select for the cognate receptor of a known ligand, hapten, or epitope, for example, by contacting host cells containing an expression library with a labeled ligand or a ligand conjugated to a toxin.
  • the known ligand may be soluble or may be associated with membrane bilayers or a solid support such as beads, etc.
  • the known ligand may be another receptor, or fragment thereof, which binds the receptor encoded by the target polynucleotide.
  • the ligand of a known receptor is identified. For example, insert polynucleotide (e.g.
  • DNA is cloned in-frame with a polynucleotide (e.g. DNA) sequence encoding a receptor transmembrane domain.
  • the transmembrane domain may be naturally occurring, or nonnaturally occurring.
  • the transmembrane domain directs the polypeptide (candidate ligand) encoded by the insert polynucleotide (e.g. DNA) to be expressed on the cell surface. See U.S. 5,866,341.
  • the cells are contacted with receptor polypeptide or a fragment thereof.
  • the receptor or receptor fragment may be labeled or may be conjugated to a toxin.
  • the receptor or receptor fragment may be soluble, hydrophobic, associated with membranes, associated with membrane vesicles, associated with lipid bilayers, associated with lipid bilayer vesicles, associated with a sohd support such as beads, etc.
  • Labels for ligands, receptors, etc. include those in Example 14 for antibodies.
  • the host cells are contacted with an antibody which recognizes the receptor or ligand encoded by the target polynucleotide.
  • the host cells are contacted with known ligand, and the antibody recognizes the ligand, the receptor, or the ligand-receptor complex.
  • receptors means any protein capable of specifically binding a ligand, hapten, or epitope.
  • receptors that may identified by binding known ligand, epitopes, or haptens, include adhesion molecules such as cadherins, selectins, fasciclins, integrins, leukocyte adhesion receptor, neuroglian, VLA family molecules and the like.
  • adhesion molecules such as cadherins, selectins, fasciclins, integrins, leukocyte adhesion receptor, neuroglian, VLA family molecules and the like.
  • receptors that may identified by binding to known growth factors include growth hormone receptors, insulin receptors, interleukin receptors and the like.
  • receptors that may identified by binding to known ligands include chemokine receptors and G-protein coupled receptors.
  • Nondividing Cells A major technical obstacle to identifying candidate pharmaceutical targets is the nondividing state of terminally differentiated such as musculoskeletal or other cells. Because vectors currently used for hbrary production do not replicate in nondividing cells, it is not possible to recover a gene of interest in nondividing cells by conventional methods. Moreover, it is difficult to identify an expressed gene in a hbrary based on its function using some current cloning vectors because of the low expression level of cloned genes. Although vaccinia-based vectors replicate in nondividing cells and express polypeptides encoded by insert polynucleotides (e.g. DNA) at a high level, the very low cloning efficiency of vaccinia using homologous recombination and the low titers obtained through direct ligation have precluded the use of vaccinia virus for hbrary production.
  • polynucleotides e.g. DNA
  • trimolecular recombination method of the present invention overcomes the obstacle of low cloning efficiency or low titer in vaccinia virus. As described herein, recombination efficiencies of 90% to 100% and relatively high titers have been achieved using trimolecular recombination. This contrasts with efficiencies of less than 1% using standard methods. For example, combining trimolecular recombination, in vitro stem cell differentiation of cells such as musculoskeletal cells, and direct or indirect selection, allows for the identification of genes that control growth and development. The genes identified are candidate pharmaceutical targets. Additionally, it will be possible to identify insert polynucleotides (e.g.
  • DNAs which stimulate proliferation of nondividing cells, for example, using a cell cycle-specific promoter or a promoter induced during proliferation, coupled to a suicide gene.
  • the nonadherent cells are those that express an insert polynucleotide (e.g. DNA) which induces proliferation, while the normally nondividing cells which do not contain such an insert remain adherent.Functional Gene Discovery.
  • One of the most powerful tools available to molecular biologists for gene discovery has been the ability to efficiently screen representative cDNA libraries constructed in lambda phage. Many microbial and some classes of mammalian genes were first detected and isolated by using functional or biochemical assays to screen lambda phage libraries.
  • the retroviral genome cannot be recovered from cells that are no longer dividing. As a result this technology also cannot be applied to bioassays for which the readout is either terminal differentiation or cell death. This is not just an uninteresting special situation. It is fundamental to identifying critical genes involved in many important biological processes including the pathways whereby stem cells give rise to fully differentiated, non-proliferating tissue components, the mechanisms of apoptosis, as well as the targets of immune cytotoxicity described above.
  • the methods described here open this door to discovery. They teach how to construct a representative cDNA library in a vector infectious for mammalian cells and they describe how genes that function in cell differentiation or cell death can be selected from such a library.
  • the viral vectors described can be employed as the "lambda phage" of mammalian cells.
  • a target polynucleotide is selected based on its induction of a suicide gene construct.
  • the target polynucleotide may encode a transcript and/or a polypeptide which stimulates expression of the suicide gene.
  • suicide gene construct or “suicide gene” is meant a nucleic acid which causes cell death when expressed.
  • Polynucleotides useful as suicide genes include apoptosis-inducing genes such as p53 and other toxic sequences and cell death-inducing sequences which are known in the art, and include those disclosed herein.
  • the host cells are engineered to comprise a suicide gene construct, as described in Examples 7, 11, and 12, and in the section below.
  • Preferred suicide genes include the toxins disclosed in Example 14 (antibody section) such as Pseudomonas exotoxin A chain, diphtheria A chain, ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, etc.
  • the method can be used to select target polynucleotides which sensitize host cells to killing by an agent.
  • an agent may include an infective agent, a DNA-damaging agent, a therapeutic agent, an antisense construct, etc.
  • the direct selection method comprises: providing a plurality of host cells which contain recombinant vector comprising insert polynucleotides; culturing the host cells under conditions such that the insert polynucleotides are expressed; exposing the host cells to a compound which induces death in a cell expressing the target polynucleotide; removing the nonviable cell or the target polynucleotide, thereby directly selecting the target polynucleotide.
  • an indirect selection method may be used, wherein the host cells are sensitive to an agent, and the target polynucleotide desensitizes cells to that agent. Thus, viable cells would be selected in an indirect selection method.
  • Antisense molecules are usually single stranded DNA or
  • RNA molecules or their substituted analogues, which down-regulate expression of gene products, for example, by binding to the target RNA through Watson and Crick base pairing and prevent the translation of these RNAs or enhancing mRNA degradation
  • Step CA Antisense Nucleic Acid Drug Dev 5(2):129-32 (1998)
  • Crooke ST Antisense Nucleic Acid Drug Dev 5(2):115-22 (1998)
  • Akhtar S J Drug Target. 5(4):225-234 (1998)
  • Mizuno, T., et al Proc. Natl. Acad. Sci.
  • a target polynucleotide which encodes a product necessary or essential for cell adherence, viability, etc. is isolated based on antisense inhibition of the endogenous transcript.
  • the insert polynucleotides are cloned in the library vector in either orientation.
  • a target polynucleotide which encodes a negative regulator of a product necessary for cell adherence, viability, etc. is selected based on antisense inhibition of the endogenous transcript.
  • the host cells are unable to adhere, or to remain viable, or to grow, etc., and a target polynucleotide is selected that allows host cells to adhere, remain viable, or grow, etc.
  • the insert polynucleotides are cloned in the library vector in either orientation.
  • antisense nucleotide sequences which are complementary to a specific DNA or RNA sequence.
  • the term “antisense strand” is used in reference to a nucleic acid strand that is complementary to the “sense” strand.
  • Antisense molecules may be produced by any method, including synthesis by ligating the insert polynucleotides in a reverse orientation to a viral promoter which permits the synthesis of a complementary strand. Once introduced into a cell, this transcribed strand combines with natural sequences produced by the cell to form duplexes. These duplexes then block either transcription or translation of the natural sequence. In this manner, nonviable phenotypes may be generated.
  • the designation “negative” is sometimes used in reference to the antisense strand, and “positive” is sometimes used in reference to the sense strand.
  • the direct or indirect selection method may be used to isolate genes involved in differentiation using any tissue or cell specific promoter to drive a suicide gene.
  • tissue or cell specific promoter for example, to identify a gene involved in myeloid cell development, hematopoietic stem cells would be engineered to contain a suicide gene under the control of a myeloid cell tissue specific promoter such as CD lib. The hematopoietic stem cells are then used as host cells in the trimolecular recombination method and/or direct or indirect selection method.
  • murine ES cells are engineered which express a suicide gene upon differentiation into myeloid cells.
  • the ES cells may be elecfroporated with GDI lb promoter driving a suicide gene and selected with hygromycin to obtain stable clones. After transfer into the ES cells, target polynucleotides would be selected by expression of the suicide gene, or alternatively, by lack of expression of the suicide gene.
  • tissue restricted promoters are listed below but this invention may be carried out using any promoter with restricted expression.
  • tissue restricted promoters are also referred to herein and elsewhere as differentiation-specific promoters, and other grammatically equivalent or art- known terms.
  • Neural Neuron specific Rl ⁇ subunit of cAMP-PK promoter
  • Tryptophan Hydroxylase promoter Neural specific enolase promoter, Tyrosine hydroxylase promoter, T ⁇ l ⁇ -tubulin promoter.
  • Lung ⁇ l collagen gene promoter, rat clara cell 10 protein promoter, human surfactant protein SP-C promoter, preproendothelin promoter.
  • Liver human apolipoprotein E promoter.
  • Heart alpha B crystallin promoter, murine alpha myosin heavy chain promoter.
  • Thymus lck proximal promoter
  • T cells CD2 promoter, CD4 promoter, CD3 promoter.
  • B cells IGE ⁇ , CD19 promoter.
  • Osteoclast and osteoblast specific promoter Mouse pro- ⁇ l(I) promoter, Tartrate Resistant Acid Phosphatase (TRAP) promoter, GDI lb promoter.
  • a wide variety of cells may be used as recipients, including stem cells, pluripotent cells such as zygotes, embryos, ES cells, other cells such as lymphoid and myeloid stem cells, neural stem cells, transformed cells such as tumour cells, infected cells, differentiated cells, etc.
  • Suicide gene constracts may be introduced into the cells by any convenient means, for example, conventional techniques such as transfection (e.g. lipofection, calcium phospate precipitation, elecfroporation, etc.), microinjection, viral infection, or any other means known in the art and/or disclosed herein.
  • ES cell lines which may be used in this invention are: porcine (e.g. U.S. Patent 5,523,226, "Transgenic Swine Compositions and Methods”); murine (e.g. D3, Rl, CGR8, ABI ES cell lines); primate (e.g. rhesus monkey); rodent; marmoset; avian (e.g. chicken); bovine; rabbit; sheep; and horse.
  • porcine e.g. U.S. Patent 5,523,226, "Transgenic Swine Compositions and Methods”
  • murine e.g. D3, Rl, CGR8, ABI ES cell lines
  • primate e.g. rhesus monkey
  • rodent e.g. rhesus monkey
  • rodent e.g. rhesus monkey
  • rodent e.g. rhesus monkey
  • rodent e.g. rhesus monkey
  • rodent e.g. rhesus monkey
  • Musculoskeletal diseases are highly prevalent in our society. With the continual aging of our population the physical, mental and financial burden of conditions like osteoarthritis (which affects 40 million Americans) and osteoporosis (which affects 25 million Americans) are predicted to increase significantly. Thus, there is a tremendous demand for the development of effective therapeutic interventions for these diseases. Unfortunately, our limited understanding of both the etiology of these diseases and the basic molecular and cellular biology of the musculoskeletal system has greatly hampered efforts to identify potential targets for therapeutic intervention.
  • Tissue culture techniques have been developed that allow progenitor stem cells to develop in vitro into functionally mature, terminally differentiated cells such as chondrocytes, osteoblasts, osteoclasts, and myocytes. Using these techniques, it is possible to investigate the regulation of musculoskeletal cell differentiation.
  • a major technical obstacle to identifying candidate pharmaceutical targets is the nondividing state of terminally differentiated musculoskeletal cells.
  • the vectors currently used for library production do not replicate in nondividing cells, thus it is not possible to recover a gene of interest by current methods.
  • vaccinia-based vectors replicate in nondividing cells and express polypeptides encoded by insert polynucleotides (e.g.
  • vaccinia vectors have not been used to identify previously unknown polynucleotides (e.g. genes) of interest from a complex population of clones (i.e. insert polynucleotides).
  • trimolecular recombination method of the present invention overcomes the obstacle of low cloning efficiency in vaccinia viras. As described herein, recombination efficiencies of 90% to 100% with relatively high titers have been achieved using trimolecular recombination. This contrasts with efficiencies of less than 1% using standard methods. Combining trimolecular recombination, in vitro musculoskeletal cell differentiation, and direct or indirect selection allows the identification of genes that control growth and development. The genes identified are candidate pharmaceutical targets.
  • Every differentiated cell is distinguished from its precursors by expression of some specific gene product. Transcriptional activation of the promoter for that gene often serves as a surrogate marker of differentiation. If a constract of that specific promoter driving expression of a toxin such as the diphtheria A chain is transfected into a proliferating precursor, then any gene that promotes differentiation will result in cell death. If a differentiation promoting insert is introduced as a recombinant in a vector (e.g. a vaccinia expression vector), then it can be readily recovered from dying differentiating cells. These methods are applicable to any stem cell population that can be induced to differentiate into a defined cell type or tissue.
  • Stem cells have been described for a wide variety of tissues including but not limited to different types of blood cells, epidermal cells, neurons, glial cells, kidney cells, and liver cells. Also among these are the different stem cells of the musculoskeletal system including the precursors of chondrocytes, osteoblasts, osteoclasts, and myocytes.
  • Osteoclasts Bone is the only organ that contains a cell type, the osteoclast, whose function is to destroy the organ in which it develops and resides. This destraction, or resorption, of bone occurs throughout life and in the healthy individual is counterbalanced by de novo bone formation in a processs called bone remodeling.
  • the genetic control of osteoclast differentiation is a well understood example of stem cell differentiation.
  • the methods and sfrategies of the present invention can be applied to identify genes that regulate stem cell differentiation in pathways leading to differentiated cells such as osteoclasts. This is illustrated specifically for the analysis of osteoclast differentiation.
  • Sfrategies are described to detect and isolate genes that positively or negatively regulate differentiation including genes that are expressed in the differentiating cell itself or that are a secreted product of another cell which influences differentiation in a paracrine fashion.
  • a cell type or cell line that can be induced to differentiate into mature osteoclasts in response to a specific signal, preferably RANK Ligand (RANKL)
  • RNKL RANK Ligand
  • polynucleotides e.g. genes expressed in a recombinant vaccinia virus library
  • RAW cells are employed.
  • RAW cells are a continuously growing murine myelomonocytic cell line that can be induced to differentiate into osteoclasts by freatment with a range of concenfrations of RANK ligand (RANKL), preferably 10 ng/ml (Hsu, H. et al, Proc Natl Acad Sci USA Pd(7):3540-3545 (1999); Owens, J. M. et al, J Cell Physiol 179:110 (1999)).
  • RANKL RANK ligand
  • These or similarly responsive cells can be transfected with a suicide gene construct comprising a promoter that normally drives expression of a gene product that is recognized as a marker of fully differentiated osteoclasts but which is linked in this construct to a suicide gene.
  • the promoter is that of the osteoclast differentiation marker TRAP and the suicide gene encodes the A chain of diphtheria toxin (TRAP/DT-A).
  • a vaccinia library preferably a cDNA library, is constructed for functional gene selection, for example, using cDNA derived from cells that include but are not limited to mature bone marrow derived osteoclasts, or RAW cells or other precursors that have been induced to differentiate into osteoclasts.
  • cDNA may be isolated from either fully mature cells or cells that have been induced to initiate the differentiation program but have not yet completed the process and may express higher levels of the downstream regulatory products.
  • RAW cells or other osteoclast progenitor cells that have been transfected with a TRAP/DT-A or similar suicide gene construct are infected with the vaccinia cDNA hbrary.
  • Any vaccinia recombinant that encodes a gene product that promotes differentiation to the mature TRAP expressing phenotype will result in synthesis of the suicide gene, and death of the host cell. Such cells and their contents will be released from the cell monolayer.
  • Vaccinia virus recombinants extracted from the cells and cell contents released into the culture supernatant are enriched for the desired vaccinia recombinants. This selection process can be repeated through multiple cycles until the desired level of enrichment is achieved.
  • TRAF6 Limaga, M. A.
  • the invention also provides for the use of this method to isolate insert polynucleotides (e.g. DNA) involved in differentiation of other cells and progenitors, such as those described herein and others well-known in the art.
  • polynucleotides e.g. DNA
  • DNA antisense strand based sfrategy DNA antisense strand based sfrategy.
  • a limitation of the insert expression sfrategy is that certain regulatory products, for example those encoded by very long cDNA, may be difficult to clone as a functional full- length product.
  • Antisense inhibition is an alternative sfrategy that does not depend on cloning of full-length cDNA. In this case, total cDNA derived from the same cells as in the sense strand based sfrategy is cloned into the vaccinia transfer plasmid in a manner preventing translation, such as in reverse orientation.
  • the resulting insert cannot produce the normal product but can down-regulate expression of the endogenous mRNA and/or protein sequence, such as by hybridizing to and inhibiting translation or promoting degradation of complementary cellular mRNA sequences.
  • RAW cells or other progenitor cells containing insert polynucleotides (e.g. DNA) in a vector and also containing TRAP DT-A or similar suicide constract are treated with an agent that induces differentiation, for example, 10 ng/ml RANKL. Under these conditions almost all host cells differentiate and undergo suicide gene- mediated death. Only cells containing a recombinant vector with an insert that inhibits expression of an essential regulator of differentiation will survive and remain adherent. Virus extracted from the remaining adherent monolayer will, therefore, be enriched for sequences homologous to the desired positive regulators of differentiation.
  • This selection process can also be repeated through several cycles until the desired degree of enrichment of recombinants in the adherent monolayer is achieved.
  • the antisense polynucleotide (e.g. gene) fragments obtained can be employed to select the actual full-length coding sequence.
  • TRAF6 Limaga, M. A. et al, Genes Dev 13:1015 (1999)
  • c-Fos Wang, Z. Q. et al, Nature 360:141 (1992)
  • c-Src Soriano, P. et al. , Cell 64:693 (1991)
  • the invention also provides for the use of this method to isolate insert polynucleotides (e.g. DNA) involved in differentiation of other cells and progenitors, such as those described herein and others well-known in the art.
  • a vaccinia library is constructed from insert polynucleotides (such as DNA, for example, cDNA) derived from cells that include but are not limited to bone marrow derived osteoclastic progenitors, an enriched fraction of progenitors, or RAW cells or other precursors that have been induced to differentiate into osteoclasts.
  • Polynucleotides such as cDNA may be isolated from cells that include but are not limited to immature precursor cells or cells that have been induced to initiate the differentiation program but have not yet completed the process and may express higher levels of the downstream regulatory products.
  • the invention also provides for the use of this method to isolate insert polynucleotides (e.g. DNA) involved in differentiation of other cells and progenitors, such as those described herein and others well-known in the art.
  • polynucleotides e.g. DNA
  • DNA antisense based strategy Polynucleotides such as total cDNA derived from the same cells as in the sense strand based sfrategy above is cloned into the vaccinia vector in reverse orientation so that the recombinant gene product synthesized in infected cells cannot encode the normal gene product but can hybridize to and inhibit translation or promote degradation of complementary cellular mRNA sequences. If the targeted sequence encodes an essential factor that inhibits cell differentiation, then in the absence of an effective inhibitory signal RAW cells or other progenitor cells transfected with TRAP/DT-A or similar suicide construct will either spontaneously differentiate or will differentiate in response to otherwise suboptimal signals.
  • RAW cells or other progenitor cells transfected with TRAP/DT-A or similar suicide construct will either spontaneously differentiate or will differentiate in response to otherwise suboptimal signals.
  • Differentiation to the mature TRAP expressing phenotype will result in synthesis of the toxin, i.e., suicide gene, and death of the infected cell.
  • Such cells and their contents will be released from the cell monolayer.
  • Vaccinia viras recombinants extracted from the cells and cell contents released into the culture supernatant are enriched for sequences homologous to the desired negative regulators of differentiation. This selection process can be repeated through multiple cycles until the desired level of enrichment is achieved.
  • the antisense gene fragments obtained can be employed to select the actual full-length coding sequence.
  • a negative intracellular regulator of osteoclast differentiation has not as yet been isolated.
  • the invention also provides for the use of this method to isolate insert polynucleotides (e.g. DNA) involved in differentiation of other cells and progenitors, such as those described herein and others well-known in the art.
  • polynucleotides e.g. DNA
  • polynucleotides e.g. genes
  • the insert polynucleotide (e.g. cDNA, gene or other DNA) populations tested are or can be both expressed and function in the same type of cell.
  • polynucleotides (e.g. cDNA, gene or other DNA) expressed in one type of cell may function in the activation or differentiation of another type of cell. If such gene products can also function in autocrine fashion when they are introduced into the target indicator cell, then the strategies that can be employed remain the same, only the source of polynucleotide (e.g.
  • cDNA, gene, or other insert DNA incorporated into a vaccinia library is different.
  • the polynucleotide e.g. cDNA, gene, or other insert DNA
  • the strategy of "lethality based" e.g. lysis/nonadherence based selection described in the previous paragraphs is not applicable since the expressing cell does not itself become non- viable or non-adherent.
  • a vaccinia library is constructed in the sense orientation from insert polynucleotides (e.g. DNA, preferably cDNA) derived from cells that include but are not limited to bone marrow derived sfromal cells and or lymphoid cells.
  • Producer cells are selected that do not either induce or inhibit induction of differentiation of RAW cells or other osteoclast progenitors. These may include but are not limited to fibroblastoid or lymphoid cells and cell Hnes or RAW cells themselves.
  • RAW cells are employed as an indicator target for differentiation.
  • These or similarly responsive cells are transfected with an indicator gene (e.g. reporter gene) constract comprising a promoter that normally drives expression of a gene product that is recognized as a marker of fully differentiated osteoclasts but which is linked in this construct to expression of an easily detected indicator gene (e.g. reporter gene) product.
  • the promoter is that of the osteoclast differentiation marker TRAP and the indicator gene (e.g. reporter gene) encodes the enzyme luciferase (TRAP/luciferase).
  • Multiple cultures of producer cells are separately infected with recombinant vaccinia virus expanded from a small initial pool, preferably an initial pool of between 1 and 1000 viral pfu is expanded to 10 to 10,000 pfu prior to infection of between 100" and 10,000 producer cells.
  • Each pool of infected producer cells is cocultured with indicator cells that have been transfected with TRAP/luciferase or a similar indicator construct.
  • the invention also provides for the use of this method to isolate insert polynucleotides (e.g. DNA) involved in differentiation of other cells and progenitors, such as those described herein and others well-known in the art.
  • polynucleotides e.g. DNA
  • the invention also provides for the use of this method to isolate insert polynucleotides (e.g. DNA) involved in differentiation of other cells and progenitors, such as those described herein and others well-known in the art.
  • polynucleotides e.g. DNA
  • RAW cells or other progenitor cells transfected with TRAP/luciferase or similar indicator construct are treated with an agent that induces differentiation, in a preferred embodiment with RANKL at the lowest concentration that, in the absence of vaccima recombinants, reproducibly induces differentiation and a positive indicator signal in every microculture of producer and indicator cells.
  • the invention also provides for the use of this method to isolate insert polynucleotides (e.g. DNA) involved in differentiation of other cells and progenitors, such as those described herein and others well-known in the art.
  • polynucleotides e.g. DNA
  • Cell Proliferation Genes Genes whose aberrant expression or function may contribute to cell proliferation disorders fall into two general categories: (Smith, E. S., et al, Nat. Med. 7:967-972 (2001)) dominant transforming genes, including oncogenes, and (Brown, M. L., et al., Annu. Rev. Public Health 22:91-113 (2001)) recessive cell proliferation genes, including tumor suppressor genes and genes encoding products involved in programmed cell death ("apoptosis").
  • Oncogenes generally encode proteins that are associated with the promotion of cell growth. Because cell division is a cracial part of normal tissue development and continues to play an important role in tissue regeneration, oncogene activity, properly regulated, is essential for the survival of the organism. However, inappropriate expression or improperly controlled activation of oncogenes may drive uncontrolled cell proliferation and result in the development of severe diseases, such as cancer. Weinberg, C.A., Cancer. . Clin. 44:160-110 (1994).
  • Tumor suppressor genes normally act as "brakes” on cell proliferation, thus opposing the activity of oncogenes. Accordingly, inactivation of tumor suppressor genes, e.g., through mutations or the removal of their growth inhibitory effects may result in the loss of growth confrol, and cell proliferative diseases such as cancer may develop. Weinberg, C.A., Cancer J. Clin. 44:160-110 (199 A).
  • tumor suppressor genes are genes whose product is involved in the confrol of apoptosis; rather than regulating proliferation of cells, they influence the survival of cells in the body.
  • surveillance systems are believed to ensure that the growth regulatory mechanisms are intact; if abnormalities are detected, the surveillance system switches on a suicide program that culminates in apoptosis.
  • Mutations in tumor suppressor genes and genes encoding products involved in the confrol of apoptosis are typically recessive; i.e., both copies of the gene, the maternally inherited copy and the paternally inherited one, must be inactivated by mutation to remove the effect of the gene product.
  • a single functional copy of such genes is sufficient to maintain tumor suppression.
  • Predisposition to certain hereditary cancers involves mutant tumor suppressor genes. For example, if an individual inherits a single defective tumor suppressor gene from her father, initially her health will be uncompromised, since each cell still contains a functional copy of the gene inherited from her mother. However, as cells divide, mutations accumulate. Thus, at one point, the remaining normal copy in a cell may be inactivated by mutation to remove the function of the tumor suppressor, thereby completing one of the steps toward tumor formation. Such a cell may give rise to descendant cells which represent the early stages of cancer.
  • Tumor suppressor genes and oncogenes participate in growth control pathways in normal cells in such a way that the appropriate level of cell division is maintained. Disruption of these pathways by mutation of the component genes, oncogenes or tumor suppressor genes, is the underlying cause of cancer. Growth control in complex organisms like humans is a very important and complicated process. Thus, multiple genetic pathways for growth confrol are involved. Some pathways operate in all cell types in the body. Other pathways are much more specific and function only in certain cells. For example, signaling pathways in cell lines derived from a certain tumor type can be studied with the present invention. The invention can be used to study the role of the HER-2/ new oncogene in breast carcinoma by expressing dominant negative mutations of signaling proteins in breast cancer cell lines.
  • HER-2/ «ew (c-erbB-2) is overexpressed in 30% of breast tumors and its presence is correlated with lower survival rates of patients with these tumors (Elledge, R.M., et al, Seminars in Oncology 19:244 (1992)).
  • the HE -2/neu protein demonstrates close sequence homology with, but is distinct from, the epidermal growth factor receptor (EGFR) (Scheuter, A.L., et al, Science 229:916 (1985)).
  • EGFR epidermal growth factor receptor
  • the unregulated growth characteristics of HER- 2/new-positive tumors is hypothesized to arise, at least in part, from the effect of HER.-2/neu on intracellular signaling pathways (Kumar, R., et al, Mol Cell. Biol.
  • the invention described herein can be used to isolate cells expressing dominant negative mutations of cellular signaling proteins known to interact with the EGF receptor such as PI3K, PLC ⁇ l, Grb2, Syp, Nek, She, and p9i in several cell lines derived from breast tumors (see Table 2).
  • Cell proliferation genes can be of medical value in the identification of individuals predisposed to cancer. Because early detection and surgical resection play a vital role in survival rates, methods that facilitate early diagnosis are extremely important. One way to decrease the length of time between the appearance of tumor tissue and its detection is to survey candidate patients more frequently and more thoroughly. However, such methods of surveillance are expensive; thus it is necessary to limit scrutiny to high risk individuals. Consequently, information about genetic predisposition to cancer is extremely desirable. Because most genes that influence hereditary cancer are also involved in tumor progression, isolation of genes by somatic cell genetics has the potential to uncover such predisposing genes. Germline testing for such genes offers the chance to rate an individual's probability of contracting cancer, and expensive cancer screening efforts may be limited to those most likely to benefit from them.
  • Cell proliferation genes can be of medical value in the classification of already existing tumors based on genotype. Lowe et al, 1994, Science 266:807-810. In the past, oncologists have relied on histological examination of biopsy specimens. Though useful, histological analyses are generally hampered by their subjectivity and imprecision. Methods that classify tumors based on their genetic composition have the potential to improve the reliability of classification. Detailed knowledge about tumor genotype may serve as a prognostic indicator for the tumor and may assist in guiding the therapeutic choice.
  • Tumor Suppressor Genes Many tumor suppressor genes cause growth arrest when overexpressed in normal cells, as well as in certain tumor cell lines. Examples for tumor suppressor genes include p53 (Lin et al, Proc. Natl. Acad. Sci. U.S.A. 89:9210-9214 (1992)), Rb (Francke et al, Cytogenet. Cell Genet. 75:131-134 (1976); Cavanee et al, Nature 305:779-784 (1983); Friend et al, Proc. Natl. Acad. Sci. U.S.A.
  • tumor suppressor genes trigger growth arrest in cells at one of several positions in the cell cycle. Most frequently, however, tumor suppressors are found to cause growth arrest at the G ⁇ /S stage.
  • a selection system has been designed based on the tumor suppressor pi 6, which is described in more detail, infra.
  • selection systems are designed based on the gene encoding Rb. Overexpression of Rb is known to cause arrest in many cell lines.
  • selection systems are generated based on the breast cancer susceptibility tumor suppressor gene BRCA1.
  • BRCA1 has been shown to arrest growth of breast epithelial cell lines (Holt et al, Nat. Genet. 72:298-302 (1996)).
  • selection systems are designed based on the p53 pathway. Regulated expression of p53 and its downstream targets, such as the CDK inhibitor p21 induces either apoptosis or GI arrest in a variety of cell lines.
  • tumor suppressor genes are used in order to design selection systems for the identification of novel cell proliferation genes.
  • any gene whose expression can be manipulated to cause cell growth arrest can be used. Examples include, but are not limited to, WT1, VHL, BRCA2, NF1, NF2, P15, P21, P18, P19, P27, P57.
  • selctions is based on genes such as those listed in Examples 26-29.
  • CDK Inhibitors In some embodiments of the invention, selection systems are generated based on expression of CDK inhibitors in suitable host cells.
  • CDK inhibitors defined to date including ⁇ l5, pl6, pl8, ⁇ l9, ⁇ 21, p27, p57 cause cell cycle arrest when they are overexpressed in certain cell lines.
  • pl6 some details are already known with respect to downstream pathway components.
  • most details of the pathway of growth confrol within which the genes function are still to be elucidated.
  • CDK4 and CDK6 in the cases of pl5, pl6, and pl8, and CDK4, CDK6, and CDK2 (and CDK4, CDK6) in the case of p21, p27, and p57
  • CDK4 and CDK6 in the cases of pl5, pl6, and pl8, and CDK4, CDK6, and CDK2 (and CDK4, CDK6) in the case of p21, p27, and p57
  • CDK4 and CDK6 in the cases of pl5, pl6, and pl8, and CDK4, CDK6, and CDK2 (and CDK4, CDK6) in the case of p21, p27, and p57
  • selection systems are generated based on dissection of oncogene pathways.
  • a dominant-negative oncogene or a dominant-negative fragment of an oncogene may be ectopically expressed such that growth is inhibited or apoptosis is induced.
  • Many forms of dominant-negative oncogene mutants have been engineered.
  • receptor tyrosine kinases receptor mutants lacking an intact enzymatic domain have been shown to dominant-negatively inhibit the function, and thus signal transduction, of the wild-type receptor. Redemann et al, Mol. Cell. Biol. 72:491-498 (1992); Kashles et al, Mol Cell Biol.
  • IL-1 friterleukin 1
  • IL-1 is a major pro-inflammatory and immunoregulatory protein that stimulates fibroblast differentiation and proliferation, the production of prostaglandins, collagenase and phospholipase by synovial cells and chondrocytes, basophil and eosinophil degranulation and neutrophil activation.
  • IL-1 is involved in the pathogenesis of chronic and acute inflammatory and autoimmune diseases.
  • IL-1 is both a mediator of inflammatory symptoms and of the destraction of the cartilage proteoglycan in afflicted joints.
  • IL-1 is also a highly potent bone resorption agent. Jandiski, J. J., J. Oral Path 17:145 (1988); Dewhirst, F. E. et al, J. Immunol. 8:2562 (1985). It is alternately referred to as "osteoclast activating factor" in destructive bone diseases such as osteoarthritis and multiple myeloma.
  • IL-1 also stimulates production of other cytokines such as IL-6, which can modulate tumor development (Tartour et al, Cancer Res. 54:6243 (1994).
  • IL-1 is predominantly produced by peripheral blood monocytes as part of the inflammatory response and exists in two distinct agonist forms, IL-l ⁇ and IL-l ⁇ .
  • IL-l ⁇ is synthesized as a biologically inactive precursor, pIL-l ⁇ .
  • pIL- l ⁇ lacks a conventional leader sequence and is not processed by a signal peptidase. March, C. j., Nature 315:641-641 (1985).
  • pIL-l ⁇ is cleaved by interleukin-l.beta. converting enzyme ("ICE") between Asp-116 and Ala-117 to produce the biologically active C-terminal fragment found in human serum and synovial fluid.
  • ICE interleukin-l.beta. converting enzyme
  • ICE is a cysteine protease localized primarily in monocytes. It converts precursor IL-l ⁇ to the mature form. Black, R. A. et al, FEBS Lett. 247: 386-390 (1989); Kostura, M. j. et al, Proc. Natl Acad. Sci. USA 55:5227-5231 (1989). Processing by ICE is also necessary for the transport of mature IL
  • ICE or its homologs, also appears to be involved in the regulation of programmed cell death or apoptosis. Yuan, j. et al, Cell 75:641-652 (1993); Miura, M. et al, Cell 75:653-660 (1993); Nett-Fiordalisi, M. A. et al, J. Cell Biochem: 17B 11 (1993).
  • ICE or ICE homologs are thought to be associated with the regulation of apoptosis in neurodegenerative diseases, such as Alzheimer's and Parkinson's disease. Marx, j. and M. Baringa, Science 259:160-162 (1993); Gagliardini, V. et al, Science 253:826-828 (1994).
  • Therapeutic applications for inhibition of apoptosis may include freatment of Alzheimer's disease, Parkinson's disease, stroke, myocardial infarction, spinal atrophy, and aging.
  • ICE has been demonstrated to mediate apoptosis (programmed cell death) in certain tissue types.
  • McGr, H. Science 267:1445 (1995); Whyte, M. and Evan, G., Nature 376:11 (1995); Martin, S. J. and Green, D. R, Cell 52:349 (1995); Alnemri, E. S., et al, J. Biol. Chem. 270:4312 (1995); Yuan, J. Curr. Opin. Cell Biol. 7:211 (1995).
  • a fransgenic mouse with a disruption of the ICE gene is deficient in Fas-mediated apoptosis (Kuida, K. et al, Science 257:2000 (1995)).
  • This activity of ICE is distinct from its role as the processing enzyme for pro-IL-l ⁇ . It is conceivable that in certain tissue types, inhibition of ICE may not affect secretion of mature IL-l ⁇ , but may inhibit apoptosis.
  • Enzymatically active ICE has been previously described as a heterodimer composed of two subunits, p20 and plO (20 kDa and 10 kDa molecular weight, respectively). These subunits are derived from a 45 kDa proenzyme (p45) by way of a p30 form, through an activation mechanism that is autocatalytic. Thornberry, N. A. et al, Nature 355:768-774 (1992). The ICE proenzyme has been divided into several functional domains: a prodomain (pi 4), a p22/20 subunit, a polypeptide linker and a plO subunit. Thornberry et al, supra; Casano et al, Genomics 20:A14-AS1 (1994).
  • Full length p45 has been characterized by its cDNA and amino acid sequences.
  • the p20 and plO cDNA and amino acid sequences are also known.
  • Murine and rat ICE have also been sequenced and cloned. They have high amino acid and nucleic acid sequence homology to human ICE. Miller, D. K. et al, Ann. N.Y. Acad. Sci. 696: 133-148 (1993); Molineaux, S. M. et al, Proc. Nat. Acad. Sci. P0:1809-1813 (1993).
  • the three-dimensional structure of ICE has been determined at atomic resolution by X-ray crystallography. Wilson, K. P., et al, Nature 370:210-215 (1994).
  • the active enzyme exists as a tetramer of two p20 and two plO subunits.
  • homologs of ICE with sequence similarities in the active site regions of the enzymes.
  • Such homologs include TX (or ICE.sub.rel- ⁇ or ICH-2) (Faucheu, et al, EMBO J. 14 91A (1995); Kamens J., et al, J. Biol Chem. 270:15250 (1995); Nicholson et al, J. Biol. Chem. 270:15810 (1995)), TY (or ICE.sub.rel- ⁇ i) (Nicholson et al, J. Biol. Chem. 270:15810 (1995); ICH-1 (or Nedd-2) (Wang, L.
  • Each of these ICE homologs, as well as ICE itself, is capable of inducing apoptosis when overexpressed in transfected cell lines. Inhibition of one or more of these homologs with the peptidyl ICE inhibitor Tyr-Nal-Ala-Asp-chloromethylketone results in inhibition of apoptosis in primary cells or cell lines. Lazebnik et al, Nature 377:346 (1994).
  • the direct or indirect selection method may identify molecules similar to ICE that are involved in diseases such as chronic and acute forms of IL-1 -mediated diseases, apoptosis-mediated diseases, as well as inflammatory, autoimmune, proliferative, infectious, or degenerative diseases.
  • Degenerative diseases include Parkinson's Disease, Pick's Disease, Alzheimer's Disease, as well as Rosenthal fibres in Cerebellar Asfrocytomas, Cytoplasmic bodies in muscle and Mallory bodies in Alcoholic Liver Disease.
  • Additional IL-1- and apoptosis-mediated diseases include inflammatory diseases, autoimmune diseases, prohferative disorders, infectious diseases, degenerative diseases, necrotic diseases, osteoarthritis, acute pancreatitis, chronic pancreatitis, asthma, adult respiratory distress syndrome, glomerulonephritis, rheumatoid arthritis, systemic lupus erythematosus, scleroderma, chronic thyroiditis, Graves' disease, autoimmune gastritis, insulin-dependent diabetes mellitus (Type I), autoimmune hemolytic anemia, autoimmune neutropenia, thrombocytopenia, chronic active hepatitis, myasthenia gravis, inflammatory bowel disease, Crohn's disease, psoriasis, graft vs.
  • inflammatory diseases include inflammatory diseases, autoimmune diseases, prohferative disorders, infectious diseases, degenerative diseases, necrotic diseases, osteoarthritis, acute pancreatitis, chronic pancreatitis, asthma
  • osteoporosis multiple myeloma-related bone disorder
  • acute myelogenous leukemia chronic myelogenous leukemia
  • metastatic melanoma Kaposi's sarcoma
  • multiple myeloma sepsis septic shock, Shigellosis, Alzheimer's disease, Parkinson's disease, cerebral ischemia, myocardial ischemia, spinal muscular atrophy, multiple sclerosis, AIDS- related encephalitis, HIN-related encephalitis, aging, alopecia, and neurological damage due to stroke.
  • Apoptosis In another embodiment, direct or indirect selection is based on the phenomenon of apoptosis, i.e., programmed cell death.
  • Cells in culture can be induced to undergo apoptotic death by a variety of stimuli, depending on the particular cells. For example, certain cells enter apoptosis after exposure to glucocorticoids, tumor necrosis factors, or other natural agents. In addition, many cell types undergo apoptosis when exposed to radiation or chemotherapeutics. Further, cells may be engineered to contain genes which have been implicated in the confrol of or participation in apoptosis under the confrol of an inducible promoter.
  • Such genes include, but are not limited to bcl-2 (Korsymeyer, Immunol. Today 73:285-288 (1992)), c-myc (Shi et al, Science 257:212-214 (1992); Evan et al, Cell 69:119-128 (1992)), p53 (Rotter et al, Trends Cell. Biol. 3:46-49 (1993)), TRPM-2/SGP (Kryprianou et al, Cancer Res. 57:162-166 (1991)), and Fas/APO-1 (Itoh et al, Cell 55:233-243 (1991)).
  • Cell types which can be induced to undergo apoptosis include, for example, lymphocytes, tumor cells derived from lymphocytes, and tumors of epithelial cell origin. Activation of the FAS antigen receptor in maturing lymphocytes activates an apoptosis program. FAS antigen can be activated either by exogenous application of a FAS antibody (Velcich et al, 1995, Cell Growth Differ. 6:749-757) or by ectopic expression of an activated form of the receptor.
  • Treatment with certain steroid hormones or cross-linking of the T cell receptors on the cell surface using, for example, an antibody, can also induce apoptosis in lymphocytes, related cell or tumor lines, and tumors of epithelial origin.
  • the 3DO line for instance, responds to receptor cross-linking by undergoing apoptosis (Vito et al, 1996, Science 271:521-525), while murine thymoma W7 cells undergo apoptosis in response to dexamethasone (Bourgeois et al, 1993, Mol. Endocrin ⁇ l. 7:840-851).
  • the method of the invention is useful to identify target polynucleotides causing cells to arrest in a growth phase or to move out of one growth phase and into another.
  • it may be desirable to identify polynucleotides causing cell arrest for example at GI.
  • a polynucleotide may cause host cells arrested in a particular growth phase to move past that phase or to move into another phase.
  • a tight scattering assay is used as the viability assay, as is well known in the art.
  • FACS fluorescence activated cell sorter
  • cells When viewed in the fluorescence activated cell sorter (FACS), cells have particular characteristics as measured by their forward and 90 degree (side) light scatter properties. These scatter properties represent the size, shape and granule content of the cells. These properties account for two parameters to be measured as a readout for the viability. Briefly, the DNA of dying or dead cells generally condenses, which alters the 90 degree scatter; similarly, membrane blebbing can alter the forward scatter. Alterations in the intensity of light scattering, or the cell- refractive index indicate alterations in viability.
  • a standard is determined for a particular cell type by the forward and side scattering properties of a live cell population. This standard for scattering is subsequently used for comparison to the host cells.
  • the viability assay utilizes a viability dye.
  • annexin N is a member of a protein family which displays specific binding to phospholipid (phosphotidylserine) in a divalent ion dependent manner. This protein has been widely used for the measurement of apoptosis (programmed cell death) as cell surface exposure of phosphatidylserine is a hallmark early signal of this process.
  • Suitable viability dyes include, but are not limited to, annexin, ethidium homodimer-1, DEAD Red, propidium iodide, SYTOX Green, etc., and others known in the art; see the Molecular Probes Handbook of Fluorescent Probes and Research Chemicals, Haugland, Sixth Edition,; see Apoptosis Assay on page 285 in particular, and Chapter 16.
  • the viability dye such as annexin is labeled, either directly or indirectly, and combined with a cell population.
  • Annexin is commercially available, i.e., from PharMingen, San Diego, California, or Caltag Laboratories, Millbrae, Cahfornia.
  • the viability dye is provided in a solution wherein the dye is in a concentration of about 100 ng/ml to about 500 ng/ml, more preferably, about 500 ng/ml to about 1 ⁇ g/ml, and most preferably, from about 1 ⁇ g/ml to about 5 ⁇ g/ml.
  • the viability dye is directly labeled; for example, annexin may be labeled with a fluorochrome such as fluorecein isothiocyanate (FITC), Alexa dyes, TRITC, AMCA, APC, tri-color, Cy5, and others known in the art or commercially available.
  • FITC fluorecein isothiocyanate
  • Alexa dyes TRITC
  • AMCA amino acid adenothiocyanate
  • APC tri-color
  • Cy5 tri-color
  • the viability dye is labeled with a first label, such as a hapten such as biotin, and a secondary fluorescent label is used, such as fluorescent streptavidin.
  • a first label such as a hapten such as biotin
  • secondary fluorescent label such as fluorescent streptavidin.
  • Other first and second labeling pairs can be used as will be appreciated by those in the art.
  • the viability dye is allowed to incubate with the cells for a period of time, and washed, if necessary.
  • the cells are then sorted as outlined below to remove the viable cells and retain the nonviable cells.
  • exclusion dye staining is used as the viability assay.
  • Exclusion dyes are those which are excluded from living cells, i.e. they are not taken up passively (they do not permeate the cell membrane of a live cell). However, due to the permeability of dead or dying cells, they are taken up by dead cells. Generally, but not always, the exclusion dyes bind to
  • exclusion dye does bind DNA; this eliminates the need for a wash step.
  • exclusion dyes that require the use of a secondary label may also be used.
  • Preferred exclusion dyes include, but are not limited to, ethidium bromide; ethidium homodimer-1; propidium iodine; SYTOX green nucleic acid stain; Calcein
  • the exclusion dye is added to the cells at a concentration of from about 100 ng/ml to about 500 ng/ml, more preferably, about 500 ng/ml to about 1 ⁇ g/ml, and most preferably, from about
  • the cells and the exclusion dye are incubated for some period of time, washed, if necessary, and then the cells sorted as outlined below, to remove non- viable cells from the population.
  • cell viability assays which may be run, including for example enzymatic assays, which can measure extracellular enzymatic activity of either live cells (i.e. secreted proteases, etc.), or dead cells (i.e. the presence of intracellular enzymes in the media; for example, intracellular proteases, mitochondrial enzymes, etc.). See The Molecular Probes Handbook of Fluorescent Probes and Research Chemicals, Haugland, Sixth Edition, chapter 16 in particular.
  • At least one cell viability assay is run, with at least two different cell viability assays being preferred, when the fluors, are compatible.
  • a preferred embodiment utilizes light scattering assays (both forward and side scattering).
  • two viability assays are run, preferred embodiments utilize light scattering and dye exclusion, with light scattering and viability dye staining also possible, and all three being done in some cases as well. Viability assays thus allow the separation of nonviable or dying cells from viable ones.
  • a preferred embodiment utilizes a cell proliferation assay.
  • proliferation assay herein is meant an assay that allows the determination that a cell is either proliferating, i.e. replicating, or not replicating.
  • the proliferation assay is a dye inclusion assay.
  • a dye inclusion assay relies on dilution effects to distinguish between cell phases. Briefly, a dye (generally a fluorescent dye as outlined below) is introduced to cells and taken up by the cells. Once taken up, the dye is trapped in the cell, and does not diffuse out. As the cell population divides, the dye is proportionally diluted. That is, after the introduction of the inclusion dye, the cells are allowed to incubate for some period of time; cells that lose fluorescence over time are dividing, and the cells that remain fluorescent are arrested in a non-growth phase.
  • a dye generally a fluorescent dye as outlined below
  • the introduction of the inclusion dye may be done in one of two ways. Either the dye cannot passively enter the cells (e.g. it is charged), and the cells must be freated to take up the dye; for example through the use of a electric pulse. Alternatively, the dye can passively enter the cells, but once taken up, it is modified such that it cannot diffuse out of the cells. For example, enzymatic modification of the inclusion dye may render it charged, and thus unable to diffuse out of the cells.
  • the Molecular Probes CellTrackerTM dyes are fluorescent chloromethyl derivatives that freely diffuse into cells, and then glutathione S-fransferase-mediated reaction produces membrane impermeant dyes.
  • Suitable inclusion dyes include, but are not limited to, the Molecular
  • Probes line of CellTrackerTM dyes including, but not limited to CellTrackerTM Blue, CellTrackerTM Yellow-Green, CellTrackerTM Green, CellTrackerTM Orange, PKH26 (Sigma), and others known in the art; see the Molecular Probes Handbook, supra; chapter 15 in particular.
  • inclusion dyes are provided to the cells at a concentration ranging from about 100 ng/ml to about 5 ⁇ g/ml, with from about 500 ng/ml to about 1 ⁇ g/ml being preferred.
  • a wash step may or may not be used.
  • the cells and the inclusion dye are incubated for some period of time, to allow cell division and thus dye dilution.
  • the length of time will depend on the cell cycle time for the particular cells; in general, at least about 2 cell divisions are preferred, with at least about 3 being particularly preferred and at least about 4 being especially preferred. Because of the length of time required, vaccinia virus MVA or another vector less cytopathic than vaccinia is preferred.
  • the cells are then sorted as outlined below, to create populations of cells that are replicating and those that are not.
  • the bright (i.e. fluorescent) cells are collected; in other embodiments, for example for screening for prohferation agents, the low fluorescence cells are collected. Alterations are determined by measuring the fluorescence at either different time points or in different cell populations, and comparing the determinations to one another or to standards.
  • At least one proliferation assay is run, with more than one being preferred.
  • a proliferation assay results in a population of proliferating cells and a population of arrested cells.
  • the cell phase assay is a DNA binding dye assay. Briefly, a DNA binding dye is introduced to the cells, and taken up passively. Once inside the cell, the DNA binding dye binds to DNA, generally by intercalation, although in some cases, the dyes can be either major or minor groove binding compounds.
  • the amount of dye is thus directly correlated to the amount of DNA in the cell, which varies by cell phase; G2 and M phase cells have twice the DNA content of GI phase cells, and S phase cells have an intermediate amount, depending on at what point in S phase the cells are.
  • Suitable DNA binding dyes are permeant, and include, but are not limited to, Hoechst 33342 and 33258, acridine orange, 7-AAD, LDS 751, DAPI, and SYTO 1 , Molecular Probes Handbook, supra; chapters 8 and 16 in particular.
  • the DNA binding dyes are added in concenfrations ranging from about 1 ⁇ g/ml to about 5 ⁇ g/ml.
  • the dyes are added to the cells and allowed to incubate for some period of time; the length of time will depend in part on the dye chosen. In some embodiments, measurements are taken immediately after addition of the dye.
  • the cells are then sorted as outlined below, to create populations of cells that contain different amounts of dye, and thus different amounts of DNA; in this way, cells that are replicating are separated from those that are not.
  • cells with the least fluorescence can be separated from those that are replicating and thus contain more than a single genome of DNA. Alterations are determined by measuring the fluorescence, for example, at different time points or in different cell populations, and comparing the determinations to one another or to standards.
  • the cell phase assay is a cyclin destraction assay.
  • the host cells contain a fusion nucleic acid which comprises nucleic acid encoding a cyclin destraction box and a nucleic acid encoding a detectable molecule.
  • Cyclin destraction boxes are known in the art and are sequences that cause destraction via the ubiquitination pathway of proteins containing the boxes during particular cell phases. That is, for example, GI cyclins may be stable during GI phase but degraded during S phase due to the presence of a GI cyclin destruction box.
  • a cyclin destruction box to a detectable molecule, for example green fluorescent protein
  • a detectable molecule for example green fluorescent protein
  • multiple boxes are used, preferably each with a different fluor, such that detection of the cell phase can occur.
  • cyclin A has a destruction box comprising the sequence RTVLGVIGD (SEQ ID NO: 12); the destraction box of cyclin Bl comprises the sequence RTALGDIGN (SEQ ID NO: 13). See Glotzer et al, Nature 349:132-138 (1991).
  • YMTVS ⁇ DRFMQDSCVPKKMLQLVGVT (rat cyclin B) (SEQ ID NO: 14); KFRLLQETMYMTVSIIDRFMQNSCVPKK (mouse cyclin B) (SEQ ID NO: 15); RAILIDWLIQVQMKFRLLQETMYMTVS (mouse cyclin 131) (SEQ ID NO: 16); DRFLQAQLVCRKKLQWGITALLLASK (mouse cyclin 132) (SEQ ID NO:17); and MSVLRGKLQLVGTAAMLL (mouse cyclin A2) (SEQ ID NO:18).
  • the nucleic acid encoding the cyclin destraction box is operably linked to nucleic acid encoding a detectable molecule.
  • the fusion proteins are constructed by methods known in the art.
  • the nucleic acids encoding the destraction box is ligated to a nucleic acid encoding a detectable molecule.
  • detectable molecule herein is meant a molecule that allows a cell or compound comprising the detectable molecule to be distinguished from one that does not contain it, i.e., an epitope, sometimes called an antigen TAG, a specific enzyme, or a fluorescent molecule.
  • Preferred fluorescent molecules include but are not limited to green fluorescent protein (GFP), blue fluorescent protein (BFP), yellow fluorescent protein (YFP), red fluorescent protein (RFP), and enzymes including luciferase and ⁇ -galactosidase.
  • GFP green fluorescent protein
  • BFP blue fluorescent protein
  • YFP yellow fluorescent protein
  • RFP red fluorescent protein
  • enzymes including luciferase and ⁇ -galactosidase When antigen TAGs are used, preferred embodiments utilize cell surface antigens.
  • the epitope is preferably any detectable peptide which is not generally found on the cytoplasmic membrane, although in some instances, if the epitope is one normally found on the cells, increases may be detected, although this is generally not preferred.
  • enzymatic detectable molecules may also be used; for example, an enzyme that generates a novel or chromogenic product.
  • the results of sorting after cell phase assays generally result in at least two populations of cells that are in different cell phases.
  • positive controls can be used.
  • agents known to alter cell cycling may be used.
  • p21 is a molecule known to arrest cells in the GI cell phase, by binding GI cyclin- CDK complexes.
  • the C35 gene is differentially expressed in human cancers including, inter alia, breast, bladder, ovarian, colon, and pancreatic cancer, and melanoma.
  • the C35 gene was described in detail in U.S. Patent Application Publication No. 2002/0155447 Al, which is hereby incorporated by reference in its entirety.
  • the C35 gene is overexpressed in human breast and bladder carcinomas (see U.S. Patent Application Publication No. 2002/0155447A1).
  • the human C35 genomic locus was identified by BLAST analysis against the public human genome sequence database. This entire C35 gene (composed of four exons and three introns) is contained within a 1401 nucleotide region between 37,795,754 and 37,797,155 on chromosome 17.
  • the ERBB2/HER2 gene terminates 505 nucleotides from the 3' end of C35 while the GRB7 gene begins 7402 nucleotides from the 5' start of C35 transcription. (Locations based on. build 33 of the human genome sequence available from The National Center for Biotechnology Information.)
  • the present invention is directed to polynucleotides that promote transcription in cancer tissue, preferably breast cancer tissue, and more preferably, human breast cancer tissue.
  • the C35 promoter of the present invention comprises a polynucleotide, the sequence of which is at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO:77, as shown below (also known herein as the "long promoter"), wherein said polynucleotide promotes transcription in cancer tissue, specifically breast cancer tissue, and more specifically, human breast cancer tissue:
  • the transcription start site is identified as nucleotide 1062 in SEQ ID NO:
  • the present invention is also directed to polynucleotides having all, part, or none of the nucleotides downstream of the C35 transcription start site (that is, a polynucleotide having all, part, or none of the nucleotides between and including nucleotides 1062 and 1107 of SEQ ID NO:77).
  • the present invention comprises a polynucleotide, the sequence of which is at at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleotide sequence from about position 1 to about position 1061 ofSEQ ID NO:77.
  • the invention is further directed to fragments and/or functional derivatives of the C35 promoter of SEQ ID NO:77 that promote transcription in cancer tissue, specifically breast cancer tissue, and more specifically, human breast cancer tissue.
  • the present invention comprises a polynucleotide, the sequence of which is at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleotide sequence from about position 706 to about position 1107 of SEQ ID NO:77 (also known herein as the "short promoter"), as shown below, wherein said polynucleotide promotes transcription in cancer tissue, specifically breast cancer tissue, and more specifically, human breast cancer tissue:
  • the transcription start site is identified as nucleotide 1062 in SEQ ID NO:
  • the present invention is also directed to "short promoter" polynucleotides having all, part, or none of the nucleotides downstream of the C35 transcription start site (that is, “short promoter” polynucleotides. having all, part, or none of the nucleotides between and including nucleotides 1062 and 1107 of SEQ ID NO:77).
  • the present invention comprises a polynucleotide, the sequence of which is at at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleotide sequence from about position 706 to about position 1061 of SEQ ID NO:77.
  • the present invention comprises a polynucleotide, the sequence of which is at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleotide sequence from about position 1006 to about position 1107 of SEQ. ID NO:77 (also known herein as "promoter fragment 1"), as shown below, wherein said polynucleotide promotes transcription in cancer tissue, specifically breast cancer tissue, and more specifically, human breast cancer tissue:
  • the transcription start site is identified as nucleotide 1062 in SEQ ID NO:
  • the present invention is also directed to "promoter fragment 1" polynucleotides having all, part, or none of the nucleotides downstream of the C35 transcription start site (that is, “promoter fragment 1" polynucleotides having all, part, or none of the nucleotides between and including nucleotides 1062 and 1107 of SEQ ID NO:77).
  • the present invention also comprises a polynucleotide, the sequence of which is at at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleotide sequence from about position 1006 to about position 1061 of SEQ ID NO:77.
  • the present invention comprises a polynucleotide, the sequence of which is at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleotide sequence from about position 906 to about position 1005 of SEQ ID NO:77 (also known herein as "promoter fragment 2"), as shown below, wherein said polynucleotide promotes transcription in cancer tissue, specifically breast cancer tissue, and more specifically, human breast cancer tissue:
  • the present invention comprises a polynucleotide, the sequence of which is at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleotide sequence from about position 806 to about position 905 of SEQ ID NO:77 (also known herein as "promoter fragment 3"), as shown below, wherein said polynucleotide promotes transcription in cancer tissue, specifically breast cancer tissue, and more specifically, human breast cancer tissue:
  • the present invention a polynucleotide, the sequence of which is at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleotide sequence from about position 706 to about position 805 of SEQ ID NO:77 (also known herein as "promoter fragment 4"), as shown below, wherein said polynucleotide promotes transcription in cancer tissue, specifically breast cancer tissue, and more specifically, human breast cancer tissue:
  • the present invention a polynucleotide, the sequence of which is at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleotide sequence from about position 906 to about position 1107 of SEQ ID NO:77 (also known herein as "promoter fragment 5"), as shown below, wherein said polynucleotide promotes transcription in cancer tissue, specifically breast cancer tissue, and more specifically, human breast cancer tissue:
  • the transcription start site is identified as nucleotide 1062 in SEQ ID NO:
  • the present invention is also directed to "promoter fragment 5" polynucleotides having all, part, or none of the nucleotides downstream of the C35 transcription start site (that is, “promoter fragment 5" polynucleotides having all, part, or none of the nucleotides between and including nucleotides 1062 and 1107 of SEQ ID NO:77).
  • the present invention also comprises a polynucleotide, the sequence of which is at at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleotide sequence from about position 906 to about position 1061 of SEQ ID NO:77.
  • the present invention comprises a polynucleotide,. the sequence of which is at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleotide sequence from about position 806 to about position 1107 of SEQ ID NO:77 (also known herein as "promoter fragment 6"), as shown below, wherein said polynucleotide promotes transcription in cancer tissue, specifically breast cancer tissue, and more specifically, human breast cancer tissue:
  • the transcription start site is identified as nucleotide 1062 in SEQ ID NO:
  • the present invention is also directed to "promoter fragment 6" polynucleotides having all, part, or none of the nucleotides downstream of the C35 transcription start site (that is, “promoter fragment 6" polynucleotides having all, part, or none of the nucleotides between and including nucleotides 1062 and 1107 of SEQ ID NO:77).
  • the present invention also comprises a polynucleotide, the sequence of which is at at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleotide sequence from about position 806 to about position 1061 of SEQ ID NO: 77.
  • the present invention comprises a polynucleotide, the sequence of which is at least 90%, 95%, 96%, 97%, 98%, 99%, or 100%) identical to the nucleotide sequence from about position 806 to about position 1005 of SEQ ID NO:77 (also known herein as "promoter fragment 7"), as shown below, wherein said polynucleotide promotes transcription in cancer tissue, specifically breast cancer tissue, and more specifically, human breast cancer tissue: 806 AACTGAAAAG CGCTTTGCAG 825
  • the present invention comprises a polynucleotide, the sequence of which is at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleotide sequence from about position 706 to about position 1005 of SEQ ID NO: 77 (also known herein as "promoter fragment 8"), as shown below, wherein said polynucleotide promotes transcription in cancer tissue, specifically breast cancer tissue, and more specifically, human breast cancer tissue:
  • the present invention comprises a polynucleotide, the sequence of which is at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the nucleotide sequence from about position 706 to about position 905 of SEQ ID NO:77 (also known herein as "promoter fragment 9"), as shown below, wherein said polynucleotide promotes transcription in cancer tissue, specifically breast cancer tissue, and more specifically, human breast cancer tissue:
  • the present invention comprises a polynucleotide, the sequence of which is at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO:78, as shown below (also known herein as the "extra long promoter"), wherein said polynucleotide promotes transcription in cancer tissue, specifically breast cancer tissue, and more specifically, human breast cancer tissue:
  • the designation "functional derivative” denotes, in the context of a functional derivative of a nucleic acid sequence, a molecule that retains a biological activity (either functional or stractural) that is substantially similar to that of the original sequence.
  • This functional derivative or equivalent may be a natural derivative or may be prepared synthetically.
  • Such derivatives include more precisely C35 promoter sequences having substitutions, deletions, or additions of one or more nucleotides, provided that promoter activity is conserved.
  • the term “functional derivatives” is intended to include “fragments”, “segments", “variants” "analogs” or “chemical derivatives” of the subject matter of the present invention.
  • variant refers to a nucleic acid molecule that is substantially similar in stracture and biological activity to the nucleic acids of the present invention.
  • Bioactivity in the context of the present invention, includes, but is not limited to, promoter activity.
  • promoter activity is determined using a reporter gene assay (as described further in the Examples, infra), wherein the polynucleotide of the present invention is operably associated with a reporter gene, e.g., luciferase, and the constract is introduced into a first host cell, specifically a "normal” (i.e., non-cancer) cell, more specifically a breast cell, and more even specifically an H16N2 cell.
  • the construct is introduced into a second host cell, specifically a cancer cell, more specifically a breast cancer cell, and even more specifically a 21MT2 cell.
  • cell lines include, but are not limited to MCF- 10A, MCF-10F, MCF-10-2A, MCF12-A, MCF-12F, ZR-75-1, ZR-75-30, UACC-812, UACC-893, HCC38, HCC70, HCC202, HCC1007 BL, HCC1008, HCC1143, HCC1187, HCC1187 BL, HCC1395, HCC1395 BL, HCC1419, HCC1428, HCC1428 BL, HCC1500, HCC569, HCC1599, HCC1599 BL, HCC1806, HCC1937, HCC1937 BL, HCC1937 BL, HCC1954, HCC1954 BL, HCC2157, HCC2157
  • the reporter gene constract may be introduced into the same host cells as the promoter-reporter constract without being operably associated with the polynucleotides of the present invention.
  • the reporter gene e.g., luciferase, the activity of which may be expressed in "Relative Light Units” or RLUs
  • the reporter gene e.g., luciferase, the activity of which may be expressed in "Relative Light Units" or RLUs
  • promoter activity of the polynucleotides that is specific for or enhanced in cancer cells is present and/or retained if the difference in luciferase expression between the cancer host cells and the normal host cells, as measured in RLUs, is at least 2-fold, specifically at least 4-fold, and more specifically, at least 6.5- fold greater in the cancer cells than in the normal cells.
  • heterologous sequence refers to two or more polynucleotide sequences not joined together in nature.
  • Non-limiting examples of heterologous sequences relative to the polynucleotides of the present invention include reporter genes such as chloramphenicol acetyl transferase (CAT), green fluorescent protein (GFP), blue fluorescent protein (BFP), yellow fluorescent protein (YFP), red fluorescent protein (RFP), beta- galactosidase, and the like, which can be juxtaposed or joined to the C35 promoter polynucleotides of the present invention.
  • CAT chloramphenicol acetyl transferase
  • GFP green fluorescent protein
  • BFP blue fluorescent protein
  • YFP yellow fluorescent protein
  • RFP red fluorescent protein
  • beta- galactosidase beta- galactosidase
  • the isolated nucleic acid molecules are also intended to include allelic variations, so long as the sequence is a functional variant of the C35 promoter sequence.
  • a C35 promoter allele does not encode the identical sequence to that found in SEQ ID NO: 77 it can be isolated and identified as a C35 promoter using the same techniques used herein, and especially PCR techniques to amplify the appropriate region with primers based on the sequences disclosed herein.
  • allele defines an alternative form of a gene that occupies a given locus on a chromosome.
  • a “mutation” is a detectable change in the genetic material which can be transmitted to a daughter cell.
  • a mutation can be, for example, a detectable change in one or more deoxyribonucleotides.
  • nucleotides can be added, deleted, substituted for, inverted, or transposed to a new position. Spontaneous mutations and experimentally induced mutations exist.
  • a or “an” entity refers to one or more of that entity; for example, “a polynucleotide,” is understood to represent one or more polynucleotides.
  • the terms “a” (or “an”), “one or more,” and “at least one” can be used interchangeably herein.
  • nucleotide sequence of the polynucleotide is identical to the reference sequence except that the polynucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence, while still retaining promoter activity.
  • nucleotide having a nucleotide sequence at least 95% identical to a reference nucleotide sequence up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence, while still retaining promoter activity.
  • This calculation is an example, and also applies to polynucleotides having other percent identities to a reference sequence (e.g., polynucleotides having a nucleic acid sequence that is at least 90%, 96%, 97%, 98%, 99%, or 100% identical to a reference sequence).
  • whether any particular nucleic acid is at least
  • nucleotide sequence of the present invention can be determined conventionally using known computer programs.
  • a preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence, also referred to as a global sequence alignment, can be determined using the FASTDB computer program based on the algorithm of
  • RNA sequence can be compared by converting U's to T's.
  • the result of said global sequence alignment is in percent identity.
  • Preferred parameters used in a FASTDB alignment of DNA sequences to calculate percent identity are:
  • Penalty 0.05, Window Size 500 or the length of the subject nucleotide sequence, whichever is shorter.
  • a manual correction must be made to the results. This is because the FASTDB program does not account for 5' and 3' truncations of the subject sequence when calculating percent identity. For subject sequences truncated at the 5 'or 3 'ends, relative to the query sequence, the percent identity is corrected by calculating the number of bases of the query sequence that are 5' and 3' of the subject sequence, which are not matched/aligned, as a percent of the total bases of the query sequence.
  • Whether a nucleotide is matched/aligned is determined by results of the FASTDB sequence alignment. This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score. This corrected score is what is used for the purposes of the present invention. Only bases outside the 5' and 3' bases of the subject sequence, as displayed by the FASTDB alignment, which are not matched/aligned with the query sequence, are calculated for the purposes of manually adjusting the percent identity score.
  • a 90 base subject sequence is aligned to a 100 base query sequence to determine percent identity.
  • the deletions occur at the 5' end of the subject sequence and therefore, the FASTDB alignment does not show a matched/alignment of the first 10 bases at 5' end.
  • the 10 unpaired bases represent 10% of the sequence (number of bases at the 5' and 3' ends not matched/total number of bases in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 bases were perfectly matched the final percent identity would be 90%.
  • a 90 base subject sequence is compared with a 100 base query sequence.
  • deletions are internal deletions so that there are no bases on the 5'or 3' of the subject sequence which are not matched/aligned with the query, hi this case the percent identity calculated by FASTDB is not manually corrected.
  • bases 5' and 3' of the subject sequence which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to be made for the purposes of the present invention.
  • Table 4 lists various transcription factors and other predicted binding sites by location within the C35 short promoter.
  • "Recognition Sequence” refers to the idealized binding site for each transcription factor and should be compared to the actual sequence within the C35 promoter fragments to determine quahty of the call.
  • call quality is a fimction of homology and length.
  • the predicted transcription factor binding sites are based on standard string searches.
  • Figure 25 shows the distribution of these sites along the length of the C35 long promoter.
  • Table 5 is a reference table for ambiguous binding sites shown in Table 4.
  • This example describes the construction and characterization of a new set of direct ligation vectors designed to be universally applicable for the generation of chimeric vaccinia genomes.
  • the aim was to modify the genome of vNotl/tk so as to acquire direct ligation vectors which are more universally useful.
  • the insertion site was changed by placing the sites for two unique restriction enzymes at the beginning of the thymidine kinase gene. This allows one to fix the orientation of the insert polynucleotide (e.g. DNA) and eliminates the production of contaminating wild type genomes after religation of viral arms.
  • the thymidine kinase gene was preceded by a strong constitutive vaccinia viras promoter.
  • Notl and Apal to eliminate rehgation of poxviras arms and fix the orientation of the insert polynucleotide (e.g. DNA) behind strongly expressing constitutive vaccinia promoters.
  • the insertion cassette has been placed at the beginning of the thymidine kinase gene in vaccinia to utilize drag selection in the isolation of recombinants.
  • oligonucleotides were constructed which, when annealed, contained the 7.5k gene promoter (MM436: GGCCAAAAATTGAAAAACTAGATCTATTTATTGCACGCGGCCGCCA TGGGCCC (SEQ ID NO:19) and MM437: GGCCGGGCCCATGGCGG CCGCGTGCAATAAATAGATCTAGTTTTTCAATTTTT (SEQ ID NO:20)), or the synthetic EL promoter (MM438: GGCCAAAAATTG AAATTTTATTTTTTTTTTTTGGAATATAAAGCGGCCGCCATGGGCCC (SEQ ID NO:21) and MM439: GGCCGGGCCCATGGCGGCCGCTT TATATTCCAAAAAAAAAAAAAAATAAAATTTCAATTTTT (SEQ ID NO:22)) and restriction sites for Notl and Apal.
  • MM436 GGCCAAAAATTGAAAAACTAGATCTATTTATTGCACGCGGCCGCCA TGGGCCC
  • MM437 GGC
  • the double-sfranded oligonucleotides were annealed by ramping from 94° C to 20 °C over two hours and ligated into the Notl site present in pJNotl/tk, a plasmid containing the Hindlll J fragment from vNotl/tk, resulting in plasmids p7.5/tk and pEL/tk.
  • PCR Polymerase Chain Reaction
  • the plasmids were excised with Notl (MM440/MM442 product) and cloned into pJNot/tk digested with Notl yielding pJNot/tk-GUS, or excised with Notl and Apal (MM440/MM441 product), and inserted into pEL/tk and p7.5/tk previously digested with Apal and Notl yielding p7.5/tk- GUS and pEL/tk-GUS.
  • oligonucleotides Two oligonucleotides were constructed which, when annealed, contained the 7.5k gene promoter and the nucleotide sequence encoding for a cytotoxic T-cell epitope for ovalbumin (Chan, W. K., et al., Nucleic Acids Res.
  • Vaccinia viras was generated from viral DNA by rescue with fowlpox virus (Scheiflinger, et al, 1992, Proc. Natl. Acad. Sci. USA 89:9977-9981). Vaccima viras was isolated from infected HeLa cells by banding and sedimentation in sucrose (Earl, et al, In Ausubel, et ⁇ /.(eds.), Current Protocols in ' Molecular Biology, Greene Publishing Associates/Wiley Interscience, New York (1991)).
  • the purified virions were freated with Proteinase K (Boehringer Mannheim) and gently extracted with buffer saturated phenol, phenokchloroform (50:50), and chloroform before precipitation with 2.5 volumes of ethanol in 0.3M sodium acetate and resuspended in TE (lOmM TrisHCl, ⁇ H8.0. ImM EDTA (Earl, et al, In
  • Confluent wells of BSC-1 cells from a 12 well dish were infected with fowlpox virus and after a two hour incubation at 37° C were transfected with 0.6 ⁇ g full length vaccinia DNA using Lipofectamine (Life Technologies Incorporated) as suggested by the manufacturer. After 24, 48, and 72 hours the cells were harvested, lysed by three freeze-thaw cycles and screened by plaque assay on
  • AGTTAGGCCCCCCCTCGA (SEQ ID NO:32) was digested with Apal and
  • vEL/tk DNA arms and the DNA fragment encoding ovalbumin were ligated overnight at room temperature, at a 4:1 (insert: viras) molar ratio, in 30 microliters with 5 units T4 DNA Ligase.
  • the ligation product was transfected using lipofectamine (Life Technologies, Inc.) into a well of confluent BSC-1 cells from a 12 well plate two hours after infection with fowlpox virus at 1 pfu/cell. Three days later the cells were harvested and isolated plaques were selected by passage in Hutk- cells in the presence of bromodeoxyuridine (Earl, et al, In Ausubel, et al. (eds.), Current Protocols in Molecular Biology, Greene Publishing Associates/Wiley Interscience, New York (1991)).
  • BSC-1 cells were infected at high multiplicity of infection (moi) by vaccinia WR, vEL/tk, v7.5/tk, or vNotl/tk. After 24 hours the cells were harvested and resuspended in Cell Suspension Buffer (Bio-Rad Genomic DNA Plug Kit) at lxlO 7 cells/ml. An equal volume of 2% CleanCut agarose (Bio-Rad) preincubated at 50° C was added and the cell suspension was formed into 100 ⁇ l plugs. After hardening at 4° C the plugs were freated as previously described to digest protein (Merchlinsky, et al, J. Virol.
  • the plugs were equilibrated in the appropriate restriction enzyme buffer and ImM PMSF for 16 hours at room temperature, incubated with restriction enzyme buffer, lOOng/ml Bovine Serum Albumin and 50 units Notl or Apal for two hours at 37 °C (Notl) or room temperature (Apal) prior to electrophoresis.
  • ImM EDTA ImM EDTA
  • 2.5 microliters were digested with HindHI, Hindm and Apal, or Hindm and Notl, electrophoresed through a 1.0% agarose gel, and transferred to Nyfran (Schleicher and Schuell) using a Turboblotter (Schleicher and Schuell).
  • the samples were probed with p7.5/tk (FIG. 2A) or pEL/tk (FIG. 2B) labeled with 32 P using Random Primer DNA Labeling Kit (Bio-Rad) in QuickHyb (Sfratagene) and visualized on Kodak XAR film.
  • MC57G cells in wells of a 6 well plate were infected at an moi of 1 with vEL/tk, v7.5/tk-ova, vEL/tk-ova, vEL/tk-ovaFL clone 1, and vEL/tk-ovaFL clone 2 (vEL/tk-ovaFL are virus clones of full length ovalbumin generated by direct ligation).
  • vEL/tk-ovaFL are virus clones of full length ovalbumin generated by direct ligation.
  • At 16 hours post infection cells were harvested, labeled with 100 microcuries 51 Chromium (Dupont) for 1 hour at 37°C, and 10 4 cells were added to wells of a 96 well round bottom plate in quadruplicate.
  • a sample of uninfected MC57G cells incubated with 1 micromolar purified ova 257-264 peptide was also incubated with 51 Cr as a positive control and untreated MC57G cells were used as a negative confrol.
  • T cells specific for ova 257- 264 were added to target cells at ratios of 2:1 and 10:1.
  • Cells were incubated at 37 °C for 4 hours, supematants were harvested, and 51 Cr release determined. Spontaneous release was derived by incubating target cells with media alone and maximal release was determined by incubating target cells with 5% Triton X 100.
  • % specific lysis ((experimental release-spontaneous release) / (maximal release- spontaneous release)) X 100. In each case the mean of quadruplicate wells was used in the above formula.
  • the vaccinia WR genome is approximately 190 kilo-bases in length and rich in A and T residues.
  • the complete sequence of the vaccinia WR genome was provided by P. Earl of the Bernard Moss laboratory (Laboratory of Viral Diseases, NIADD, NIH, Bethesda, MD).
  • a restriction enzyme search of the complete sequence of the vaccinia WR genome using Mac Vector (IBI) revealed a lack of restriction sites for Apal, Ascl, Bs ⁇ l20I, Fsel, RsrII, Sfil, Srfl and Sgfl.
  • the ready availability of highly active and pure preparations of the enzyme as well as the generation of a staggered end upon digestion led us to choose to use Apal as the second site in conjunction with the Notl site already present in vNot/tk.
  • Vaccinia virus based expression vectors are most useful when the foreign protein is expressed constitutively.
  • the expression of foreign proteins during the early stage of viral replication is essential for cytotoxic T cell response (Bennick, et al. Topics Microbiol. Immunol. /65:153-184 (1990)) and high levels of total protein expression have been observed using promoters active during the late stage of viral replication.
  • a useful feature of vNotl/tk that must be retained in any new vector is the ability to discriminate for recombinant viral genomes using selection against an active thymidine kinase gene.
  • the introduction of the Apal site within the coding sequence for the tk gene necessitates an increase in the total number of amino acids in order to accommodate the restriction enzyme site.
  • a comparison of the amino acid sequence for thymidine kinase genes from a variety of animal and viral species showed the region of greatest heterogeneity was at the N terminus of the protein, suggesting that this region of the protein could tolerate a modest increase in the number of amino acids.
  • the recombination-independent cloning vectors were constructed by making plasmid intermediates containing the modified thymidine kinase (tk) gene and replacing the tk sequence in the vNotl tk genome by homologous recombination. Two sets of oligonucleotide pairs were constructed which, when annealed, contained the promoter for the 7.5k gene or the synthetic EL sequence and restriction sites for Notl and Apal.
  • tk modified thymidine kinase
  • the modified thymidine kinase genes were constructed by annealing the double-sfranded oligonucleotides and ligating the product into the Notl site present at the beginning of the thymidine kinase gene in pJNotl tk, a plasmid containing the Hindlll J fragment from vNotl/tk.
  • the acquisition of the Apal site was verified by restriction enzyme analysis of plasmid DNA and the nucleotide sequence of the thymidine kinase gene promoter was determined and found to be as depicted in FIG. 1.
  • the recombinant viruses derived from p7.5/tk and pEL/tk were isolated using a strategy relying on positive drug selection in the presence of HAT (hypoxanthine, aminopterin, thymidine) (Weir, et al, Proc. Nat. Acad. Sci. USA 7P:1210-1214 (1982)).
  • HAT hypoxanthine, aminopterin, thymidine
  • viruses vpNotl a virus that contains a copy of pBR322 inserted at the Notl site of vNotl/tk (Merchlinsky, et al, Virology 190:522-526 (1992)), and vNotl/lacZ/tk, a virus with a copy of the lacZ gene interrupting the thymidine kinase in vNotl " (Merchlinsky, et al, Virology. 190:522-526 (1992)) are thymidine kinase negative (tk " ) viruses that are identical to vNotl/tk except for the inserted DNA at the beginning of the tk gene.
  • the plasmids p7.5/tk and pEL/tk were recombined with vpNotl and vNo acZ/tk helper viruses in CV-1 cells and the infected monolayers were harvested and passaged in the presence of HAT media on Hutk " cells. Individual plaques were passaged and isolated an additional three rounds on Hutk " cells before expansion and analysis.
  • v7.5/tk and vEL/tk viras in HAT supplemented media implies these viruses, in contrast to vpNot and vNot/lacZ/tk, contain an active thymidine kinase (tk) gene.
  • tk active thymidine kinase
  • an active tk gene could arise from multiple crossovers which delete the 7.5k or EL promoter sequences, generating a virus with the normal tk promoter.
  • the v7.5/tk and vEL/tk genomes should contain a unique site for both Notl and Apal within the HindlH J fragment.
  • Uncut vaccinia WR migrates at a size of 190 kilobase pairs as compared to multimers of bacteriophage lambda (lane 1).
  • both v7.5/tk and vEL/tk contain a unique site for both Apal and Notl, the sites are at the same locus as the Notl site in vNot/tk, and the sites are in a more central location in the genome than the HindlH F fragment which contains the Notl site in vaccinia WR.
  • the background of cellular DNA fragments was more pronounced in the Apal digestion, which has a six base pair recognition site, than for the Notl digest.
  • the genomes for vEL/tk and v7.5/tk were analyzed by Southern blotting to confirm the location of the Apal and Notl sites in the HindlH J fragment as shown in FIG.2.
  • the filters were hybridized to 32 P labeled HindlH J fragment derived from the p7.5/tk or pEL/tk.
  • the genomes for v7.5/tk and vEL/tk have an Apal site that does not appear in vNotl/tk (compare lanes 7 and 8 to lane 5 in each blot) whereas digestion with Notl and HindlH yield a set of fragments of equivalent size.
  • the 0.5 kilobase Hind.II/NotI or HindlH/ Apal fragment from the left hand side of HindlH J produced from Notl or Apal digestion has electrophoresed off the bottom of the agarose gel.
  • the definitive characterization of the promoter sequence utilized products of Polymerase Chain Reaction (PCR).
  • PCR Polymerase Chain Reaction
  • a pair of primers flanking the beginning of the tk gene were used to generate a DNA fragment from the viruses vNotl/tk, v7.5/tk, or vEL/tk and their cognate plasmids as shown in FIG. 4.
  • the PCR products for v7.5/tk and vEL/tk are the same size as those observed for the plasmids used to generate the viruses (p7.5/tk and pEL/tk) and larger than those seen for vaccinia WR and vNotl/tk.
  • the PCR fragments were cloned into the plasmid pCRH, the nucleotide sequence was determined and shown to match the sequence displayed in FIG. 1.
  • RNA synthesis was measured by infecting confluent BSC-1 cells in the presence and absence of cytosine arabinoside (AraC) at an moi of 5, harvesting the cells, isolating the RNA using Trizol (Life Technologies) and analyzing the level of thymidine kinase RNA synthesis by primer extension (Weir, et al, Nucleic Acids Research 16:10261-10282 (1990)). Incubation with AraC blocks viral DNA replication, allowing one to identify the class of viral promoter.
  • cytosine arabinoside AraC
  • the early class of viral promoters are active prior to DNA replication and will be unaffected by AraC in the infection. Late promoters are only expressed after the onset of DNA replication and their activity is abrogated in the presence of AraC. Perasal ofthe products on a denaturing polyacrylamide gel demonstrated that significantly more (estimated to be at least ten fold) tk RNA primer extension products were synthesized in vEL/tk infections as compared to vNot/tk.
  • RNA start site insensitive to AraC incubation In cells infected with vNot/tk a single RNA start site insensitive to AraC incubation was observed whereas in vEL/tk infections two distinct start sites, one resistant to AraC and corresponding to the appropriate early start site (Davison, et al, J. Mol. Biol. 210:149-169 (1989)), and one species sensitive to AraC and corresponding to the appropriate late start of RNA (Davison, et al. J. Mol. Biol. 210:111-184 (1989)) were observed (data not shown).
  • the pattern of RNA species derived from infection with v7.5/tk was similar to that observed for vEL/tk with the absolute levels of RNA expression intermediate to that observed for vEL/tk and vNot/tk.
  • the plasmids were recombined with vNotl/tk and the recombinant viruses identified by staining with X-glu (Carroll, et al, BioTechniques 79:352-355 (1995)), passaged for three rounds through Hutk " cells, and expanded to generate the viral stocks vNotl/ tk-GUS, v7.5/ tk-GUS and vEL/tk-GUS.
  • the structures ofthe recombinant virases were verified by Southern blot analysis.
  • the level of expression of ⁇ -glu by vNotl/tk-GUS, v7.5/ tk-GUS and vEL/tk-GUS was measured from infected confluent monolayers of BSC-1 cells in the presence or absence of AraC (FIG. 5).
  • the level of ⁇ -glu expression for the v7.5/tk-GUS and vEL/tk was much higher than that observed for vNotl/tk-GUS and highest (approximately twenty fold higher) in the vEL/tk-GUS.
  • Expression of ⁇ -glu was observed for all three virases in the presence of cytosine arabinoside, indicating that each promoter is a member of the early class of viral promoters.
  • ⁇ -glu in vNotl/ tk-GUS was unchanged in the presence or absence of AraC indicating that this promoter is only active early during infection, whereas the ⁇ -glu levels in v7.5/ tk-GUS and vEL/tk-GUS were lower in the presence of AraC, indicating these promoters are active both early and late times during infection.
  • v7.5/tk and vEL/tk vectors were initially isolated by growth in the presence of HAT supplemented media and are designed to contain an active tk gene to allow selection for viruses with inserts via passage in Hutk " cells in the presence of bromodeoxyuridine (Earl, et al, In Ausubel, et al. (eds.), Current Protocols in Molecular Biology, Greene Publishing Associates/Wiley Interscience, New York (1991)). Both vectors were tested by plaque assay in Hutk " cells using drug selection and the results for vEL/tk are shown in FIG. 6.
  • Fowlpox should be a superior helper viras as it is used at 37°C, will not revert to a highly replicating strain, and, since it does not recombine with vaccinia DNA or productively infect primate cell lines, can be used at higher moi than vaccinia.
  • fowlpox can serve as an efficient helper virus a series of wells from a 12 well plate containing BSC-1 cells were infected with varying Mois of fowlpox and transfected with full length vaccinia WR DNA, the cells were harvested after 24, 48, or 72 hours and the virus titer was determined as shown in Table 6. Transfection of DNA without fowlpox or fowlpox infection alone resulted in no plaques. The level of rescued vaccinia increased with later harvest and was proportional to the moi of the fowlpox infection.
  • DNA was fransfected into BSC-1 cells infected with fowlpox viras using lipofectamine as described in Materials and Methods. The cells were harvested at 1, 2, or 3 days post fransfection, lysed by freeze-thaw cycles and assayed for infectious viras by plaque assay on BSC-1 cells.
  • the ovalbumin insert was modified to include a Notl site at its 5' end, translation stop codons, a vaccinia transcription stop signal and an Apal site at its 3' end. This insert was digested with Notl and Apal and ligated with purified vEL/tk DNA arms that had been digested with Notl and Apal.
  • the ligation mix was fransfected into fowlpox infected BSC-1 cells, cells were harvested, and after three days the cell extract was passaged on Hutk " cells in the presence or absence of 125mM bromodeoxyuridine.
  • the titer obtained without drug selection was 2.7 X 10 3 pfu and with drug selection 2.8 X 10 3 pfu.
  • Individual plaques were picked from Hutk " cells in the presence and absence of bromodeoxyuridine and tested for the presence ofthe ovalbumin insert by dot blot hybridization with an ovalbumin cDNA probe. All 15 plaques picked in the presence of bromodeoxyuridine, and all 10 plaques picked in its absence contained the ovalbumin insert.
  • vEL/tk-ovaFL Two individual clones were expanded further and tested for the ability to sensitize host cells to lysis by ova 257-264 specific cytotoxic T lymphocytes (CTL). The results of this experiment are shown in Table 7. As controls, vaccinia recombinant for an ova 257-264 minigene, v7.5/tk-ova and vEL/tk- ova, were generated by homologous recombination. These ova peptide recombinant virases were tested in concert with the vEL/tk-ovaFL clones for the ability to sensitize host cells to lysis by ova specific CTL.
  • CTL cytotoxic T lymphocytes
  • CML Cell mediated lymphocytotoxicity assay on recombinant vaccinia viras infected cells.
  • Virally infected MC57G cells were generated as described in (Materials and Methods).
  • One sample of MC57G cells was treated with ova257-264 peptide (l ⁇ M), another sample of cells was left untreated.
  • Cells were incubated with two different ratios of ova specific cytotoxic T lymphocytes for 4 hours at 37 °C and percent specific lysis was determined as described in Materials and Methods.
  • DNA virases are particularly useful expression vectors for the study of cellular processes as they can express many different proteins in their native form in a variety of cell lines.
  • gene products expressed in recombinant vaccima virus have been shown to be efficiently processed and presented in association with MHC class I for stimulation of cytotoxic T cells.
  • the gene of interest is normally cloned in a plasmid under the confrol of a promoter flanked by sequences homologous to a non-essential region in the virus and the cassette is introduced into the genome via homologous recombination.
  • a panoply of vectors for expression, selection and detection have been devised to accommodate a variety of cloning and expression strategies.
  • homologous recombination is an ineffective means of making a recombinant virus in situations requiring the generation of complex libraries or when the insert polynucleotide (e.g. DNA) is large.
  • Poxviruses are ubiquitous vectors for studies in eukaryotic cells as they are easily constructed and engineered to express foreign proteins at high levels. The wide host range of the viras allows one to faithfully express proteins in a variety of cell types.
  • Fowlpox viras will efficiently generate infectious viras but not replicate in the host cell or recombine with the vaccinia DNA products.
  • Fowlpox viras has the properties of an ideal helper virus as it is used at 37°C, will not revert to a highly replicating strain, and, since it does not recombine with vaccinia DNA or productively infect primate cell lines, can be used at relatively high moi.
  • vNotl/tk The utility of the vaccinia based direct ligation vector vNotl/tk, has been described by Merchlinsky, et al, Virology 190:522-526 (1992). This genome lacks the Notl site normally present in the HindlH F fragment and contains a unique Notl site at the beginning of the thymidine kinase gene in frame with the coding sequence. This allows the insertion of DNA fragments into the Notl site and the identification of recombinant genomes by drag selection.
  • the vNotl/tk vector will only express foreign proteins at the level ofthe thymidine kinase gene, a weakly expressed gene only made early during viral infection. Thus, the vNotl/tk vector can be used to efficiently clone large DNA fragments but does not fix the orientation of the DNA insert or lead to high expression ofthe foreign protein.
  • This example describes the constraction and characterization of a pair of vaccinia DNA vector genomes v7.5/tk and vEL/tk suitable for direct ligation.
  • the v7.5/tk and vEL/tk vectors were designed to contain unique restriction sites for Notl and Apal at the beginning of the thymidine kinase gene allowing the oriented cloning of DNA and eliminating the intact genomes arising from relegation of vaccima vector arms.
  • thymidine kinase gene as the insertion site for foreign DNA allows selection for recombinants and against helper or wild type genomes.
  • the level of tk expression in v7.5/tk and vEL/tk should be much higher than in vaccinia WR or vNot/tk.
  • the Apal site at the beginning of the tk gene in v7.5/tk and vEL/tk was formed from vNot/tk by adding extra nucleotides at the Notl site.
  • the additional nucleotides increase the amino acid sequence at the N terminus ofthe wild type tk gene from Met-Asn-Gly to Met-Gly-Pro-Ala-Ala-Asn-Gly (SEQ ID NO:35) in v7.5/tk and vEL/tk.
  • Modifications in the expression level and N terminal amino acid sequence of the thymidine kinase gene may increase (more protein) or decrease (different . sequence) the sensitivity of the viras to bromodeoxyuridine. Plaques, albeit smaller, were observed with v7.5/tk and vEL/tk infection at a concenfration of bromodeoxyuridine sufficient to completely suppress plaque formation for wild type vaccinia WR.
  • bromodeoxyuridine Plaque formation was suppressed at five-fold higher concenfrations of bromodeoxyuridine, a level of drug that does not interfere with the viability ofthe cells or impede the ability of tk " virus to form plaques.
  • the explanation for the altered sensitivity to bromodeoxyuridine awaits further characterization of the protein as the altered thymidine kinase gene may have a different reaction rate for formation ofthe triphosphate form ofthe bromodeoxyuridine or a reduced ability to bind bromodeoxyuridine.
  • the development of direct ligation vectors has increased the possible applications for poxvirus expression vectors.
  • the v7.5/tk and vEL/tk vectors were designed to incorporate the advantages of oriented cloning, high levels of expression of foreign protein, and the selection for recombinant virases, into direct ligation vectors. They were shown to express high levels of proteins at all times during infection.
  • the utility of these vectors was demonstrated by constructing recombinants containing a CTL epitope for ovalbumin (constructed by homologous recombination with a plasmid) or the ovalbumin coding sequence (constructed by direct ligation protocol) and showing how both recombinants were able to elicit a strong CTL response.
  • v7.5/tk and vEL/tk vectors provide a set of universally applicable direct hgation cloning vectors for poxviruses.
  • p7.5/tk and pEL/tk included, respectively, either the 7.5K vaccinia viras promoter or a strong synthetic early/late (E/L) promoter (FIG. 1).
  • the Apa I site was preceded by a strong translational initiation sequence including the ATG codon. This modification was introduced within the vaccinia viras thymidine kinase (tk) gene so that it was flanked by regulatory and coding sequences of the viral tk gene.
  • Recombinants Standard methods for generation of recombinants in vaccinia viras exploit homologous recombination between a recombinant vaccinia fransfer plasmid and the viral genome.
  • Table 8 shows the results of a model experiment in which the frequency of homologous recombination following fransfection of a recombinant fransfer plasmid into vaccinia virus infected cells was assayed under standard conditions.
  • a minigene encoding the immunodominant 257-264 peptide epitope of ovalbumin in association with H-2K b was inserted at the Not 1 site in the fransfer plasmid tk gene.
  • the disrupted tk gene is substituted for the wild type viral tk+ gene in any recombinant viras.
  • This serves as a marker for recombination since tk- human 143B cells infected with tk- virus are, in contrast to cells infected with wild type tk+ virus, resistant to the toxic effect of BrdU.
  • Recombinant viras can be . scored by the viral pfu on 143B cells cultured in the presence of 125 mM BrdU. [0656] The frequency of recombinants derived in this fashion is ofthe order of
  • This recombination frequency is too low to permit efficient construction of a cDNA library in a vaccinia vector.
  • the following two procedures were used to generate an increased frequency of vaccinia virus recombinants.
  • (1) One factor limiting the frequency of viral recombinants generated by homologous recombination following transfection of a plasmid fransfer vector into vaccinia virus infected cells is that viral infection is highly efficient whereas plasmid DNA transfection is relatively inefficient. As a result many infected cells do not take up recombinant plasmids and are, therefore, capable of producing only wild type viras.
  • FPV Fowl Pox Virus
  • This insert is flanked by vaccinia tk DNA.
  • One sample was fransfected with 600ng untreated genomic V7.5/tk DNA.
  • Three days later cells were harvested, and the virus was extracted by three cycles of freeze/thaw in dry ice isopropanol 37° C water bath. Crude viral stocks were plaqued on TK- 143 B cells with and without BrdU selection.
  • each PCR product would contain either no ATG start codon, or an ATG start codon in each ofthe three possible reading frames.
  • each PCR product contains at its 3 prime end, franslation stop codons in all three reading frames, and a vaccinia virus transcription double stop signal.
  • These 4 PCR products were ligated separately into the ⁇ otI/ Apal sites of pBS.Ntk.MCS-, generating the 4 vectors, p7.5/ATG0/tk, p7.5/ATGl/tk, p7.5/ATG3/tk, and p7.5/ATG4/tk whose sequence modifications relative to the p7.5/tk vector are shown in FIG. 8.
  • Each vector includes unique BamHI, Smal, Pstl, and SaU sites for cloning D ⁇ A inserts that employ either their own endogenous translation initiation site (in vector p7.5/ ATGO/tk) or make use of a vector translation initiation site in any one of the three possible reading frames (p7.5/ATGl/tk, p7.5/ATG3/tk, and p7.5/ATG4/tk).
  • cD ⁇ A was synthesized from poly-A+ mR ⁇ A of a murine tumor cell line (BCA39) and ligated into each ofthe four modified p7.5/tk fransfer plasmids.
  • the fransfer plasmid is amplified by passage through procaryotic host cells such as E. coli as described herein or as otherwise known in the art.
  • DMEM fetal bovine serum
  • BrdU BrdU
  • DMEM-2.5 DMEM-2.5. At the end of the two hour infection period the cells were overlayed with 3 ml of DMEM-2.5. Cells were incubated in a CO 2 incubator at 37°C for three days. On the third day the cells were harvested, pelleted by centrifugation, and resuspended in 300 ⁇ l PBS. Viras was released from the cells by three cycles of freeze/ thaw as described above. One hundred microliters of crude viras stock was fransferred to a 1.5 ml tube, an equal volume of melted 2% low melting point agarose was added, and the virus/agarose mixture was fransferred into a pulsed field gel sample block.
  • agar worms When the agar worms were solidified they were removed from the sample block and cut into three equal sections. All three sections were fransferred to the same 1.5 ml tube, and 250 ⁇ l of 0.5M EDTA, 1% Sarkosyl, 0.5mg/ml
  • Proteinase K was added.
  • the worms were incubated in this solution at 37°C for 24 hours.
  • the worms were washed several times in 500 ⁇ l 0.5X TBE buffer, and one section of each worm was fransferred to a well of a 1% low melting point agarose gel. After the worms were added the wells were sealed by adding additional melted 1% low melting point agarose. This gel was then electorphoresed in a Bio-Rad pulsed field gel electrophoresis apparatus at
  • Vaccinia DNA was purified from the agarose using ⁇ -Agarase (Gibco) following the recomendations of the manufacturer. Purified vaccinia DNA was resuspended in 50 ⁇ l ddH 2 O. One microliter of each DNA. stock was used as the template for a Polymerase Chain Reaction (PCR) using vaccinia TK specific primers MM428 and MM430 (which flank the site of insertion) and Klentaq Polymerase (Clontech) following the recommendations of the manufacturer in a 20 ⁇ l final volume. Reaction conditions included an initial denaturation step at 95°C for 5 minutes, followed by 30 cycles of: 94°C 30 seconds, 55°C 30 seconds, 68°C 3 minutes.
  • PCR Polymerase Chain Reaction
  • the tri-molecular recombination method as herein disclosed may be used with other virases such as mammalian virases including vaccinia and herpes virases.
  • virases such as mammalian virases including vaccinia and herpes virases.
  • two viral arms which have no homology are produced.
  • the only way that the viral arms can be linked is by bridging through homologous sequences that flank the insert in a fransfer vector such as a plasmid.
  • a fransfer vector such as a plasmid
  • Libraries constructed in vaccinia and other mammalian viruses by the tri-molecular recombination method ofthe present invention may have similar advantages to those described here for vaccinia virus and its use in identifying target antigens in the CTL screening system of the invention. Similar advantages are expected for DNA libraries constructed in vaccinia or other mammalian virases when carrying out more complex assays in eukaryotic cells. Such assays include but are not limited to screening for DNA encoding receptors and ligands of eukaryotic cells.
  • a model system was assayed to determine the level of enrichment that can be obtained through a procedure that selects for DNA recombinants that encode the target epitopes of tumor specific cytotoxic T cells.
  • a specific vaccinia recombinant that encodes a well characterized ovalbumin peptide (SHNFEKL) (SEQ ID NO:26) was diluted with non-recombinant virus so that it constituted either 0.2%, 0.01%, or 0.001% of viral pfu.
  • This ovalbumin peptide is known to be processed and presented to specific CTL in association with the murine class I MHC molecule H-2K .
  • MC57G cells do not themselves express ovalbumin peptide, but do express H-2K , which allows them to associate with and present ovalbumin peptide to the T cells.
  • ovalbumin peptide-specific CTL derived by repeated in vitro stimulation of ovalbumin primed splenic T cells with the immunodominant ovalbumin SHNFEKL peptide, were added for 30 min.
  • Virus extracted from each cell population was tifred for the frequency of ovalbumin recombinant viral pfu. Virus extracted from floating cells was then used as input to another enrichment cycle with fresh adherent MC57G cells and ovalbumin peptide-specific CTL. It was observed that, following enrichment of Wova to greater than 10% of total virus, further enrichment of the recombinant viras was accelerated if the m.o.i. in succeeding cycles was reduced from 1 to 0.1. The results, presented in Table 12, demonstrate marked enrichment of Wova recombinant virus from an initial concenfration of 0.2% to 49% or from 0.01% to 39% in 5 enrichment cycles and from 0.001% to 18% in 6 enrichment cycles.
  • the above-described selection method for isolating DNA clones that encode target epitopes of specific cytotoxic T cells from a viral library is far more efficient than existing methods for accomplishing this same goal.
  • the most widely employed method requires fransfection of numerous small pools of recombinant plasmids into separate target populations in order to assay T cell stimulation by a minor component of some pool. Because this requires screening out many negative plasmid pools, it is a far more labor intensive procedure than the positive selection method described herein.
  • the method described here can detect positive DNA clones that occur at a much lower frequency than would otherwise be possible.
  • the design principle of this strategy can be directly extended to screening and selection of DNA clones with specific antibodies as well as with CTL.
  • a Deregulated Ribosomal Protein L3 Gene Encodes a Shared Murine Tumor
  • the ribosomal protein described may be representative of a class of shared tumor antigens that arise as a result of deregulated expression of a self-protein without compromising immune tolerance to normal tissues. Such antigens would be suitable for immunotherapy of cancer in vital organs.
  • RNA Total RNA Isolation KitTM (5 Prime 3 Prime, Inc., Boulder, CO). Poly A+ mRNA was isolated from the total RNA using DynabeadsTM (Dynal, Lake Success, NY). Two micrograms of poly A+ mRNA was converted to double stranded cDNA using the Great Lengths cDNA Synthesis KitTM (Clontech, Palo Alto, CA). The double sfranded cDNA was then inserted in vaccima virus vector v7.5/tk (Rosenwald, I. B., Cancer Lett. 102:113-123 (1996)).
  • mice were immunized infraperitoneally with

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Wood Science & Technology (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

L'invention concerne une méthode à haut rendement qui permet d'introduire un ADN dans des virus à ADN linéaire, tels que la poxvirose, une méthode qui permet de produire des bibliothèques dans des virus à ADN linéaire, tels que la poxvirose, et des méthodes qui permettent de rechercher des polynucléotides d'intérêt par criblage en fonction de la non-viabilité cellulaire ou d'autres phénotypes de cellules eucaryotes, notamment des cellules mammaliennes. L'invention concerne également des polynucléotides promoteurs C35 et des dérivés fonctionnels de ces derniers.
PCT/US2003/033557 2002-10-22 2003-10-22 Methodes permettant de produire une bibliotheque et methodes permettant de selectionner des polynucleotides d'interet WO2004037993A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003294234A AU2003294234A1 (en) 2002-10-22 2003-10-22 Methods of producing a library and methods of selecting polynucleotides of interest

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US10/277,161 2002-10-22
US10/277,161 US20030194696A1 (en) 2000-03-28 2002-10-22 Methods of producing a library and methods of selecting polynucleotides of interest
US50677103P 2003-09-30 2003-09-30
US60/506,771 2003-09-30

Publications (2)

Publication Number Publication Date
WO2004037993A2 true WO2004037993A2 (fr) 2004-05-06
WO2004037993A3 WO2004037993A3 (fr) 2005-09-15

Family

ID=32179469

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/033557 WO2004037993A2 (fr) 2002-10-22 2003-10-22 Methodes permettant de produire une bibliotheque et methodes permettant de selectionner des polynucleotides d'interet

Country Status (2)

Country Link
AU (1) AU2003294234A1 (fr)
WO (1) WO2004037993A2 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7067251B2 (en) 2000-03-28 2006-06-27 University Of Rochester Methods of directly selecting cells expressing inserts of interest
EP2029172A2 (fr) * 2006-06-22 2009-03-04 Vaccinex, Inc. Anticorps anti-c35 pour le traitement du cancer
US7858559B2 (en) 2000-11-17 2010-12-28 University Of Rochester In vitro methods of producing and identifying immunoglobulin molecules in eukaryotic cells

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030124128A1 (en) * 2001-06-21 2003-07-03 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention, and therapy of breast cancer

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030124128A1 (en) * 2001-06-21 2003-07-03 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention, and therapy of breast cancer

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DATABASE GENBANK [Online] 29 July 1996 HILLER ET AL., XP002987736 Database accession no. AA010615 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7067251B2 (en) 2000-03-28 2006-06-27 University Of Rochester Methods of directly selecting cells expressing inserts of interest
US7858559B2 (en) 2000-11-17 2010-12-28 University Of Rochester In vitro methods of producing and identifying immunoglobulin molecules in eukaryotic cells
US8741810B2 (en) 2000-11-17 2014-06-03 University Of Rochester In vitro methods of producing and identifying immunoglobulin molecules in eukaryotic cells
EP2029172A2 (fr) * 2006-06-22 2009-03-04 Vaccinex, Inc. Anticorps anti-c35 pour le traitement du cancer
EP2029172A4 (fr) * 2006-06-22 2010-07-28 Vaccinex Inc Anticorps anti-c35 pour le traitement du cancer

Also Published As

Publication number Publication date
AU2003294234A8 (en) 2004-05-13
WO2004037993A3 (fr) 2005-09-15
AU2003294234A1 (en) 2004-05-13

Similar Documents

Publication Publication Date Title
US7067251B2 (en) Methods of directly selecting cells expressing inserts of interest
US6800442B2 (en) Methods of selecting polynucleotides encoding antigens
AU776865B2 (en) T cells specific for target antigens and methods and vaccines based thereon
US5885827A (en) Eukaryotic high rate mutagenesis system
JP4368196B2 (ja) 真核細胞において免疫グロブリン分子を製造および同定するインビトロにおける方法
US20030104402A1 (en) Methods of producing or identifying intrabodies in eukaryotic cells
US20230130799A1 (en) Compositions and methods for making antibodies based on use of expression-enhancing loci
KR20230074515A (ko) 개선된 기능성 및 지속성을 갖는 붕괴된 레그나제-1 및/또는 tgfbrii를 지니는 유전자 조작된 t 세포
KR20180134894A (ko) 발현 강화 유전자좌의 사용에 기초하여 항체를 만들기 위한 조성물 및 방법
KR19990077015A (ko) 진핵 세포 발현 시스템을 위한 발현 증강 서열 요소 (ease)
KR102449406B1 (ko) 백시니아 바이러스/진핵 세포에서 폴리뉴클레오티드 라이브러리를 생산하기 위한 개선된 방법
CN108474005B (zh) 转位子系统、试剂盒及其用途
CA2272823A1 (fr) Procede pour identifier des genes codant de nouvelles proteines secretees ou associees a des membranes
CN114502731A (zh) 基于转座子的免疫细胞的修饰
KR20210089707A (ko) 항-liv1 면역세포 암 치료법
US20020192675A1 (en) Methods of identifying regulator molecules
US20030194696A1 (en) Methods of producing a library and methods of selecting polynucleotides of interest
WO2004037993A2 (fr) Methodes permettant de produire une bibliotheque et methodes permettant de selectionner des polynucleotides d'interet
KR101646597B1 (ko) Pten 융합 단백질 및 이의 용도
JP4732338B2 (ja) Es細胞の変異方法およびシステム
Sainova et al. Development of Animal Models, Presenting Appropriate Analogues to Respective Human Systems
KR20010032473A (ko) 시그널 서열 트래핑법
EA044725B1 (ru) Композиции и способы получения антител, основанные на применении локусов, обеспечивающих повышенную экспрессию
Bhattacharya NF-κB-mediated regulation of immunoglobulin isotype switching and apoptosis
CA2206994A1 (fr) Genes et produits geniques pour le diagnostic de lesions nerveuses degeneratives

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR 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 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: A2

Designated state(s): GH GM KE LS MW MZ 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 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
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)