WO2004046330A2 - Methods of generating high-production of antibodies from hybridomas created by in vitro immunization - Google Patents

Methods of generating high-production of antibodies from hybridomas created by in vitro immunization Download PDF

Info

Publication number
WO2004046330A2
WO2004046330A2 PCT/US2003/036702 US0336702W WO2004046330A2 WO 2004046330 A2 WO2004046330 A2 WO 2004046330A2 US 0336702 W US0336702 W US 0336702W WO 2004046330 A2 WO2004046330 A2 WO 2004046330A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
antibodies
producing
hybridoma
mismatch repair
Prior art date
Application number
PCT/US2003/036702
Other languages
French (fr)
Other versions
WO2004046330A3 (en
Inventor
Luigi Grasso
Shaohong Liang
Nicholas E. Nicolaides
Philip M. Sass
Original Assignee
Morphotek, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Morphotek, Inc. filed Critical Morphotek, Inc.
Priority to JP2004570421A priority Critical patent/JP4555089B2/en
Priority to DE60329526T priority patent/DE60329526D1/en
Priority to AU2003295576A priority patent/AU2003295576B2/en
Priority to AT03786774T priority patent/ATE444359T1/en
Priority to CA2506127A priority patent/CA2506127C/en
Priority to EP20030786774 priority patent/EP1572971B1/en
Publication of WO2004046330A2 publication Critical patent/WO2004046330A2/en
Publication of WO2004046330A3 publication Critical patent/WO2004046330A3/en

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
    • C12N15/01Preparation of mutants without inserting foreign genetic material therein; Screening processes therefor
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • C07K16/1282Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria from Clostridium (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/243Colony Stimulating Factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • 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
    • C12N15/102Mutagenizing nucleic acids
    • C12N15/1024In vivo mutagenesis using high mutation rate "mutator" host strains by inserting genetic material, e.g. encoding an error prone polymerase, disrupting a gene for mismatch repair
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation

Definitions

  • the invention relates to the generation of hybridoma cells that produce high-affinity antibodies in high titers. More specifically, the invention relates to the use of an in vitro ii riunization method in conjunction with hybridoma technology using dominant negative mismatch repair genes or chemical inhibitors of mismatch repair to produce high titers of antigen specific antibodies of the IgG subclass, that bind to the antigen with high affinity.
  • antibodies to block the activity of foreign and/or endogenous polypeptides provides an effective and selective strategy for treating the underlying cause of disease.
  • MAb monoclonal antibodies
  • MAb monoclonal antibodies
  • the FDA approved ReoPro Gibcose, (1996) Nat. Biotechnol 14:1216-1217), an anti-platelet MAb from Centocor; Herceptin (Weiner, (1999) Semin. Oncol. 26:43-51), an anti-Her2/neu MAb from Genentech; and Synagis (SaezLlorens, et al. (1998) Pediat. Infect Dis. J. 17:787-791), an anti- respiratory syncytial virus MAb produced by Medimmune.
  • Standard methods for generating MAbs against candidate protein targets are known by those skilled in the art. Briefly, rodents such as mice or rats are injected with a purified antigen in the presence of adjuvant to generate an immune response (Shield, et al. (1996) Am. J. Kidney Dis. 27: 855-864). Rodents with positive immune sera are sacrificed and splenocytes are isolated. Isolated splenocytes are fused to melanomas to produce immortalized cell lines that are then screened for antibody production. Positive lines are isolated and characterized for antibody production.
  • rodent MAbs as human therapeutic agents were confounded by the fact that human anti-rodent antibody (HARA) responses occurred in a significant number of patients treated with the rodent-derived antibody (Khazaeli, et al, (1994) Immunother. 15:42-52).
  • HAA human anti-rodent antibody
  • CDRs complementarity determining regions
  • Ig immunoglobulin subunits making up the antigen binding domain
  • HAb rodent-derived MAbs
  • Another problem that exists in antibody engineering is the generation of stable, high yielding producer cell lines that is required for manufacturing of the molecule for clinical materials.
  • Several strategies have been adopted in standard practice by those skilled in the art to circumvent this problem.
  • One method is the use of Chinese Hamster Ovary (CHO) cells transfected with exogenous Ig fusion genes containing the grafted human light and heavy chains to produce whole antibodies or single chain antibodies, which are a chimeric molecule containing both light and heavy chains that form an antigen-binding polypeptide (Reff, M. E. (1993) Curr. Opin. Biotechnol. 4:573-576).
  • Another method employs the use of human lymphocytes derived from transgenic mice containing a human grafted immune system or transgenic mice containing a human Ig gene repertoire.
  • Yet another method employs the use of monkeys to produce primate MAbs, which have been reported to lack a human anti-monkey response (Neuberger and Gruggermann (1997) Nature 386:25-26).
  • the generation of a cell line that is capable of generating sufficient amounts of high affinity antibody poses a major limitation for producing sufficient materials for clinical studies. Because of these limitations, the utility of other recombinant systems such as plants are currently being explored as systems that will lead to the stable, high-level production of humanized antibodies (Fiedler and Conrad (1995) Bio/Technology 13:1090-1093).
  • the invention described herein is directed to the use of random genetic mutation throughout an antibody structure in vitro by blocking the endogenous mismatch repair (MMR) activity of a host cell producing immunoglobulins that encode biochemically active antibodies.
  • MMR mismatch repair
  • the invention also relates to methods for repeated in vitro genetic alterations and selection for antibodies with enhanced binding and pharmacokinetic profiles.
  • the invention described herein is further directed to the creation of genetically altered cell hosts with increased antibody production via the blockade of MMR.
  • the invention facilitates the generation of high affinity antibodies and the production of cell lines with elevated levels of antibody production derived from hybridoma cells.
  • the invention described herein provides methods for generating antigen-specific monoclonal antibodies (mAbs). Other advantages of the present invention are described in the examples and figures described herein.
  • the invention provides methods for producing hybridoma cells producing high-affinity antibodies from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobuliii-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells express a dominant negative allele of a mismatch repair gene; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; thereby producing hybridoma cells producing high-affinity antibodies.
  • the dominant negative allele of a mismatch repair gene comprises a truncation mutation of the PMS2 gene (e.g., a PMS2-134 gene).
  • antibodies are screened using an ELISA-based assay or other assays that can measure antibody-antigen binding.
  • the screening assays screen for hypermutated hybridomas that produce higher affinity antibodies than those produced by the parental hybridomas.
  • the screening assays screen for hypermutated hybridomas that produce antibodies in higher titers than the parental hybridomas.
  • the method further comprises inactivation of the dominant negative allele of the mismatch repair gene, thereby stabilizing the genome of said hypermutated hybridoma.
  • the dominant negative mismatch repair gene is introduced into the hybridoma cell after the fusion of said myeloma with the immunoglobulin-producing cells. In other embodiments, the dominant negative mismatch repair gene is introduced into the myeloma cell prior to the fusion with the immunoglobulin- producing cells.
  • the invention also comprises antibodies produced by the hybridoma cells.
  • the invention also comprises methods for producing hybridoma cells that produce high titers of antibodies from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor blood cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells express a dominant negative allele of a mismatch repair gene; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen of the hypermutated hybridoma cells for antigen-specific antibodies produced in higher titers than that produced by the parental hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce higher titers of antibodies than that produced by the parental hybridoma cells.
  • the dominant negative allele of a mismatch repair gene comprises a truncation mutation of the PMS2 gene (e.g., a PMS2-134 gene).
  • antibodies are screened using an ELISA-based assay.
  • the screening assays screen for hypermutated hybridomas that produce higher affinity antibodies than those produced by the parental hybridomas.
  • the screening assays screen for hypermutated hybridomas that produce antibodies in higher titers than the parental hybridomas.
  • the method further comprising inactivation of the dominant negative allele of the mismatch repair gene, thereby stabilizing the genome of said hypermutated hybridoma.
  • the dominant negative mismatch repair gene is introduced into the hybridoma cell after the fusion of said myeloma with the immunoglobulin-producing cells. In other embodiments, the dominant negative mismatch repair gene is introduced into the myeloma cell prior to the fusion with the immunoglobulin- producing cells.
  • the dominant negative allele of the mismatch repair gene is subsequently inactivated in order to restabilize the genome of the cell.
  • the dominant negative allele of the mismatch repair gene may be introduced into the myeloma cell prior to fusion with the immunoglobulin producing cells.
  • the resulting hybridoma cells express the same dominant negative allele of the mismatch repair gene as the myeloma cells.
  • the dominant negative allele of the mismatch repair gene may be introduced into the hybridoma cells.
  • the invention also comprises antibodies produced by the hybridoma cells.
  • the invention further provides recombinant myeloma cells comprising a polynucleotide sequence encoding a dominant negative mismatch repair protein.
  • the dominant negative mismatch repair protein may be a dominant negative form of, for example, aPMS2, PMS1, PMSR3, PMSR2, PMSR6, MLH1, GTBP, MSH3, MSH2, MLH3, or MSH1, and PMSR proteins encoded by homologs of the PMSR genes as described in Nicolaides et al. (1995) Genomics 30:195-206 and Horii et al. (1994) Biochem. Biophys. Res. Commun. 204:1257-1264.
  • the recombinant myeloma cell expresses a polynucleotide encoding a dominant negative allele of a PMS2 gene (e.g., a truncation mutation of the PMS2 gene, such as the PMS2-134 gene).
  • the recombinant myeloma cell is a human cell.
  • the recombinant myeloma cell does not express immunoglobulin genes and/or Epstein-Barr virus.
  • the myeloma cells are HAT sensitive.
  • the invention also provides a method for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing said immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for binding of antibodies produced from said hybridoma cells to antigen; (d) cloning immunoglobulin genes from said hybridoma into a mammalian expression cell, wherein said mammalian expression cell expresses a dominant negative allele of a mismatch repair gene; (e) performing a screen for mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from said hybridoma cells; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin
  • the dominant negative allele of a mismatch repair gene is introduced into said mammalian expression cell prior to introduction of the immunoglobulin genes. In other embodiments, the dominant negative allele of a mismatch repair gene is introduced into said mammalian expression cell after introduction of said immunoglobulin genes. In other embodiments, the dominant negative allele of a mismatch repair gene is introduced into the mammalian expression cell with the immunoglobulin genes simultaneously.
  • the invention also comprises antibodies produced by the mammalian expression cells.
  • the invention also provides a method for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing said immunoglobulin-producing cells with myeloma cells to form hybridoma cells, wherein said hybridoma cells express a dominant negative allele of a mismatch repair gene; (c) incubating said parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from said hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for said antigen than antibodies produced by said parental hybridoma cells; (f) cloning immunoglobulin
  • the dominant negative allele of a mismatch repair gene is present in the myeloma cell prior to cell fusion. In other embodiments, the dominant negative allele of the mismatch repair gene is introduced into the hybridoma cell after cell fusion.
  • the invention also comprises antibodies produced by the mammalian expression cells.
  • the invention also provides a method for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for binding of antibodies produced from the hybridoma cells to antigen; (d) cloning immunoglobulin genes from the hybridoma into a parental mammalian expression cell, wherein the mammalian expression cell expresses a dominant negative allele of a mismatch repair gene; (e) incubating the parental mammalian expression cell to allow for mutagenesis, thereby forming hypermutated mammalian expression cells; (f) performing a screen of hypermutable mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from the
  • the dominant negative allele of a mismatch repair gene is introduced into said mammalian expression cell prior to introduction of the immunoglobulin genes. In other embodiments, the dominant negative allele of a mismatch repair gene is introduced into said mammalian expression cell after introduction of said immunoglobulin genes. In other embodiments, the dominant negative allele of a mismatch repair gene is introduced into the mammalian expression cell with the immunoglobulin genes simultaneously.
  • the invention also provides antibodies produced by the mammalian expression cells.
  • the invention also provides recombinant, hypermutable mammalian expression cells comprising a polynucleotide sequence encoding a dominant negative mismatch repair protein.
  • the mismatch repair gene may be a dominant negative mismatch repair gene, including, but not limited to a dominant negative form of PMS2, PMS1, PMSR3, PMSR2, PMSR6, MLH1, GTBP, MSH3, MSH2, MLH3, or MSH1, and homologs of PMSR genes as described in Nicolaides et al (1995) Genomics 30:195-206 and Horii et al. (1994) Biochem. Biophys. Res. Commun. 204:1257-1264.
  • a non-limiting example includes a dominant negative truncation mutant of PMS2 (e.g., a.PMS2-134 gene).
  • the invention also provides methods for producing hybridoma cells producing high- affinity antibodies from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising ⁇ rimunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells; (c) incubating the parental hybridoma cells in the presence of at least one chemical inhibitor of mismatch repair, thereby forming hypermutated hybridoma cells; (d) performing a screen for antigen binding for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for the antigen than antibodies produced by said parental hybridoma cells; thereby producing hybridoma cells producing high-affinity antibodies.
  • the invention also comprises antibodies produced by the hybridoma cells.
  • the invention also provides methods for producing hybridoma cells that produce high titers of antibodies from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells; (c) incubating the parental hybridoma cells in the presence of at least one chemical inhibitor of mismatch repair, thereby forming hypermutated hybridoma cells; (d) performing a screen of the hypermutated hybridoma cells for antigen-specific antibodies produced in higher titers than that produced by the parental hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce higher titers of antibodies than that produced by said parental hybridoma cells; thereby producing hybridoma cells producing high titers of antibodies.
  • the hypermutated hybridoma cells also are screened for the production of higher titers of antibodies than that produced by the parental hybridomas.
  • the screening may be using an ELISA-based assay, or any other means to measure antibody-antigen binding.
  • the invention also comprises antibodies produced by the hybridoma cells.
  • the invention also provides methods for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for antigen binding of antibodies produced from the hybridoma cells; (d) cloning immunoglobulin genes from the hybridoma cells into a mammalian expression cell; (e) incubating the mammalian expression cell in the presence of at least one chemical inhibitor of mismatch repair; (f) performing a screen for mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from the hybridoma cells; thereby producing mammalian expression cells that produce high titers of
  • the method may further comprise the removal of the chemical inhibitor from the hypermutated mammalian expression cells, thereby stabilizing the genome of said hypermutated mammalian expression cells.
  • the invention also comprises antibodies produced by the mammalian expression cells [0042]
  • the invention also provides methods for producing mammalian expression cells that produce high titers of high affinity antibodies to a selected antigen from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) incubating the hybridoma cells in the presence of at least one chemical inhibitor of mismatch repair to form hypermutated hybridoma cells; (d) performing a screen for antigen binding for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that
  • the parental mammalian expression cell is further incubated in the presence of at least one chemical inhibitor of mismatch repair, thereby forming a hypermutated mammalian expression cell; and the hypermutated mammalian expression cells are screened for higher production of antibodies than that of the parental mammalian expression cells.
  • the method may further comprise the removal of the chemical inhibitor from the hypermutated hybridoma and/or hypermutated mammalian expression cells, thereby stabilizing the genome of said hypermutated hybridoma cells and/or hypermutated mammalian expression cells.
  • the invention also comprises antibodies produced by the mammalian expression cells.
  • the invention also provides a method for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing said immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for binding of antibodies produced from said hybridoma cells to antigen; (d) cloning immunoglobulin genes from said hybridoma into a mammalian expression cell; (e) incubating said mammalian expression cell in the presence of at least one chemical inhibitor of mismatch repair, thereby forming a hypermutated mammalian expression cell; (f) performing a screen for hypermutated mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from said parental mammalian expression cells; and (g) performing a second screen for hypen
  • the method may further comprise the removal of the chemical inhibitor from the hypermutated hybridoma and/or hypermutated mammalian expression cells, thereby stabilizing the genome of said hypermutated hybridoma cells and/or hypermutated mammalian expression cells.
  • the invention also comprises antibodies produced by the mammalian expression cells.
  • the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro, wherein the donor cells are derived from a donor that is naturally deficient in mismatch repair; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma
  • the method may further comprise introducing a wild-type gene for mismatch repair into said selected hypermutated hybridoma cell to complement the mismatch repair deficiency, thereby restabilizing the genome of said selected hypermutated hybridoma cell.
  • the invention also comprises antibodies produced by the hybridoma cells.
  • the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells, wherein the myeloma cells are naturally deficient in mismatch repair, thereby forming parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby fonning hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; thereby producing hybridoma cells producing high-affinity antibodies.
  • the method may further comprise introducing a wild-type gene for mismatch repair into said selected hypermutated hybridoma cell to complement the mismatch repair deficiency, thereby restabilizing the genome of said selected hypermutated hybridoma cell.
  • the invention also comprises antibodies produced by the hybridoma cells.
  • the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells in high titers comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro, wherein the donor cells are derived from a donor that is naturally deficient in mismatch repair; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; (f) performing a second screen for hypermutated
  • the method may further comprise introducing a wild-type gene for mismatch repair into said selected hypermutated hybridoma cell to complement the mismatch repair deficiency, thereby restabilizing the genome of said selected hypermutated hybridoma cell.
  • the invention also comprises antibodies produced by the hybridoma cells.
  • the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells in high titers comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells, wherein the myeloma cells are naturally deficient in mismatch repair, thereby forming parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; (f) performing a second screen for hypermutated
  • the method may further comprise introducing a wild-type gene for mismatch repair into said selected hypermutated hybridoma cell to complement the mismatch repair deficiency, thereby restabilizing the genome of said selected hypermutated hybridoma cell.
  • the invention also comprises antibodies produced by the hybridoma cells.
  • the invention comprises a method for producing mammalian expression cells that produce high-affinity antibodies in high titers from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro, wherein the donor cells are derived from a donor that is naturally deficient in mismatch repair; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; (f) cloning immunoglobulin
  • the parental mammalian expression cell is further incubated in the presence of at least one chemical inhibitor of mismatch repair, thereby forming a hypermutated mammalian expression cell; and the hypermutated mammalian expression cells are screened for higher production of antibodies than that of the parental mammalian expression cells.
  • the method may further comprise the removal of the chemical inhibitor from the hypermutated mammalian expression cells, thereby stabilizing the genome of said hypermutated mammalian expression cells.
  • the invention also comprises antibodies produced by the mammalian expression cells.
  • the invention comprises a method for producing mammalian expression cells that produce high-affinity antibodies in high titer from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells, wherein the myeloma cells are naturally deficient in mismatch repair, thereby forming parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; and (f) cloning immunoglobulin
  • the parental mammalian expression cell is further incubated in the presence of at least one chemical inhibitor of mismatch repair, thereby forming a hypermutated mammalian expression cell; and the hypermutated mammalian expression cells are screened for higher production of antibodies than that of the parental mammalian expression cells.
  • the method may further comprise the removal of the chemical inhibitor from the hypermutated mammalian expression cells, thereby stabilizing the genome of said hypermutated mammalian expression cells.
  • the invention also comprises antibodies produced by the hybridoma cells.
  • the immunoglobulin-producing cells are mammalian cells, including but not limited to, mouse cells, rat cells, goat cells, cow cells, horse cells, dog cells, cat cells, rabbit cells, bird cells, monkey cells and human cells. In preferred embodiments, the cells are human cells.
  • the dominant negative allele of a mismatch repair gene is a dominant negative allele of PMS2, PMS1, PMSR3, PMSR2, PMSR6, MLH1, GTBP, MSH3, MSH2, MLH3, or MSH1, and homologs of PMSR genes as described in Nicolaides et al. (1995) Genomics 30:195-206 and Horii et al (1994) Biochem. Biophys. Res. Commun. 204:1257-1264.
  • the mismatch repair genes are not limit to these examples.
  • the immunogenic antigen is conjugated to a mitogenic polypeptide comprising at least a portion of a polypeptide including, but not limited to tetanus toxoid, ovalbumin, bovine serum albumen, thyroglobulin, diptheria toxoid, BCG, and cholera toxin.
  • the antigen is generated by denaturing the mature protein.
  • the antibodies produced have an affinity of at least about 1 x 10 7 M "1 . In other embodiments, the antibodies have an affinity of at least about 1 x 10 8 M "1 . In other embodiments, the antibodies have an affinity of at least about 1 x 10 9 M "1 . In other embodiments, the antibodies have an affinity of at least about 1 x 10 10 M '1 . In other embodiments, the antibodies have an affinity of at least about 1 x 10 11 M "1 . In other embodiments, the antibodies have an affinity of at least about 1 x 10 12 M "1 . In other embodiments, the antibodies have an affinity of at least about 1 x 10 13 M "1 .
  • the antibodies have an affinity of at least about 1 x 10 14 M "1 .
  • the antibodies are produced in a higher titer than the parental cell lines, such as in an amount of at least about 1.5 fold higher than the parental cell line.
  • the titer is at least about 1.5-3 fold higher than the parental cell line.
  • the titer is at least about 3-5 fold higher than the parental cell line.
  • the titer is at least about 5-7 fold higher than the parental cell line.
  • the titer is at least about 7-9 fold higher than the parental cell line.
  • the titer is at least about 9-10 fold higher than the parental cell line.
  • mutation rates are further enhanced by incubating the hybridoma cells with a chemical mutagen, such as, but not limited to N-ethyl-N-nitrosourea, N-methyl-N-nitrosourea, procarbazine hydrochloride, chlorarnbucil, cyclophosphamide, methyl methanesulfonate, ethyl methanesulfonate, diethyl sulfate, acrylamide monomer, triethylene melamin, melphalan, nitrogen mustard, vincristine, dimethylnitrosamine, N-methyl-N'-nitro-nitrosoguanidine, 7,12 dimethylbenz (a) anthracene, ethylene oxide, hexamethylphosphoramide, and bisulfan.
  • a chemical mutagen such as, but not limited to N-ethyl-N-nitrosourea, N-methyl-N-nitrosourea, procarbazine hydrochloride, chlorarnbucil
  • the chemical inhibitors of mismatch repair used in certain embodiments of the methods of the invention include, but are not limited to, at least one of an anthracene, an ATPase inhibitor, a nuclease inhibitor, an RNA interference molecule, a polymerase inhibitor and an antisense oligonucleotide that specifically hybridizes to a nucleotide encoding a mismatch repair protein.
  • the chemical inhibitor is an anthracene having the formula:
  • Ri-Rio are independently hydrogen, hydroxyl, amino, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S- alkenyl, N-alkenyl, O-alkynyl, S-alkynyl, N-alkynyl, aryl, substituted aryl, aryloxy, substituted aryloxy, heteroaryl, substituted heteroaryl, aralkyloxy, arylalkyl, alkylaryl, alkylaryloxy, arylsulfonyl, alkylsulfonyl, alkoxycarbonyl, aryloxycarbonyl, guanidino, carboxy, an alcohol, an amino acid, sulfonate, alkyl sulfonate, CN, NO 2 , an aldehyde group
  • R 5 and R ⁇ are hydrogen.
  • Ri-Rio are independently hydrogen, hydroxyl, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, phenyl, tolyl, hydroxymethyl, hydroxypropyl, or hydroxybutyl.
  • Non-limiting examples of the anthracenes include 1,2-dimethylanthracene, 9,10-dimethylanthracene, 7,8- dimethylanthracene, 9,10-duphenylanthracene, 9,10-dihydroxymethylanthracene, 9- hydroxymethyl- 10-methylanthracene, dimethylanthracene- 1 ,2-diol, 9-hydroxymethyl- 10- methylanthracene-l,2-diol, 9-hydroxymethyl- 10-methylanthracene-3,4-diol, and 9,10-di-m- tolylanthracene.
  • the chemical inhibitor may be introduced into the growth medium of the cells.
  • the chemical inhibitor may be withdrawn from the hypermutated hybridoma cells in order to re-stabilize the genome of the cells.
  • the invention also comprises a method for in vitro production of antigen-specific immunoglobulin-producing cells comprising: (a) isolating donor cells from an animal; (b) treating said cells with L-leucyl-L-leucine methy ester hydrobromide; (c) incubating said donor cells with an immunogenic antigen in vitro, at 25-37°C, 5-10% CO 2 , in medium supplemented with 5-15% serum, and a growth promoting cytokine for 4 days; (d) washing said cells in medium; and (e) culturing said cells in medium supplemented with 5-15% serum an additional 8 days; thereby stimulating the production of antigen-specific immunoglobulin- producing cells.
  • the immunoglobulin-producing cells are human cells.
  • the immunogenic antigen is conjugated to a mitogenic polypeptide comprising at least a portion of a polypeptide including, but not limited to tetanus toxoid, ovalbumin, bovine serum albumen, thyroglobulin, diptheria toxoid, BCG, and cholera toxin.
  • the antigen is generated by denaturing the mature protein.
  • the antibodies produced have an
  • the antibodies have an affinity of at least about 1 x 10 8 M “1 . In other embodiments, the antibodies have an affinity of at least about 1 x 10 9 M “1 . In other embodiments, the antibodies have an affinity of at least about 1 x 10 10 M “1 . In other embodiments, the antibodies have an affinity of at least about 1 x 10 11 M “1 . In other embodiments, the antibodies have an affinity of at least about 1 x 10 12 M “1 . In other embodiments, the antibodies have an affinity of at least about 1 x 10 13 M “1 . In other embodiments, the antibodies have an affinity of at least about 1 x 10 14 M “1 .
  • the antibodies are produced in a higher titer than the parental cell lines, such as in an amount of at least about 1.5 fold higher than the parental cell line.
  • the titer is at least about 1.5-3 fold higher than the parental cell line.
  • the titer is at least about 3-5 fold higher than the parental cell line.
  • the titer is at least about 5-7 fold higher than the parental cell line.
  • the titer is at least about 7-9 fold higher than the parental cell line.
  • the titer is at least about 9-10 fold higher than the parental cell line.
  • mutation rates are further enhanced by incubating the hybridoma cells and/or mammalian expression cells with a chemical mutagen, such as, but not limited to N-ethyl-N-nitrosourea, N-methyl-N-nitrosourea, procarbazine hydrochloride, chlorambucil, cyclophosphamide, methyl methanesulfonate, ethyl methanesulfonate, diethyl sulfate, acrylamide monomer, triethylene melamin, melphalan, nitrogen mustard, vincristine, dimethylnitrosamine, N-methyl-N'-nitiO-nitrosoguanidine, 7,12 dimethylbenz (a) anthracene, ethylene oxide, hexamethylphosphoramide, and bisulfan.
  • a chemical mutagen such as, but not limited to N-ethyl-N-nitrosourea, N-methyl-N-nitrosourea, procarbazine
  • the mammalian expression cells used in the methods of the invention may include, but are not limited to, Chinese Hamster Ovary, baby hamster kidney cells, human embryonic kidney line 293, normal dog kidney cell lines, normal cat kidney cell lines, monkey kidney cells, African green monkey kidney cells, COS cells, and non-tumorigenic mouse myoblast G8 cells, fibroblast cell lines, myeloma cell lines, mouse NTH/3T3 cells, LMTK 31 cells, mouse sertoli cells, human cervical carcinoma cells, buffalo rat liver cells, human lung cells, human liver cells, mouse mammary tumor cells, TRI cells, MRC 5 cells, and FS4 cells. [0084] These and other embodiments are described more fully in the next section and include certain non-limiting examples.
  • Figure 1 shows the immune response of PBMCs to antigen stimulation.
  • PBMCs were cultured in the presence or absence of TT for 4 days then washed with medium and cultured in the presence or absence of TT for an additional eight days. Culture supernates were collected and tested for the presence of antibody reactive to TT. Antibodies bound to TT pre-coated on the solid phase were detected with HRP-labeled goat anti-human IgG, or HRP-labeled goat anti-human IgM.
  • Fig. 2A shows reactivity of donor serum to TT by detection of donor anti-TT IgG.
  • Fig. 2B shows reactivity of donor serum to TT by detection of donor anti-TT IgM.
  • Figure 3 shows the frequency of the anti-TT response of PBMCs upon in vitro immunization with TT, or with TT in combination with IL-2, or CD40L.
  • Figure 4 shows the intensity of the response of PBMCs upon in vitro immunization with TT, or with TT in combination with IL-2, or CD40L.
  • Figure 5 shows the response of hybridomas expressing anti-TT antibodies.
  • Fig. 6A shows the reactivity of unstimulated PBMCs to EGFR.
  • Fig. 6B shows the reactivity of PBMCs to EGFR after immunization with EGFR-TT.
  • Fig. 6C shows the reactivity of unstimulated PBMCs to EGFR-TT.
  • Fig. 6D shows the reactivity of PBMCs to
  • Figure 7 shows the response of hybridomas expressing antibodies against human
  • Antibodies bound to EGFR or BSA (control) pre-coated on the solid phase were detected with HRP-labeled goat anti-human IgG or HRP-labeled goat anti-human IgM.
  • Figure 8 shows the IgG and IgM responses of cells immunized with tumor cells in vitro.
  • Figure 9 shows reactivity of clones to GM-CSF, chick ovalbumin (CAB), or keyhole limpet hemocyanin.
  • Figure 10 shows inhibitory effect of anti-GM-CSF antibodies on proliferation of TF-1 cells. Shown are the effects of a GM-CSF-specific, blocking antibody; a GM-CSF-specific, non-blocking antibody; and a non-specific antibody.
  • the invention provides various embodiments of a method for producing antibody- producing cells and antibodies from in vitro immunized cells with high affinity, and/or increased production.
  • the cells that produce the antibodies are hybridoma cells, formed by fusing myeloma cells with the lymphoid cells that have been immunized against an antigen in vitro.
  • the cells that produce the antibodies are mammalian cells that have been transfected with immunoglobulin genes cloned from lymphoid cells that have been immunized against an antigen in vitro.
  • the method employs both hybridoma cells and mammalian cells.
  • the invention provides a method for generating hybridoma cells producing high-affinity antibodies from in vitro immunized, immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells express a dominant negative allele of a mismatch repair gene; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for the antigen than antibodies produced by the parental hybridoma cells; thereby producing hybridoma cells producing high-affinity antibodies.
  • the invention provides methods of producing hybridoma cells that produce high titers of antibodies from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells express a dominant negative allele of a mismatch repair gene; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen of the hypermutated hybridoma cells for antigen-specific antibodies produced in higher titers than that produced by the parental hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce higher titers of antibodies than that produced by the parental hybridoma cells; thereby producing hybridoma cells that produce high titers of antibodies.
  • the invention provides a method for producing hybridoma cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing said immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for binding of antibodies produced from said hybridoma cells to antigen; (d) cloning immunoglobulin genes from said hybridoma into a mammalian expression cell, wherein said mammalian expression cell expresses a dominant negative allele of a mismatch repair gene; and (e) performing a screen for mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from said hybridoma cells; thereby producing hybridoma cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells comprising
  • the invention provides a method for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells are produced by: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing said immuiioglobulin-producing cells with myeloma cells to form hybridoma cells, wherein said hybridoma cells express a dominant negative allele of a mismatch repair gene; (c) incubating said parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from said hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for said antigen than antibodies produced by said parental hybridoma cells; and (f) cloning immunoglobulin genes from said hybridoma into
  • the invention provides mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin- producing cells are produced by: (a) combining donor cells comprising immunoglobulin- producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin- producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for binding of antibodies produced from the hybridoma cells to antigen; (d) cloning immunoglobulin genes from the hybridoma into a parental mammalian expression cell, wherein the mammalian expression cell expresses a dominant negative allele of a mismatch repair gene; (e) incubating the parental mammalian expression cell to allow for mutagenesis, thereby forming hypermutated mammalian expression cells; (f) performing a screen of hypermutable mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from the
  • the invention provides a method of producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells are produced by: (a) combining donor cells comprising immunoglobulin- producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin- producing cells with myeloma cells to form parental hybridoma cells; (c) incubating the parental hybridoma cells in the presence of at least one chemical inhibitor of mismatch repair, thereby forming hypermutated hybridoma cells; (d) performing a screen for antigen binding for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for the antigen than antibodies produced by said parental hybridoma cells; thereby producing hybridoma cells that produce high-affinity antibodies.
  • the invention provides a method of producing hybridoma cells that produce high titers of antibodies from in vitro immunized immunoglobulin- producing cells are produced by: (a) combining donor cells comprising immunoglobulin- producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin- producing cells with myeloma cells to form parental hybridoma cells; (c) incubating the parental hybridoma cells in the presence of at least one chemical inhibitor of mismatch repair, thereby forming hypermutated hybridoma cells; (d) performing a screen of the hypermutated hybridoma cells for antigen-specific antibodies produced in higher titers than that produced by the parental hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce higher titers of antibodies than that produced by said parental hybridoma cells; thereby producing hybridoma cells producing high titers of antibodies.
  • the invention provides methods for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells are produced by: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for antigen binding of antibodies produced from the hybridoma cells; (d) cloning immunoglobulin genes from the hybridoma cells into a mammalian expression cell; (e) incubating the mammalian expression cell in the presence of at least one chemical inhibitor of mismatch repair; and (f) performing a screen for mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from the hybridoma cells; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized
  • the invention provides a method for producing mammalian expression cells that produce high affimty antibodies to a selected antigen from in vitro immunized immunoglobulin-producing cells are produced in high titers by: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) incubating the hybridoma cells in the presence of at least one chemical inhibitor of mismatch repair to form hypermutated hybridoma cells; (d) performing a screen for antigen binding for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for the antigen than antibodies produced by the parental hybridoma cells; and (f) cloning immunoglobulin genes from the hypermutated hybridoma cells into a mammalian expression cell, thereby forming parental mamma
  • the invention also provides methods for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing said immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for binding of antibodies produced from said hybridoma cells to antigen; - (d) cloning immunoglobulin genes from said hybridoma into a mammalian expression cell; (e) incubating said mammalian expression cell in the presence of at least one chemical inhibitor of mismatch repair, thereby forming a hypermutated mammalian expression cell; (f) performing a screen for hypermutated mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from said parental mammalian expression cells; and (g) performing a
  • the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro, wherein the donor cells are derived from a donor that is naturally deficient in mismatch repair; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; thereby producing hybridoma cells producing high-affinity antibodies.
  • the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells, wherein the myeloma cells are naturally deficient in mismatch repair, thereby forming parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; thereby producing hybridoma cells producing high-affinity antibodies.
  • the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells in high titers comprising: (a) combimng donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro, wherein the donor cells are derived from a donor that is naturally deficient in mismatch repair; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; (f) performing a second screen for hypermut
  • the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells in high titers comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells, wherein the myeloma cells are naturally deficient in mismatch repair, thereby forming parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; (f) performing a second screen for hypermutated
  • the invention comprises a method for producing mammalian expression cells that produce high-affinity antibodies in high titers from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro, wherein the donor cells are derived from a donor that is naturally deficient in mismatch repair; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypennutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; and (f) cloning immunoglobin-producing cells comprising:
  • the invention comprises a method for producing mammalian expression cells that produce high-affinity antibodies in high titer from in vitro immunized immunoglobulin-producing cells comprising: (a) combimng donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells, wherein the myeloma cells are naturally deficient in mismatch repair, thereby forming parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; and (f) cloning immunoglobin-producing cells comprising:
  • antigen-specific immunoglobulin-producing cells are produced by: (a) isolating donor cells from an animal; (b) treating said cells with L-leucyl-L- leucine methy ester hydrobromide; (c) incubating said donor cells with an immunogenic antigen in vitro, at 25-37°C, 5-10% CO 2 , in medium supplemented with 5-15% serum, and a growth promoting cytokine for 4 days; (d) washing said cells in medium; and (e) culturing said cells in medium supplemented with 5-15% serum an additional 8 days; thereby stimulating the production of antigen-specific immunoglobulin-producing cells.
  • the blood cells used in the methods of the invention may be derived from any animal that produces antibodies.
  • the donor cells are derived from mammals, including, but not limited to humans, monkeys, mice, rats, guinea pigs, hamsters, gerbils, birds, rabbits, sheep, goats, pigs, horses, and cows.
  • the source of blood is not necessarily limited, but may be whole blood or fractions containing lymphocytes.
  • the blood may be donor or cord blood, for example.
  • the blood cells are preferably human donor cells.
  • the myeloma cells used to create the hybridoma cells in the method of the invention may be derived from any species known to have suitable myeloma cells.
  • the myeloma cells may be conveniently derived from humans or mice. Suitable examples of myeloma cells include, but are not limited to the HuNSl myeloma as described in U.S. Patent No. 4,720,459 to Winkelhake, and deposited with the American Type Culture Collection (ATCC) as CRL 8644; GM4672; RPMI 8226; and murine myeloma cell lines (e.g., P3-NSl/l-Ag4-l; P3-x63-Ag8.653; Sp2/O-Agl4; NS/O, NS/1, SP2 and S194).
  • HuNSl myeloma as described in U.S. Patent No. 4,720,459 to Winkelhake, and deposited with the American Type Culture Collection (ATCC) as CRL 8644; GM4672; RPMI 8226; and murine myeloma cell lines (e.g., P3-NSl
  • the mammalian expression cells suitable for use in certain embodiments of the method of the invention include, but are not limited to Chinese Hamster Ovary cells (CHO cells, Urlaub and Chasin (1980) Proc. Natl. Acad. Sci. USA, 11: 4216), baby hamster kidney (BHK cells), human embryonic kidney line 293 (HeLa cells, Graham et al, (1977) J.
  • Chinese Hamster Ovary cells CHO cells, Urlaub and Chasin (1980) Proc. Natl. Acad. Sci. USA, 11: 4216
  • BHK cells baby hamster kidney
  • HeLa cells Graham et al, (1977) J.
  • Gen Virol, 36: 59 normal dog kidney cell line (e.g., MDCK, ATCC CCL 34), normal cat kidney cell line (CRFK cells), monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587), COS (e.g., COS-7) cells, and non-tumorigenic mouse myoblast G8 cells (e.g., ATCC CRL 1246), fibroblast cell lines (e.g., human, murine or chicken embryo fibroblast cell lines), myeloma cell lines, mouse NIH/3T3 cells, LMTK cells, mouse sertoli cells (TM4, Mather, (1980) Biol.
  • MDCK normal dog kidney cell line
  • CCFK cells normal cat kidney cell line
  • CCL 70 monkey kidney cells
  • V1 ATCC CCL 70 African green monkey kidney cells
  • COS e.g., COS-7 cells
  • non-tumorigenic mouse myoblast G8 cells e.g., ATCC C
  • non-mammalian cells include, but are not limited to insect cells (e.g., Spodoptera frugiperda cells and the like).
  • Vectors and non- mammalian host cells are well known in the art and are continually being optimized and developed. Any host cell system capable of expressing antibodies may be used in the methods of the invention.
  • mismatch repair gene refers to an allele of a mismatch repair gene that, when expressed, exerts a dominant phenotype in the cell or organism that leads to an inhibition of the mismatch repair system, even in the presence of a wild-type allele.
  • Cells expressing a dominant negative allele of a mismatch repair gene are hypermutable and accumulate mutations at a higher rate than wild-type cells.
  • nucleic acid sequences encoding mismatch repair proteins useful in the method of the invention include, but are not limited to the following: PMS1 (SEQ LD NO:l); PMS2 (SEQ LO NO:3); PMS2-134 (SEQ TD NO:5); PMSR2 (SEQ ID NO:7); PMSR3 (SEQ ID NO:9); MLHl (SEQ ID NO:ll); MLH3 (SEQ ID NO:13); MSH2 (SEQ ID NO:15); MSH3 (SEQ ID NO:17); MSH4 (SEQ ID NO: 19); MSH5 (SEQ LD NO:21); MSH6 (SEQ ID NO:23); PMSR6 (SEQ ID NO:25); PMSL9 (SEQ ID NO:27); yeast MLHl (SEQ LD NO:29); mouse PMS2 (SEQ LD NO:31); mouse PMS2-134 (SEQ LD NO:33); Arabidopsis thaliana PMS2 (SEQ ID NO:35); A.
  • thaliana PMS2-134 (SEQ ID NO:37) A. thaliana PMS1 (SEQ ID NO:39); A. thaliana MSH7 (SEQ ID NO:41) A. thaliana MSH2 (SEQ JD NO:43); A. thaliana MSH3 (SEQ ID NO:45); A. thaliana MSH6-1 (SEQ ID NO:47); and Oryza satvia MLHl (SEQ ID NO:49).
  • the corresponding amino acid sequences for the listed nucleic acid sequences are: PMS1 (SEQ ID NO:2); PMS2 (SEQ LD NO:4); PMS2-134 (SEQ ID NO:6); PMSR2 (SEQ ID NO:8); PMSR3 (SEQ ID NO:10); MLHl (SEQ 3D NO:12); MLH3 (SEQ LD NO:14); MSH2 (SEQ LD NO: 16); MSH3 (SEQ ID NO: 18); MSH4 (SEQ ID NO:20); MSH5 (SEQ ID NO:22); MSH6 (SEQ ID NO:24); PMSR6 (SEQ ID NO:26); PMSL9 (SEQ ID NO:28); yeast MLHl (SEQ LD NO:30); mouse PMS2 (SEQ ID NO:32); mouse PMS2-134 (SEQ ID NO:34); Arabidopsis thaliana PMS2 (SEQ ID NO:36); A.
  • thaliana PMS2-134 SEQ ID NO:38
  • A. thaliana PMS1 SEQ ID NO:40
  • A. thaliana MSH7 SEQ ID NO:42
  • A. thaliana MSH2 SEQ ID NO:44
  • A. thaliana MSH3 SEQ LD NO:46
  • A. thaliana MSH6-1 SEQ ID NO:48
  • Oryza satvia MLHl SEQ ID NO:50
  • high titer refers to an titer of at least about 1.5 fold higher than the parental cell line. In some embodiments, the titer is at least about 1.5-3 fold higher, 3-5 fold higher, 5-7 fold higher, 7-9 fold higher, or 9-10 fold higher than the parental cell line.
  • "high affinity" is at least about 1 x 10 7 M "1 .
  • the antibodies have an affinity of at least about 1 x 10 s M "1 .
  • the antibodies have an affinity of at least about 1 x 10 9 M "1 .
  • the antibodies have an affinity of at least about 1 x 10 10 M "1 .
  • the antibodies have an affinity of at least about 1 x 10 11 M "1 .
  • the antibodies have an affinity of at least about 1 x 10 12 M . In other embodiments, the antibodies have an affinity of at least about 1 x 10 13 M "1 . In other embodiments, the antibodies have an affinity of at least about 1 x 10 14 M "1 . [0129] As used herein, "antigen-specific" refers to an interaction between the CDR regions of the immunoglobulin molecule with an epitope of the antigen wherein the CDR regions of the immunoglobulin molecule binds to the epitope.
  • cured refers to a state of the cells wherein the dominant negative mismatch repair gene has been eliminated from the cell or wherein the expression of the dominant negative allele has been turned off, leading to a stabilized genome, producing stable biological products such as immunoglobulins.
  • mismatch repair is inhibited by introducing a dominant negative allele of a mismatch repair gene into a cell.
  • mismatch repair is inhibited by exposing cells that express an antibody to a compound that inhibits mismatch repair.
  • the compound is an ATPase inhibitor.
  • Suitable ATPase inliibitors include, but not limited to ATP analogs that are capable of blocking the ATPase activity necessary for mismatch repair in the cell.
  • ATP analogs examples include, but are not limited to non-hydrolyzable forms of ATP, such as AMP-PNP and ATP ⁇ S, which block mismatch repair activity (Galio et al. (1999) Nucl. Acids Res. 27:2325-2331; Allen et al. (1997) EMBO J. 16:4467-4476; Bjornson et al. (2000) Biochem. 39:3176-3183).
  • Other suitable ATPase inhibitors may be identified using mismatch repair reporter cells that may be screened with candidate ATPase inhibitors to identify those compounds which effectively block ATPase activity in the cells.
  • mismatch repair is inhibited by exposing cells that express an antibody to a nuclease inhibitor.
  • the nuclease inhibitors are capable of blocking exonuclease activity in the mismatch repair biochemical pathway.
  • Mismatch repair reporter cells may be screened with candidate nuclease inhibitors to identify compounds that effectively block the exonuclease activity of the mismatch repair system.
  • Suitable nuclease inhibitors which may be used in the methods of the invention include, but are not limited to analogs of N-ethylmaleimide, an endonuclease inhibitor (Huang et al. (1995) Arch. Biochem. Biophys.
  • nuclease inhibitors may be identified using mismatch repair reporter cells that may be screened with candidate nuclease inhibitors to identify those compounds which effectively block nuclease activity in the cells.
  • DNA polymerase inhibitors are capable of blocking the polymerization of DNA which is required for functional mismatch repair.
  • suitable DNA polymerase inhibitors include, but are not limited to actinomycin D (Martin et al. (1990) J. Immunol. 145:1859); aphidicolin (Kuwakado et al. (1993) Biochem. Pharmacol. 46:1909); l-(2'-deoxy-2'-fluoro-beta-L-arabinofuranosyl)-5- methyluracil (L-FMAU) (Kukhanova et al.
  • DNA polymerase inhibitors may be identified using mismatch repair reporter cells that may be screened with candidate DNA polymerase inhibitors to identify those compounds which effectively block DNA polymerase activity in the cells.
  • mismatch repair is inhibited by exposing the cells producing antibody to an anthracene.
  • anthracene refers to the compound anthracene.
  • anthracene refers to the compound anthracene.
  • alkyl refers to a hydrocarbon containing from 1 to about 20 carbon atoms. Alkyl groups may straight, branched, cyclic, or combinations thereof. Alkyl groups thus include, by way of illustration only, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, cyclopentyl, cyclopentylmethyl, cyclohexyl, cyclohexylmethyl, and the like.
  • alkyl fused and/or polycyclic aliphatic cyclic ring systems such as, for example, adamantane.
  • alkenyl denotes an alkyl group having at least one carbon-carbon double bond.
  • alkynyl denotes an alkyl group having at least one carbon-carbon triple bond.
  • the alkyl, alkenyl, alkynyl, aryl, aryloxy, and heteroaryl substituent groups described above may bear one or more further substituent groups; that is, they may be "substituted".
  • these substituent groups can include halogens (for example fluorine, chlorine, bromine and iodine), CN, NO 2 , lower alkyl groups, aryl groups, heteroaryl groups, aralkyl groups, aralkyloxy groups, guanidino, alkoxycarbonyl, alkoxy, hydroxy, carboxy and amino groups.
  • substituted alkyl groups include, for example, alkyl groups fluoro-, chloro-, bromo- and iodoalkyl groups, aminoalkyl groups, and hydroxyalkyl groups, such as hydroxymethyl, hydroxyethyl, hydroxypropyl, hydroxybutyl, and the like. In some prefened embodiments such hydroxyalkyl groups contain from 1 to about 20 carbons.
  • aryl means a group having 5 to about 20 carbon atoms and which contains at least one aromatic ring, such as phenyl, biphenyl and naphthyl. Prefened aryl groups include unsubstituted or substituted phenyl and naphthyl groups.
  • aryloxy denotes an aryl group that is bound through an oxygen atom, for example a phenoxy group.
  • hetero denotes the presence of at least one hetero (i.e., non- carbon) atom, which is in some preferred embodiments independently one to three O, N, S, P, Si or metal atoms.
  • heteroaryl denotes an aryl group in which one or more ring carbon atom is replaced by such a heteroatom.
  • Prefened heteroaryl groups include pyridyl, pyrimidyl, pynolyl, furyl, thienyl, and imidazolyl groups.
  • aralkyl (or “arylalkyl”) is intended to denote a group having from 6 to 15 carbons, consisting of an alkyl group that bears an aryl group.
  • aralkyl groups include benzyl, phenethyl, benzhydryl and naphthylmethyl groups.
  • alkylaryl (or “alkaryl”) is intended to denote a group having from 6 to 15 carbons, consisting of an aryl group that bears an alkyl group.
  • aralkyl groups include methylphenyl, ethylphenyl and methylnaphthyl groups.
  • arylsulfonyl denotes an aryl group attached through a sulfonyl group, for example phenylsulfonyl.
  • alkylsulfonyl denotes an alkyl group attached through a sulfonyl group, for example methylsulfonyl.
  • arylalkyloxy or “aralkyloxy” are equivalent, and denote a group of formula -O-R ⁇ R 7 , where R ; is R is alkyl, alkenyl, or alkynyl which can be optionally substituted as described herein, and wherein R 7/ denotes a aryl or substituted aryl group.
  • alkylaryloxy or “alkaryloxy” are equivalent, and denote a group of formula -O-R'-R 77 , where R ; is an aryl or substituted aryl group, and R / is alkyl, alkenyl, or alkynyl which can be optionally substituted as described herein.
  • ketone denotes a moiety containing a group of formula -R-
  • esters denotes a moiety having a group of formula -R-
  • R are independently alkyl, alkenyl, alkynyl, aryl, heteroaryl, aralkyl, or alkaryl, each of which may be substituted as described herein.
  • R are independently alkyl, alkenyl, alkynyl, aryl, heteroaryl, aralkyl, or alkaryl, each of which may be substituted as described herein.
  • cyclic ether has its usual meaning of a cyclic ether containing several oxygen atoms.
  • organosulfur compound denotes aliphatic or aromatic sulfur containing compounds, for example thiols and disulfides.
  • organometallic group denotes an organic molecule containing at least one metal atom.
  • organosilicon compound denotes aliphatic or aromatic silicon containing compounds, for example alkyl and aryl silanes.
  • carboxylic acid denotes a moiety having a carboxyl group, other than an amino acid.
  • Suitable anthracenes that may be used in the method of the invention comprise compounds having the formula:
  • R Rio are independently hydrogen, hydroxyl, amino, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S- alkenyl, N-alkenyl, O-alkynyl, S-alkynyl, N-alkynyl, aryl, substituted aryl, aryloxy, substituted aryloxy, heteroaryl, substituted heteroaryl, aralkyloxy, arylalkyl, alkylaryl, alkylaryloxy, arylsulfonyl, alkylsulfonyl, alkoxycarbonyl, aryloxycarbonyl, guanidino, carboxy, an alcohol, an amino acid, sulfonate, alkyl sulfonate, CN, NO 2 , an aldehyde group, an este
  • R 5 and R ⁇ are hydrogen.
  • Ri-Rio are independently hydrogen, hydroxyl, methyl, ethyl, propyl, isopropyl, butyl, isoburyl, phenyl, tolyl, hydroxymethyl, hydroxypropyl, or hydroxybutyl.
  • Suitable anthracenes for use in the methods of the invention include, but are not limited to 1,2-dimethylanthracene, 9,10- dimethylanthracene, 7,8-dimethylanthracene, 9,10-duphenylanthracene, 9,10- dihydroxymethylanthracene, 9-hydroxymethyl- 10-methylanthracene, dimethylanthracene- 1 ,2- diol, 9-hydroxymethyl- 10-methylanthracene- 1 ,2-diol, 9-hydroxymethyl- 10-methylanthracene- 3,4-diol, and 9,10-di-m-tolylanthracene.
  • RNA interference molecule that is homologous to a mismatch repair gene of the invention.
  • the technique for generating sequence-specific RNA interference molecules is well-known in the art and may be found in, for example, Sharp et al. (2000) Science 287 r :2431-2433; Marx (2000) Science 288:1370-1372; Grishok et al. (2001) Science 287:2494-2497; and Fire et al. (1998) Nature 391:806-811, the disclosures of which are specifically incorporated by reference in their entirety.
  • mismatch repair is inhibited by exposing the cells producing antibody to "antisense compounds" which specifically hybridize with one or more nucleic acids encoding a mismatch repair gene.
  • target nucleic acid and “nucleic acid encoding a mismatch repair gene” encompass DNA encoding a mismatch repair gene, RNA (including pre-mRNA and mRNA) transcribed from such DNA, and also cDNA derived from such RNA.
  • RNA including pre-mRNA and mRNA
  • RNA disrupted by antisense compounds include such functions as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, and splicing of the RNA to yield one or more mRNA species.
  • the antisense compound thereby inhibits the expression or function of a mismatch repair gene.
  • It is prefened to target specific nucleic acids for antisense inhibition of mismatch repair in order to reversibly disrupt the function of a given mismatch repair gene.
  • “Targeting" an antisense compound to a particular nucleic acid is a multistep process, beginning with the identification of a nucleic acid sequence whose function is to be modulated.
  • mismatch repair genes there are several mismatch repair genes that may be targeted by an antisense strategy.
  • various mismatch repair genes that may be targeted are PMS2, PMSl, PMSR3, PMSR2, PMSR6, MLHl, GTBP, MSH3, MSH2, MLH3, or MSH1, and homologs of PMSR genes as described in Nicolaides et al. (1995) Genomics 30:195-206 and Horii er a/. (1994) Rtoc/ ⁇ em. Biophys. Res. Commun. 204:1257-1264, including DNA or RNA.
  • the next step of targeting involves the determination of a site or sites within this gene for the antisense interaction to occur, such that inhibition of the function of the mismatch repair gene occurs.
  • an intragenic site is targeted.
  • An "intragenic site” is a region encompassing the translation initiation or termination codon of the open reading frame (ORF) of the gene. Since, as is known in the art, the translation initiation codon is typically 5'-AUG (in transcribed mRNA molecules; 5'-ATG in the conesponding DNA molecule), the translation initiation codon is also refened to as the "AUG codon," the “start codon” or the "AUG start codon.” A minority of genes have a translation initiation codon having the RNA sequence 5'-GUG, 5'-UUG or 5'-CUG, and 5*-AUA, 5'-ACG and 5'- CUG have been shown to function in vivo.
  • translation initiation codon and “start codon” can encompass many codon sequences, even though the initiator amino acid in each instance is typically methionine in eukaryotes. It is also known in the art that eukaryotic and prokaryotic genes may have two or more alternative start codons, any one of which may be preferentially utilized for translation initiation in a particular cell type or tissue, or under a particular set of conditions, hi the context of the invention, “start codon” and “translation initiation codon” refer to the codon or codons that are used in vivo to initiate translation of an mRNA molecule transcribed from a gene encoding a mismatch repair gene, regardless of the sequence(s) of such codons.
  • a translation termination codon (or "stop codon”) of a gene may have one of three sequences, i.e., 5'-UAA, 5'-UAG and 5'-UGA (the corresponding DNA sequences are 5'-TAA, 5'-TAG and 5'-TGA, respectively).
  • start codon region and “translation initiation codon region” refer to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (t ' .e., 5' or 3') from a translation initiation codon.
  • stop codon region and “translation termination codon region” refer to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5' or 3') from a translation termination codon.
  • Other target regions include the 5' untranslated region (5'UTR), known in the art to refer to the portion of an mRNA in the 5' direction from the translation initiation codon, and thus including nucleotides between the 5' cap site and the translation initiation codon of an mRNA or conesponding nucleotides on the gene, and the 3' untranslated region (3'UTR), known in the art to refer to the portion of an mRNA in the 3' direction from the translation termination codon, and thus including nucleotides between the translation tennination codon and 3' end of an mRNA or conesponding nucleotides on the gene.
  • 5'UTR 5' untranslated region
  • 3'UTR 3' untranslated region
  • the 5' cap of an mRNA comprises an N7-methylated guanosine residue joined to the 5'-most residue of the mRNA via a 5'-5' triphosphate linkage.
  • the 5' cap region of an mRNA is considered to include the 5' cap structure itself as well as the first 50 nucleotides adjacent to the cap.
  • the 5' cap region may also be a prefened target region.
  • mRNA splice sites i.e., intron-exon junctions
  • intron-exon junctions may also be prefened target regions, and are particularly useful in situations where abenant splicing is implicated in disease, or where an overproduction of a particular mRNA splice product is implicated in disease.
  • Abenant fusion junctions due to reanangements or deletions are also prefened targets. It has also been found that introns can also be effective, and therefore prefened, target regions for antisense compounds targeted, for example, to DNA or pre-mRNA.
  • oligonucleotides are chosen which are sufficiently complementary to the target, i.e., hybridize sufficiently well and with sufficient specificity, to give the desired effect.
  • hybridization means hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleoside or nucleotide bases.
  • adenine and thymine are complementary nucleobases which pair through the formation of hydrogen bonds.
  • “Complementary,” as used herein, refers to the capacity for precise pairing between two nucleotides.
  • oligonucleotide and the DNA or RNA are considered to be complementary to each other at that position.
  • the oligonucleotide and the DNA or RNA are complementary to each other when a sufficient number of conesponding positions in each molecule are occupied by nucleotides which can hydrogen bond with each other.
  • An antisense compound is specifically hybridizable when binding of the compound to the target DNA or RNA molecule interferes with the normal function of the target DNA or RNA to cause a loss of utility, and there is a sufficient degree of complementarity to avoid non-specific binding of the antisense compound to non-target sequences under conditions in which specific binding is desired, i.e., under physiological conditions in the case of in vivo assays or therapeutic treatment, and in the case of in vitro assays, under conditions in which the assays are performed.
  • Complementarity of the antisense oligonucleotide is preferably 100%, however, degeneracy may be introduced into the oligonucleotide such that the complementarity, in some embodiments, is 80-85%, 85-90%, 90- 95% or 95-100%.
  • Antisense and other compounds of the invention which hybridize to the target and inhibit expression of the target are identified through experimentation, and the sequences of these compounds are herein below identified as prefened embodiments of the invention.
  • the target sites to which these prefened sequences are complementary comprise the region of PMS2, for example, which inhibits the translation of the C-terminal portion of the PMS2 protein, effectively forming a truncation mutant.
  • the region targeted comprises a portion of the PMS2 gene that encodes the 134 amino acid of PMS2, for example.
  • oligonucleotide refers to an oligomer or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) or mimetics thereof.
  • RNA ribonucleic acid
  • DNA deoxyribonucleic acid
  • oligonucleotides composed of naturally-occurring nucleobases, sugars and covalent internucleoside (backbone) linkages as well as oligonucleotides having non-naturally- occurring portions which function similarly.
  • backbone covalent internucleoside
  • Such modified or substituted oligonucleotides are often prefened over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for nucleic acid target and increased stability in the presence of nucleases.
  • antisense oligonucleotides are a prefened form of antisense compound
  • the present invention comprehends other oligomeric antisense compounds, including but not limited to oligonucleotide mimetics such as are described below.
  • the antisense compounds in accordance with this invention preferably comprise from about 8 to about 50 nucleobases (i.e., from about 8 to about 50 linked nucleosides).
  • Particularly prefened antisense compounds are antisense oligonucleotides, even more preferably those comprising from about 12 to about 30 nucleobases.
  • Antisense compounds include ribozymes, external guide sequence (EGS) oligonucleotides (oligozymes), and other short catalytic RNAs or catalytic oligonucleotides which hybridize to the target nucleic acid and modulate its expression.
  • the oligonucleotides are at least about 15 nucleotides in length and may be at least 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more nucleotides in length.
  • the antisense oligonucleotides comprise a sequence that is complementary to a portion of the mismatch repair sequence shown in SEQ ID NO:l; SEQ LD NO:3; SEQ LD NO:5; SEQ LD NO:7; SEQ ID NO:9; SEQ ID NO.ll; SEQ ID NO:13; SEQ ID NO:15; SEQ J-D NO:17; SEQ J-D NO:19; SEQ ID NO:21; SEQ JD NO:23; SEQ LD NO:25; SEQ J-D NO:27; SEQ ID NO:29; SEQ ID NO:31; SEQ J-D NO:33; SEQ ID NO:35; SEQ ID NO:37; SEQ ID NO:39; SEQ ID NO:41; SEQ ID NO:43; SEQ ID NO:45; SEQ ID NO:47; or SEQ ID NO:49.
  • the oligonucleotide is at least 15-50 nucleotides in length
  • nucleoside is a base-sugar combination.
  • the base portion of the nucleoside is normally a heterocychc base.
  • the two most common classes of such heterocychc bases are the purines and the pyrimidines.
  • Nucleotides are nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside.
  • the phosphate group can be linked to either the 2', 3 Or 5' hydroxyl moiety of the sugar.
  • the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound.
  • this linear polymeric structure can be further joined to form a circular structure, however, open linear structures are generally prefened.
  • the phosphate groups are commonly refened to as fonning the internucleoside backbone of the oligonucleotide.
  • the normal linkage or backbone of RNA and DNA is a 3' to 5' phosphodiester linkage.
  • prefened antisense compounds useful in this invention include oligonucleotides containing modified backbones or non-natural internucleoside linkages.
  • oligonucleotides having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone.
  • modified oligonucleotides that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleosides.
  • Prefened modified oligonucleotide backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates, 5'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates and boranophosphates having normal 3'-5' linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a 3' to 3', 5' to 5' or 2' to 2' linkage.
  • Prefened oligonucleotides having inverted polarity comprise a single 3' to 3' linkage at the 3'-most internucleotide linkage, i.e., a single inverted nucleoside residue which may be a basic (the nucleobase is missing or has a hydroxyl group in place thereof).
  • Various salts, mixed salts and free acid forms are also included. [0169] Representative United States patents that teach the preparation of the above phosphorus-containing linkages include, but are not limited to, U.S. Pat. Nos.
  • Prefened modified oligonucleotide backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocychc internucleoside linkages.
  • morpholino linkages formed in part from the sugar portion of a nucleoside
  • siloxane backbones sulfide, sulfoxide and sulfone backbones
  • formacetyl and thioformacetyl backbones methylene formacetyl and thioformacetyl backbones
  • riboacetyl backbones alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH 2 component parts.
  • Representative United States patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Pat. Nos. 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; 5,792,608; 5,646,269 and 5,677,439, each of which is herein incorporated by reference.
  • both the sugar and the internucleoside linkage, i.e., the backbone, of the nucleotide units are replaced with novel groups.
  • the base units are maintained for hybridization with an appropriate nucleic acid target compound.
  • One such oligomeric compound, an oligonucleotide mimetic that has been shown to have excellent hybridization properties is refened to as a peptide nucleic acid (PNA).
  • PNA peptide nucleic acid
  • the sugar-backbone of an oligonucleotide is replaced with an amide containing backbone, in particular an aminoethylglycine backbone.
  • nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone.
  • Representative United States patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Pat. Nos. 5,539,082; 5,714,331; and 5,719,262, each of which is herein incorporated by reference. Further teaching of PNA compounds can be found in Nielsen et al, (1991) Science 254:1497-1500.
  • Most prefened embodiments of the invention are oligonucleotides with phosphorothioate backbones and oligonucleosides with heteroatom backbones, and in particular -CH 2 -NH-O-CH 2 -, -CH 2 ⁇ N(CH 3 )-O ⁇ CH 2 ⁇ [known as a methylene (methylimino) or MMI backbone], -CH 2 -O ⁇ N(CH 3 )-CH 2 --, ⁇ CH 2 ⁇ N(CH 3 ) ⁇ N(CH 3 )-CH 2 ⁇ and ⁇ O ⁇ N(CH 3 ) ⁇ CH ⁇ CH 2 — [wherein the native phosphodiester backbone is represented as -O-P-O ⁇ CH 2 -] of the above referenced U.S.
  • Modified oligonucleotides may also contain one or more substituted sugar moieties.
  • Prefened oligonucleotides comprise one of the following at the 2' position: OH; F; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted to o alkyl or C 2 to C 10 alkenyl and alkynyl.
  • Particularly prefened are O[(CH 2 ) n O] m CH 3 , O(CH 2 ) n OCH 3 , O(CH 2 ) complicatNH 2 , O(CH 2 ) n CH 3 , O(CH 2 ) n ONH 2 , and O(CH 2 ) n ON[(CH 2 ) n CH 3 )] 2 , where n and m are from 1 to about 10.
  • oligonucleotides comprise one of the following at the 2 1 position: to Cio lower alkyl, substituted lower alkyl, alkenyl, alkynyl, alkaryl, aralkyl, O-alkaryl or O- aralkyl, SH, SCH 3 , OCN, Cl, Br, CN, CF 3 , OCF 3 , SOCH 3 , SO 2 CH 3 , ONO 2 , NO 2 , N 3 , NH 2 , heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties.
  • a prefened modification includes 2' -methoxyethoxy (2'-O ⁇ CH 2 CH 2 OCH 3 , also known as 2'-O-(2-methoxyethyl) or 2'-MOE) (Martin et al. (1995) Helv. Chim. Acta 78:486- 504) i.e., an alkoxyalkoxy group.
  • a further prefened modification includes 2'- dimethylaminooxyethoxy, i.e., a O(CH 2 ) 2 ON(CH 3 ) 2 group, also known as 2'-DMAOE, as described in examples hereinbelow, and 2'-dimethylaminoethoxyethoxy (also known in the art as 2'-O-dimethylaminoethoxyethyl or 2'-DMAEOE), i.e., 2'-O ⁇ CH 2 ⁇ O-CH 2 ⁇ N(CH 2 ) 2 , also described in examples hereinbelow.
  • 2'- dimethylaminooxyethoxy i.e., a O(CH 2 ) 2 ON(CH 3 ) 2 group, also known as 2'-DMAOE, as described in examples hereinbelow
  • 2'-dimethylaminoethoxyethoxy also known in the art as 2'-O-dimethylaminoethoxyethyl or 2'-DMAE
  • a further prefened modification includes Locked Nucleic Acids (LNAs) in which the 2'-hydroxyl group is linked to the 3 ' or 4' carbon atom of the sugar ring thereby forming a bicyclic sugar moiety.
  • the linkage is preferably a methelyne ( ⁇ CH 2 ⁇ ) n group bridging the 2' oxygen atom and the 3' or 4' carbon atom wherein n is 1 or 2.
  • LNAs and preparation thereof are described in WO 98/39352 and WO 99/14226.
  • the 2'-modification may be in the arabino (up) position or ribo (down) position.
  • a prefened 2'-arabino modification is 2'-F.
  • oligonucleotide Similar modifications may also be made at other positions on the oligonucleotide, particularly the 3' position of the sugar on the 3' terminal nucleotide or in 2'-5' linked oligonucleotides and the 5' position of 5' terminal nucleotide. Oligonucleotides may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. Representative United States patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S. Pat. Nos. 4,981,957; 5,118,800;
  • Oligonucleotides may also include nucleobase (often refened to in the art simply as "base”) modifications or substitutions.
  • unmodified or “natural” nucleobases include the piirine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U).
  • Modified nucleobases include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2- thiocytosine, 5-halouracil and cytosine, 5-propynyl ( ⁇ C ⁇ C ⁇ CH 2 ) uracil and cytosine and other alkynyl derivatives of pyrimidine bases, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8- substituted adenines and guanines
  • nucleobases include tricyclic pyrimidines such as phenoxazine cytidine(lH-pyrimido[5,4-b][l,4]benzoxazin-2(3H)-one), phenothiazine cytidine (lH-pyrimido[5,4-b][l,4]benzothiazin-2(3H)-one), G-clamps such as a substituted phenoxazine cytidine (e.g., 9-(2-aminoethoxy)-H-pyrimido[5,4-b][l,4]benzoxazin-2(3H)-one), carbazole cytidine (2H-pyrimido[4,5-b]indol-2-one), pyridoindole cytidine (H- pyrido[3',2':4,5]pynolo[2,3-d]pyrimidin-2-one).
  • Modified nucleobases may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example 7- deaza-adenine, 7-deazaguanosine, 2-aminopyridine and 2-pyridone.
  • Further nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in THE CONCISE ENCYCLOPEDIA OF POLYMER SCIENCE AND ENGINEERING, Kroschwitz, (Ed.) John Wiley & Sons, 1990, pages 858-859, those disclosed by Englisch et al.
  • nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2°C.
  • Another modification of the oligonucleotides of the invention involves chemically linking to the oligonucleotide one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide.
  • the compounds of the invention can include conjugate groups covalently bound to functional groups such as primary or secondary hydroxyl groups.
  • Conjugate groups of the invention include intercalators, reporter molecules, polyamines, polyamides, polyethylene glycols, polyethers, groups that enhance the pharmacodynamic properties of oligomers, and groups that enhance the pharmacokinetic properties of oligomers.
  • Typical conjugates groups include cholesterols, lipids, phosphohpids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes.
  • Groups that enhance the pharmacodynamic properties include groups that improve oligomer uptake, enhance oligomer resistance to degradation, and/or strengthen sequence-specific hybridization with RNA.
  • Groups that enhance the pharmacokinetic properties include groups that improve oligomer uptake, distribution, metabolism or excretion. Representative conjugate groups are disclosed in International Patent Application PCT/US92/09196, filed Oct.
  • Conjugate moieties include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al. (1989) Proc. Natl. Acad. Sci. USA 86:6553-6556), cholic acid (Manoharan et al. (1994) Bioorg. Med. Chem. Let. 4:1053-1060), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al. (1992) Ann. N.Y. Acad. Sci. 660:306-309; Manoharan et al. (1993) Bioorg. Med. Chem. Let.
  • lipid moieties such as a cholesterol moiety (Letsinger et al. (1989) Proc. Natl. Acad. Sci. USA 86:6553-6556), cholic acid (Manoharan et al. (1994) Bioorg. Med. Chem. Let. 4:1053-10
  • a phospholipid e.g., di-hexadecyl-rac-glycerol or triethyl-ammonium 1,2-di-O- hexadecyl-rac-glycero-3-H-phosphonate (Manoharan et al. (1995) Tetrahedron Lett. 36:3651- 3654; Shea et al. (1990) Nucl. Acids Res. 18:3777-3783), a polyamine or a polyethylene glycol chain (Manoharan et al.
  • the present invention also includes antisense compounds which are chimeric compounds.
  • "Chimeric” antisense compounds or “chimeras,” in the context of this invention, are antisense compounds, particularly oligonucleotides, which contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of an oligonucleotide compound.
  • oligonucleotides typically contain at least one region wherein the oligonucleotide is modified so as to confer upon the oligonucleotide increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid.
  • An additional region of the oligonucleotide may serve as a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids.
  • RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex.
  • RNA target Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of oligonucleotide inhibition of gene expression. Consequently, comparable results can often be obtained with shorter oligonucleotides when chimeric oligonucleotides are used, compared to phosphorothioate deoxyoligonucleotides hybridizing to the same target region.
  • Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.
  • Chimeric antisense compounds of the invention may be formed as composite structures of two or more oligonucleotides, modified oligonucleotides, oligonucleosides and/or oligonucleotide mimetics as described above. Such compounds have also been refened to in the art as hybrids or gapmers. Representative United States patents that teach the preparation of such hybrid structures include, but are not limited to, U.S. Pat. Nos.
  • donor cells comprising immunoglobulin-producing cells or “donor cells comprising immunoglobulin-producing cells” sometimes refened to simply as “donor cells” or “donor blood cells” refers to cells that are capable of producing antibodies when immunized with an antigenic compound.
  • sources of such donor cells suitable for use in the invention include, but are not limited to spleen cells, lymph node cells, bone manow cells, and immortalizing tumor infiltrating lymphocytes.
  • amino acid denotes a molecule containing both an amino group and a carboxyl group.
  • the amino acids are ⁇ -, ⁇ -, ⁇ - or ⁇ -amino acids, including their stereoisomers and racemates.
  • L-amino acid denotes an ⁇ -amino acid having the L configuration around the ⁇ -carbon, that is, a carboxylic acid of general formula CH(COOH)(NH 2 )-(side chain), having the L-configuration.
  • D-amino acid similarly denotes a carboxylic acid of general formula CH(COOH)(NH 2 )-(side chain), having the D-configuration around the ⁇ -carbon.
  • Side chains of L-amino acids include naturally occurring and non-naturally occurring moieties.
  • Non- naturally occurring (i.e., unnatural) amino acid side chains are moieties that are used in place of naturally occurring amino acid side chains in, for example, amino acid analogs. See, for example, Lehninger, BIOCHEMISTRY, Second Edition, Worth Publishers, Inc., 1975, pages 72- 77 (incorporated herein by reference).
  • polynucleotide refers to a nucleic acid molecule and includes genomic DNA cDNA, RNA, mRNA and the like.
  • inhibitor of mismatch repair refers to an agent that interferes with at least one function of the mismatch repair system of a cell and thereby renders the cell more susceptible to mutation.
  • hypomutable refers to a state in which a cell in vitro or in vivo is made more susceptible to mutation through a loss or impairment of the mismatch repair system.
  • agents chemicals
  • inhibitors when used in connection with inhibition of MMR refers to chemicals, oligonucleotides, RNA interference molecules, analogs of natural substrates, and the like that interfere with normal function of MMR.
  • about refers to an amount within a range of +/- 10% of the cited value.
  • mitogenic polypeptide refers to a polypeptide when in combination with the antigen provides stimulation of appropriate cells to increase the immune response against the subject antigen.
  • hybrida refers to the result of a cell fusion between an immunoglobulin-producing cell and a transformed cell, such as a myeloma cell.
  • IgG subclass refers to a category of immunoglobulins comprising IgGl, IgG2, IgG2a, IgG2b, IgG3, and IgG4.
  • mismatch repair gene refers to a gene that encodes one of the proteins of the mismatch repair complex.
  • a mismatch repair complex is believed to detect distortions of the DNA helix resulting from non-complementary pairing of nucleotide bases. The non- complementary base on the newer DNA strand is excised, and the excised base is replaced with the appropriate base which is complementary to the older DNA strand. In this way, cells eliminate many mutations that occur as a result of mistakes in DNA replication. Dominant negative alleles cause a mismatch repair defective phenotype even in the presence of a wild- type allele in the same cell.
  • a non-limiting example of a dominant negative allele of a mismatch repair gene is the human gene hPMS2-134, which carries a truncation mutation at codon 134.
  • the mutation causes the product of this gene to abnormally terminate at the position of the 134th amino acid, resulting in a shortened polypeptide containing the N- terminal 133 amino acids.
  • Such a mutation causes an increase in the rate of mutations which accumulate in cells after DNA replication.
  • expression of a dominant negative allele of a mismatch repair gene results in impairment of mismatch repair activity, even in the presence of the wild-type allele.
  • HAT-sensitive refers to a lethal effect on cells when cultured in medium containing hypoxanthine, aminopterin and thymidine.
  • EBV-negative refers to lack of infection of Epstein-Ban vims in a cell as measured by production of EBNA protein, or detection of EBV nucleic acids.
  • Ig-negative refers to lack of production in a cell of any light or heavy chains of immunoglobulins.
  • screening refers to an assay to assess the genotype or phenotype of a cell or cell product including, but not limited to nucleic acid sequence, protein sequence, protein function (e.g., binding, enzymatic activity, blocking activity, cross-blocking activity, neutralization activity, and the like). The assays include ELISA-based assays, Biacore analysis, and the like.
  • isolated refers to a nucleic acid or protein that has been separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials which would interfere with diagnostic or therapeutic uses for the polypeptide, and may include enzymes, hormones, and other proteinaceous or non- proteinaceous solutes.
  • the nucleic acid or protein is purified to greater than 95% by weight of protein. In other embodiments, the nucleic acid or protein is purified to greater than 99% by weight of protein.
  • Determination of protein purity may be by any means known in the art such as the Lowry method, by SDS-PAGE under reducing or non-reducing conditions using a stain such as a Coomassie blue or silver stain.
  • Purification of nucleic acid may be assessed by any known method, including, but not limited to spectroscopy, agarose or polyacrylamide separation with fluorescent or chemical staining such as methylene blue, for example.
  • the invention provides an in vitro immunization method for obtaining antigen-specific immunoglobulin producing cells wherein the cells produce immunoglobulins of the IgG subclass, and cells produced by this method.
  • the in vitro immunization procedure comprises combining donor cells with an immunogenic antigen in culture.
  • the buffy coat of donor cells is used.
  • the donor may be from any source, including, but not limited to cord blood, venous blood, and the like.
  • the source of the blood cells may be from any animal producing immune cells, particularly mammals. Non-limiting examples of blood cell sources include, mice, rats, humans, monkeys, dogs, cats, horses, pigs, sheep, goats, rabbits, birds, cows, guinea pigs and fish.
  • the blood or buffy coat may be further enriched for lymphocytes by any known method, such as, but not limited to differential centrifugation, filtration, and the like.
  • Donor cells such as peripheral blood mononuclear cells (PBMC) may be incubated in L-leucyl-L-lysine methyl ester hydrobromide (LLOMe). While not wishing to be bound by any particular theory of operation, LLOme is believed to lysosomotropic and specifically kills cytotoxic cells in the PBMC pool such as NK cells, cytotoxic T cells, and CD8+ suppressor T cells, while not having an effect on B cells, T helper cells accessory cells and fibroblasts (Bonebaeck (1988) Immunol. Today 9(11):355-359). Generally, the PBMCs may be incubated with LLOMe for a period of 1-30 minutes.
  • the incubation is performed for 10-20 minutes. In other embodiments, the incubation is performed for 15 minutes.
  • the LLOMe is generally a component of culture medium, such as, for example, RPMI 1640, and is provided in a concentration of about 0.10 to ImM. In some embodiments, LLOMe is provided in an amount of about 0.10 to 0.50 mM. In other embodiments, LLOMe is provided in an amount of about 0.25 mM.
  • the antigen may be any antigen provided that it is immunogenic.
  • Whole proteins or peptides may be used.
  • membrane preparations including those from tumors
  • lymphoma cells whole cells
  • single cells homogenized cells
  • pathogens inclusion bodies
  • cell lysates protein preparations
  • minced tissue including tumor tissue.
  • Whole proteins may be in native or denatured conformation.
  • Peptides may be conjugated to carrier molecules to provide immunogenicity. While not wishing to be bound by any particular theory of operation, carrier molecules may provide additional T cell epitopes which may be useful in stimulating a more robust in vitro antibody response.
  • Examples of carriers that are suitable for use in the method of the invention include tetanus toxoid, diptheria toxin, thyroglobulin, cholera toxin, BCG, bovine serum albumen (BSA), ovalbumin (OVA), and the like. These carriers are refened to herein as "mitogenic polypeptides.”
  • Antigens may be conjugated to mitogenic polypeptides in any way known in the art.
  • fusion proteins may be generated by expressing a polypeptide in a recombinant expression system comprising the polynucleotide encoding at least a portion of the antigen joined in-frame to a polynucleotide encoding at least a portion of the mitogenic polypeptide.
  • the fusion protein may have the mitogenic polypeptide joined at either the amino- or carboxy terminus of the antigen.
  • more that one antigen may be expressed as a fusion protein in combination with a mitogenic polypeptide.
  • more than one mitogenic polypeptide may be expressed as a fusion protein with the antigen or antigens.
  • more than one mitigenic polypeptide and more than one antigen may be expressed together as a single fusion protein.
  • At least a portion of the mitogenic polypeptide is conjugated to at least a portion of the antigen using chemical cross-linkers.
  • chemical cross-linkers include, but are not limited to gluteraldehyde, formaldehyde, 1,1 -bis (diazoacetyl)-2-phenylethane, N-hydroxysuccinimide esters (e.g., esters with 4-azidosalicylic acid, homobifunctional imidoesters including disuccinimidyl esters such as 3,3'-dithiobis (succinimidyl-propionate). and bifunctional maleimides such as bis-N-maleimido-l,8-octane).
  • Derivatizing agents such as methyl-3-[(p-azido-phenyl)dithio] propioimidate yield photoactivatable intermediates which are capable of forming cross-links in the presence of light.
  • a lysine residue in the mitogenic polypeptide or antigen may be coupled to a C-terminal or other cysteine residue in the antigen or mitogenic polypeptide, respectively, by treatment with N- ⁇ -maleimidobutyryloxy-succinimide (Kitagawa and Aikawa (1976) J. Biochem. 79, 233-236).
  • a lysine residue in the mitogenic polypeptide or antigen may be conjugated to a glutamic or aspartic acid residue in the antigen or mitogenic polypeptide, respectively, using isobutylchloroformate (Thorell and De Larson (1978) RADIOIMMUNOASSAY AND RELATED TECHNIQUES: METHODOLOGY AND CLINICAL APPLICATIONS, p. 288).
  • isobutylchloroformate Thiorell and De Larson (1978) RADIOIMMUNOASSAY AND RELATED TECHNIQUES: METHODOLOGY AND CLINICAL APPLICATIONS, p. 288).
  • Other coupling reactions and reagents have been described in the literature.
  • the conditions for the in vitro immunization procedure comprise incubating the cells at about 25-37°C, (preferably 37°C ) supplied with about 5-10% CO 2 . In some embodiments, the incubation is performed with between about 6-9% CO 2 . In other embodiments the incubation is performed in about 8% CO 2 .
  • the cell density is between about 2.5 to 5 x 10 6 cells/ml in culture medium. In some embodiments, the culture medium is supplemented with about 2- 20% FBS. In other embodiments, the culture medium is supplemented with about 5-15% FBS. In other embodiments, the culture medium is supplemented with about 7-12% FBS. In other embodiments, the culture medium is supplemented with about 10% FBS.
  • the in vitro stimulation culture medium is supplemented with cytokines to stimulate the cells and increase the immune response.
  • cytokines In general IL-2 is supplied in the culture medium.
  • other cytokines and additives may also be included to increase the immune response.
  • cytokines and factors may include, for example, LL-4 and anti-CD40 antibodies.
  • the fusion of myeloma cells with the immunoglobulin-producing cells may be by any method known in the art for the creation of hybridoma cells. These methods include, but are not limited to, the hybridoma technique of Kohler and Milstein, (1975, Nature 256:495-497; and U.S. Patent No. 4,376,110) (see also, Brown et al. (1981) J. Immunol. 127:539-546; Brown et al. (1980) J. Biol. Chem. 255 (ll):4980-4983; Yeh et al. (1976) Proc. Natl. Acad. Sci. USA 76:2927-2931; and Yeh et al.
  • the hybridoma producing the mAb of this invention may be cultivated in vitro or in vivo.
  • any myeloma cell may be used in the method of the invention.
  • the myeloma cells are human cells, but the invention is not limited thereto or thereby.
  • the cells are sensitive to medium containing hypoxanthine, aminopterin, an thymidine (HAT medium).
  • HAT medium hypoxanthine, aminopterin, an thymidine
  • the myeloma cells do not express iimnunoglobulin genes.
  • the myeloma cells are negative for Epstein- Ban virus (EBV) infection.
  • EBV Epstein- Ban virus
  • the myeloma cells are HAT-sensitive, EBV negative and Ig expression negative. Any suitable myeloma may be used.
  • Murine hybridomas may be generated using mouse myeloma cell lines (e.g., the P3-NSl/l-Ag4-l, P3- x63-Ag8.653 or Sp2/O-Agl4 myeloma lines). These murine myeloma lines are available
  • the hybridoma cells and/or mammalian expression cells may be rendered hypermutable by the introduction of a dominant negative allele of a mismatch repair gene.
  • the dominant negative allele of the mismatch repair gene may be introduced into the hybridoma cell (i.e., after the fusion of immunoglobulin-producing cells with the myeloma cells) or may be introduced into the myeloma cell prior to the fusions.
  • the invention therefore, also provides hypermutable myeloma cells for use in the generation of hybridoma cells.
  • the dominant negative allele may also be introduced into the mammalian expression cells.
  • the dominant negative allele of the mismatch repair gene is in the form of a polynucleotide which may be in the form of genomic DNA, cDNA, RNA, or a chemically synthesized polynucleotide.
  • the polynucleotide can be cloned into an expression vector containing a constitutively active promoter segment (such as, but not limited to, CMV, SV40, EF-1 Dor LTR sequences) or to inducible promoter sequences such as those from tefracycline, or ecdysone/glucocorticoid inducible vectors, where the expression of the dominant negative mismatch repair gene can be regulated.
  • the polynucleotide can be introduced into the cell by transfection.
  • Transfection is any process whereby a polynucleotide is introduced into a cell.
  • the process of transfection can be carried out in vitro, e.g., using a suspension of one or more isolated cells in culture.
  • the cell can be any immortalized cell used for creating hybridomas for the production of monoclonal antibodies, or the cell may be the hybridoma itself.
  • the hybridomas may be heterohybridoma cells (e.g. human-mouse cell fusions) or homohybridoma cells (e.g., human-human hybridoma cells and mouse-mouse hybridoma cells).
  • transfection will be carried out using a suspension of cells, or a single cell, but other methods can also be applied as long as a sufficient fraction of the treated cells or tissue incorporates the polynucleotide so as to allow transfected cells to be grown and utilized.
  • the protein product of the polynucleotide may be transiently or stably expressed in the cell.
  • Techniques for transfection are well known. Available techniques for introducing polynucleotides include but are not limited to electroporation, transduction, cell fusion, the use of calcium chloride, and packaging of the polynucleotide together with lipid for fusion with the cells of interest.
  • the dominant negative allele of the mismatch repair gene may be derived from any known mismatch repair gene including, but not limited to PMS2, PMSl, PMSR3, PMSR2, PMSR6, MLHl, GTBP, MSH3, MSH2, MLH3, or MSH1, and homologs of PMSR genes as described in Nicolaides et ⁇ l (1995) Genomics 30:195-206 and Horii et ⁇ l (1994) Biochem. Biophys. Res. Commun.
  • Dominant negative alleles refers to the ability of the allele to confer a hypermutable status to the cell expressing the allele. Any allele which produces such effect can be used in this invention.
  • the dominant negative alleles of a mismatch repair gene can be obtained from the cells of humans, animals, yeast, bacteria, or other organisms. Dominant negative alleles of mismatch repair genes that are suitable for use in the invention have certain functional characteristics associated with structural features.
  • a non-limiting example of a dominant negative mismatch repair gene is the PMS2 truncation mutant, PMS2-134. This gene contains a mutation which truncates the PMS2 protein after amino acid 133.
  • the lack of the C-terminus in the PMS2 protein is believed to interfere with the binding of PMS2 with Screening cells for defective mismatch repair activity can identify such alleles.
  • Cells from animals or humans with cancer can be screened for defective mismatch repair.
  • Cells from colon cancer patients may be particularly useful.
  • Genomic DNA, cDNA, or mRNA from any cell encoding a mismatch repair protein can be analyzed for variations from the wild type sequence.
  • Dominant negative alleles of a mismatch repair gene can also be created artificially, for example, by producing variants of the hPMS2-134 allele or other mismatch repair genes.
  • Various techniques of site-directed mutagenesis can be used.
  • the suitability of such alleles, whether natural or artificial, for use in generating hypermutable cells or animals can be evaluated by testing the mismatch repair activity caused by the allele in the presence of one or more wild-type alleles, to determine if it is a dominant negative allele.
  • Dominant negative alleles of such genes when introduced into cells or transgenic animals, increase the rate of spontaneous mutations by reducing the effectiveness of DNA repair and thereby render the cells or animals hypermutable.
  • the degree of elevation of the spontaneous mutation rate can be at least 2-fold, 5-fold, 10-fold, 20-fold, 50-fold, 100-fold, 200-fold, 500-fold, or 1000-fold that of the normal cell or animal.
  • the hypermutable hybridoma cells will accumulate new mutations in gene(s) to produce new output traits within the hybridoma.
  • the hybridoma cells can be screened for desired characteristics and cell lines bearing these characteristics may be expanded.
  • the hybridoma cells may be "cured" of the mismatch repair defect by eliminating the dominant negative mismatch repair gene in the cell or by turning of its expression, leading to stable biological products consisting of altered genes, RNAs, or polypeptides.
  • the dominant negative alleles of the mismatch repair gene may be introduced as part of a vector.
  • the polynucleotide encoding the dominant negative mismatch repair protein allele may be operably linked to a promoter that functions in the cell to drive expression of the dominant negative allele of the mismatch repair gene.
  • Other elements of the vector may include an origin of replication, one or more selectable markers, such as a drug resistance gene that allows the cells to grow in the presence of a growth inhibitory compound.
  • the invention may further comprise the step of restoring genetic stability of the hybridoma by introducing a wild-type mismatch repair gene into the cell to complement the deficiency and restore genetic stability.
  • Another aspect of the invention is the use of cells lacking MMR (either due to defects in endogenous mismatch repair genes, or due to the introduction of a dominant negative MMR gene) and chemical mutagens to cause an enhanced rate of mutations in a host's genome. The lack of MMR activity has been known to make cells more resistant to the toxic effects of DNA damaging agents.
  • This invention comprises making proficient MMR cells mismatch repair defective via the expression of a dominant negative MMR gene allele and then enhancing the genomic hypermutability with the use of a DNA mutagen.
  • Chemical mutagens are classifiable by chemical properties, e.g., alkylating agents, cross-linking agents, etc.
  • N-ethyl-N-nitrosourea ENU
  • N-methyl-N-nitrosourea MNU
  • procarbazine hydrochloride chlorambucil, cyclophosphamide, methyl methanesulfonate (MMS), ethyl methanesulfonate (EMS), diethyl sulfate, acrylamide monomer, triethylene melamin (TEM), melphalan, nitrogen mustard, vincristine, dimethylnitrosamine, N-methyl-N'-nitro- nitrosoguanidine (MNNG), 7,12 dimethylbenz (a) anthracene (DMBA), ethylene oxide, hexamethylphosphoramide, bisulfan.
  • a mutagenesis technique is employed that confers a mutation rate in the range of 1 mutation out of every 100 genes; 1 mutation per 1,000 genes.
  • MMR deficiency and chemical mutagens will allow for the generation of a wide anay of genome alterations (such as but not limited to expansions or deletions of DNA segments within the context of a gene's coding region, a gene's intronic regions, or 5'or 3' proximal and/or distal regions, point mutations, altered repetitive sequences) that are preferentially induced by each particular agent.
  • Mutations can be detected by analyzing for alterations in the genotype of the cells or animals, for example by examining the sequence of genomic DNA, cDNA, messenger RNA, or amino acids associated with the gene of interest. Mutations can also be detected by screening the phenotype of the gene. An altered phenotype can be detected by identifying alterations in electrophoretic mobility, spectroscopic properties, or other physical or structural characteristics of a protein encoded by a mutant gene. One can also screen for altered function of the protein in situ, in isolated form, or in model systems. One can screen for alteration of any property of the cell or animal associated with the function of the gene of interest, such as but not limited to measuring protein secretion, chemical-resistance, pathogen resistance, etc.
  • inducible vectors that control the expression of a dominant negative and normally functioning MMR gene are used.
  • This strategy restores DNA stability once a host cell or organism exhibiting a new output trait, altered gene, RNA or polypeptide has been generated via trait selection with or without the combination of chemical mutagens to establish a genetically stable version of this cell or organism.
  • MMR defective cells as a result of ectopically expressing a dominant negative MMR gene allele, the MMR activity is decreased or completely eliminated by removing the inducer molecule from the cell culture or organism's environment.
  • the expression of a dominant negative MMR gene can be suppressed by knocking out the MMR gene allele using methods that are standard to those skilled in the art of DNA knockout technology in germ or somatic cells (Waldman et al. (1995) Cancer Res. 55:5187-5190).
  • the chiral position of the side chains of the anthracenes is not particularly limited and may be any chiral position and any chiral analog.
  • the anthracenes may also comprise a stereoisomeric form of the anthracenes and include any isomeric analog.
  • hosts are but not limited to cells or whole organisms from human, primate, mammal, rodent, plant, fish, reptiles, amphibians, insects, fungi, yeast or microbes of prokaryotic origin.
  • lymphocytes were isolated from whole blood by centrifugation through Ficoll-Paque according to the manufacturer's instructions. Isolated lymphocytes were incubated with 0.25mM Leu-Leu methyl ester hydrobromide (LLOMe) prepared in RPMI 1640 medium containing 2% fetal bovine serum (FBS) for 15 minutes at room temperature. The cells were then washed three times with culture medium.
  • LLOMe Leu-Leu methyl ester hydrobromide
  • FBS fetal bovine serum
  • the cells were incubated at 37°C in a incubator, supplied with 8% CO 2 , at a density between 2.5 to 5 x 10 cells/ml in culture medium supplemented with 10% FBS and TT and IL-2 at various concentrations. After four days of culture, the cells were washed four times with medium and the culture was continued for additional eight days.
  • TT or BSA at 0.5 ⁇ g/ml in 0.05 M carbonate-bicarbonate buffer was immobilized onto an EIA plate. After blocking with 1% bovine serum albumin (BSA) in PBS containing 0.05% Tween 20, the supernatant was added to the wells. Antibodies bound to TT were detected with peroxidase-labeled goat anti-human IgG or anti-human IgM. TMB was used for color development. The plate was read using a Microplate reader with a 450 nm filter. A supernatant sample that had antibody bound to TT, but not to BSA, and in which the signal was two times the assay background was considered positive. The positive cells were pooled, and used for hybridoma production (Fig. 1).
  • peripheral blood mononuclear cells from some donors contain a fraction of B cells that secret TT-specific antibodies in culture. This is due to the fact that about 90%) of the population in the United States has been vaccinated against TT. Such sera also has a titer of higher than 1000 (Fig. 2). However, the percentage of positive events is greatly increased when PBMCs are immunized in vitro with TT (Fig. 3). The intensity of the PBMC response is also enhanced with the stimulation of TT alone or in combination with IL-2 or CD40L (Fig. 4). Similar effects were observed with other antigens (data not shown). [0227] Generation of hybridomas secreting human antibodies.
  • mice were pooled and cultured in T flasks at 0.5 - 1 x 10 6 cells/ml in culture medium supplemented with 10% FBS one day prior to the fusion.
  • mouse myeloma NS0 cells were transfected with human PMS2-134 expression vector as described in Nicolaides et al. (1998) Mol. Cell. Biol. 18(3):1635-1641.
  • the cells were cultured in RPMI 1640 supplemented with 10% FBS and 2 mM glutamine (Complete Medium) and the culture was kept in log phase.
  • lymphocytes were harvested and counted. An equal number of myeloma cells was harvested. Both types of cells were combined and washed three times with RPMI 1640 medium. Polyethylene glycol (PEG) was added dropwise to the loosened cell pellet, and the PEG was subsequently diluted out slowly with 25 ml of RPMI medium in a course of 2.5 minutes. After diluting out the PEG, fused cells were suspended in Complete Medium supplemented with HAT and 20% FBS, and seeded onto 96-well plates. [0229] Screening and characterization of antigen-specific hybridoma clones.
  • PEG Polyethylene glycol
  • hybridomas derived from TT- immunized lymphocytes were tested. Briefly, TT or BSA at 0.5 ug/ml in 0.05 M carbonate- bicarbonate buffer was immobilized onto the EIA plate. After blocking with 1% bovine serum albumen in PBS containing 0.05% ⁇ Tween 20, the cell culture supernate was added to the wells. Antibodies bound to TT were detected with peroxidase-labeled goat anti-human IgG or anti-human IgM. TMB was used for color development.
  • the plate was read in the Microplate reader with a 450 nm filter. A cell clone that showed reactivity to TT but not to BSA was considered positive (Fig. 5). Positive clones were expanded and subcloned by limiting dilution to generate monoclonal cells.
  • Example 2 Generation of hybridomas secreting human monoclonal antibodies to epidermal growth factor receptor (EGFR) (self antigen)
  • EGFR epidermal growth factor receptor
  • EGFR Human epidermal growth factor receptor
  • A431 cells Human epidermal growth factor receptor
  • EGFR-TT tetanus toxin C
  • TT-C C fragment
  • TT-C was dissolved in sterile MilliQ-grade water to yield a 2 mg/ml TT-C solution.
  • Crosslinking was performed in 50 mM sodium carbonate buffer pH 9.0 at equimolar ratios of EGFR to TT-C, using glutaraldehyde at a final concentration of 0.5% for 3 hours at room temperature, followed by 4°C overnight.
  • Glutaraldehyde was quenched by addition of a fresh 100 mg/ml solution of sodium borohydride in 50 mM sodium carbonate pH 9.0, under open atmosphere for 1 hour at 4°C.
  • Crosslinked products were dialyzed against Ca 2+ -, Mg 2+ - free phosphate-buffered saline overnight at 4°C, using 3.5K MWCO Slide- A-Lyzer cassettes.
  • the reaction was monitored by Western blotting, using commercial anti-EGFR (mAb- 15) and anti-TT-C (Roche) monoclonal antibodies. By this method, greater than 70%) of the components are crosslinked, and appear as immunoreactive species of greater MW than the starting material (data not shown).
  • PBMC peripheral blood mononuclear cells
  • EGFR-TT at a concentration of 50 ng/ml with or without recombinant human IL-2 at 20 IU, mouse anti-human CD40 antibody as CD40L at 0.5 ug/ml (used to enhance IgG class switching).
  • the cells were re-fed with complete medium, in the absence of added stimulus, every three or four days. Culture supernatants were collected on days 12-18 and tested for EGFR-specific antibodies.
  • EGFR-specific antibody response The PBMC response to the stimulation was examined in a EGFR-specific ELISA. Briefly, EGFR, TT, or BSA at 0.5 ug/ml in 0.05 M carbonate-bicarbonate buffer, pH 9.6, was immobilized onto EIA plates. After blocking the plates with 5% non-fat dry milk in PBS containing 0.05% Tween 20, the supernatant was added to the wells. Antibodies from the supernatant bound to immobilized antigens were detected with peroxidase-labeled goat anti-human IgG+IgM (H+L). TMB substrate kit was used for color development.
  • the plates were read in a Microplate reader with a 450 nm filter. A supernatant sample containing antibody that bound to EGFR, but not to TT and BSA, was considered positive. A robust response was observed in cultures immunized to the EGFR-TT as compared to controls. While anti-EGFR responses were observed in PBMCs for a small fraction of donors, the percentage of positive clones was greatly increased when PBMCs were immunized in vitro with EGFR complexed with TT (Fig. 6). Positive cells were pooled and used for hybridoma production.
  • Hybridomas To prepare activated lymphocytes, cells were pooled and cultured in T flasks at 0.5 - 1 x 10 6 cells/ml in culture medium supplemented with 10% FBS one day prior to the fusion. To prepare the fusion partner, mouse myeloma NS0 cells were transfected with human PMS2-134 expression vector as described in Nicolaides et al. (1998) Mol. Cell. Biol. 18(3): 1635-1641. The cells were cultured in RPMI 1640 supplemented with 10% FBS and 2 mM glutamine (Complete Medium) and the culture was kept in log phase. [0235] Screening and characterization of antigen-specific hybridoma clones.
  • hybridomas derived from EGFR- immunized lymphocytes were tested. Briefly, EGFR, TT TNFRl or BSA at 0.5 ug/ml in 0.05 M carbonate-bicarbonate buffer was immobilized onto the EIA plate. After blocking with 1% bovine serum albumen in PBS containing 0.05% Tween 20, the cell culture supernate was added to the wells. Antibodies bound were detected with peroxidase-labeled goat anti-human IgG or anti-human IgM. TMB was used for color development.
  • the pellet was resuspended in 10 ml ACK Lysing Buffer and incubated for 5 minutes at room temperature. Following lysis, 35 ml PBS " " was added to the tubes and the tubes were centrifuged at 1000 rpm for 7 minutes. The cells were then washed with 45 ml RPMI medium.
  • the non-adherent cell culture was centrifuged at 1000 rpm for 7 minutes. The supernatant fluid was aspirated and the pellet was resuspended in 10 ml RPMI supplemented with 2% FBS and freshly thawed 85 ⁇ g/ml LLOMe. The cells were incubated for 15 minutes at room temperature. The cells were washed twice with cRPMI and resuspended in 45 ml cRPMI. The cells were transfened to an upright T25 flask at a density of 5 x 10 cells/ml in cRPMI supplemented with 2 ⁇ g/ml PHA and incubated at 37°C/8% CO 2 for 24 hours.
  • the non-adherent cells were harvested, centrifuged at 1000 rpm for 7 minutes, and the cell pellet was resuspended in 5 ml cold cRPMI/5%DMSO.
  • the tubes containing the cells were wrapped in paper towels and stored at -80° C until needed.
  • Cells were distributed in a culture plate at a density of 1 x 10 6 cells/well. A tumor sample was chopped into fine pieces of approximately 1-3 mm 3 . An aliquot of the tumor suspension was transfened to all but 1 well, titrating the amount of tumor per well. An aliquot of 0.25 ml cRPMI was added to the control well. The total volume in the wells was 0.5ml/well. The dendritic cells and tumor cells were co-cultured at 37°C/8% CO 2 for 24 hours.
  • Frozen PBMC were thawed by adding 40 ml cRPMI/30 JU/ml IL-2/600 JU/ml IL- 4/0.75 ⁇ g/ml CD-40L pre-warmed to 50°C to the frozen cells. When thawed, the cells were incubated for 1-2 hours at 37°C. The cells were centrifuged at 1000 rpm for 7 minutes and the pellet was resuspended in 5 ml of a 2X cocktail of cRPMI/ 60 JU/ml IL-2/1200 IU/ml IL- 4/1.5 ⁇ g/ml CD-40L.
  • the cell suspension was divided among wells in a tissue culture plate at 0.5 ml/well of suspension and diluted with 0.5 ml medium for a final concentration of 30 IU/ml IL-2, 600 IU/ml IL-4, and 0.75 ⁇ g/ml CD-40L.
  • Cells were fed with cRPMI supplemented with 20 IU/ml IL-2, 400 JU/ml IL-4, 100 IU/ml IL-10, and 0.5 ⁇ g/ml CD-40L.
  • Tumor-immunized PBMCs were then fused with A6 myeloma cells to generate hybridomas. Briefly, lymphocytes were harvested from 75% tumor and 100% tumor wells, rinsed with 1 ml RPMI, transfer to conical tubes, and the volume was adjusted to 5 ml with cRPMI. The cells were centrifuged through Ficoll-Paque, and the supernatant fluid was aspirated. Interfaces containing cells from all tubes were combined and the cells were rinsed with cRPMI. The cells were then resuspended in 7.5 ml cRPMI. Viable cells were assessed by trypan blue exclusion. A6 cell viability was also assessed by trypan blue exclusion.
  • A6 cells and tumor-immunized lymphocytes were centrifuged separately at 1200 m for lOminutes. The supernatant fluids were aspirated and the cells were washed with 10ml DPBS "7 Jtube. Each cell line was washed three times with 2 ml cold Mannitol Fusion Medium (MFM) (0.3M Mannitol, 0.18mM MgCl 2 , 0.18mM CaCl 2 , ImM Hepes) and the cells were combined and resuspended in MFM at a density of 3 x 10 6 A6 cells and 3 x 10 6 PBMCs in 200 ⁇ l for a total of 6 x 10 6 cells in 200 ⁇ l.
  • MFM cold Mannitol Fusion Medium
  • BTX 450 microslides were sterilized with 65 ⁇ L 100% EtOH and pre-wetted with 65 ⁇ l MFM. A 40 ⁇ l aliquot of cell suspension was distributed evenly onto a BTX 450-1 microslide.
  • the ECM 2001 conditions were set as follows: alignment conditions, 20V for 30 seconds; pulse conditions, 150V for 30 ⁇ seconds (IX); compression conditions, 20V for 9 seconds.
  • the cells were transfened to one well of a 24 well plate containing 1 ml phenol red-free cRPMI. The fusion steps were repeated for the remaining cell suspensions, rinsing slide between fusions with 65 ⁇ L MFM. The culture plate containing fused cell cultures was incubated overnight at 37°C/8%CO 2 .
  • the fused cells were cloned and assessed by ELISA for IgG and IgM production. The results are shown in Fig. 8.
  • Purified GM-CSF from a commercial source is administered in vitro to peripheral blood mononuclear cells (PBMC).
  • PBMC peripheral blood mononuclear cells
  • Lymphocytes are isolated from whole blood by centrifugation through Ficoll-Paque according to the manufacturer's instructions. Isolated lymphocytes are incubated with 0.25 mM Leu-Leu methyl ester hydrobromide (LLOMe) prepared in RPMI 1640 medium containing 2% fetal bovine serum (FBS) for 15 minutes at room temperature. The cells are then washed three times with culture medium.
  • LLOMe Leu-Leu methyl ester hydrobromide
  • FBS fetal bovine serum
  • the cells are incubated at 37 C in an incubator, supplied with 8% CO 2 , at a density between 2.5 to 5 x 10 6 cells/ml in culture medium supplemented with 10% FBS and GM-CSF and IL-2 at various concentrations. After four days of culture, the cells are washed four times with medium and the culture was continued for additional eight days.
  • Lymphocyte culture supematants are collected on day 12 of the culture and tested in an ELISA for the presence of anti-GM-CSF antibodies. Briefly, GM-CSF or BSA at 0.5 ⁇ g/ml in 0.05 M carbonate-bicarbonate buffer is immobilized onto an EIA plate. After blocking with 1% bovine serum albumin (BSA) in PBS containing 0.05% Tween 20, the supernatant is added to the wells. Antibodies bound to GM-CSF are detected with peroxidase-labeled goat anti-human IgG or anti-human IgM. TMB is used for color development. The plate is read using a Microplate reader with a 450 nm filter. A supernatant sample that had antibody bound to GM-CSF, but not to BSA, and in which the signal was two times the assay background is considered positive. The positive cells are pooled, and used for hybridoma production.
  • BSA bovine serum albumin
  • activated lymphocytes cells are pooled and cultured in T flasks at 0.5 - 1 x 10 6 cells/ml in culture medium supplemented with 10% FBS one day prior to the fusion.
  • mouse myeloma NS0 cells are transfected with human PMS2-134 expression vector as described in Nicolaides et al. (1998) Mol. Cell. Biol. 18(3):1635-1641.
  • the cells are cultured in RPMI 1640 supplemented with 10% FBS and 2 mM glutamine (Complete Medium) and the culture is kept in log phase.
  • lymphocytes are harvested and counted. An equal number of myeloma cells is harvested. Both types of cells are combined and washed three times with RPMI 1640 medium. Polyethylene glycol (PEG) is added dropwise to the loosened cell pellet, and the PEG is subsequently diluted out slowly with 25 ml of RPMI medium in a course of 2.5 minutes. After diluting out the PEG, fused cells are suspended in Complete Medium supplemented with HAT and 20% FBS, and seeded onto 96-well plates.
  • PEG Polyethylene glycol
  • hybridomas derived from TT-immunized lymphocytes are tested. Briefly, TT or BSA at 0.5 ug/ml in 0.05 M carbonate-bicarbonate buffer is immobilized onto the EIA plate. After blocking with 1% bovine serum albumin in PBS containing 0.05% Tween 20, the cell culture supernate is added to the wells. Antibodies bound to GM-CSF are detected with peroxidase-labeled goat anti- human IgG or anti-human IgM. TMB is used for color development.
  • the plate is read in the Microplate reader with a 450 nm filter.
  • a cell clone that showed reactivity to GM-CSF but not to BSA is considered positive. Positive clones are expanded and subcloned by limiting dilution to generate monoclonal cells.
  • Example 5 Generation of hybridomas secreting human monoclonal antibodies to GM- CSF-KLH
  • GM-CSF Human GM-CSF was purchased from a vendor. In order to enhance immunization, GM-CSF was conjugated to keyhole limpet hemocyanin (KLH) (GM-CSF-KLH) and the conjugate was used as immunogen for in vitro immunization in order to overcome any immunotolerance.
  • KLH keyhole limpet hemocyanin
  • Purified GM-CSF was reconstituted in sterile MilliQ-grade water to yield a lmg/ml solution.
  • Purified, lyophilized recombinant KLH was dissolved in sterile MilliQ-grade water to yield a 1 mg/ml KLH solution.
  • a 0.2% solution of glutaraldehyde in PBS was prepared.
  • Crosslinking was performed by combining 25 ul of 1 mg/ml KLH, 25 ul of 1 mg/ml GM-CSF, and 50 ul 0.2% glutaraldehyde in a microcentrifuge tube wrapped in aluminum foil at room temperature, with shaking for 1 hour.
  • PBMC peripheral blood mononuclear cells
  • LLOMe-pretreated PBMC were incubated at a density of 3 x 10 6 cells/ml in culture medium supplemented with 10%o FBS and a stimuli mixture.
  • the stimuli mixture was composed of GM-CSF-KLH at a concentration of 50 ng/ml with or without recombinant human IL-2 at 20 IU, mouse anti-human CD40 antibody as CD40L at 0.5 ug/ml (used to enhance IgG class switching).
  • the cells were re-fed with complete medium, in the absence of added stimulus, every three or four days. Culture supematants were collected on days 12-18 and tested for GM-CSF-specific antibodies.
  • GM-CSF-specific antibody response The PBMC response to the stimulation was examined in a GM-CSF-specific ELISA. Briefly, GM-CSF, KLH, or chick ovalbumin (CAB) at 0.5 ug/ml in 0.05 M carbonate- bicarbonate buffer, pH 9.6, was immobilized onto EIA plates. After blocking the plates with 1%) BSA containing 0.05% Tween 20, the supernatant were added to the wells. Antibodies from the supernatant bound to immobilized antigens were detected with peroxidase-labeled goat anti-human IgG+IgM (H+L). TMB substrate kit was used for color development.
  • the plates were read in a Microplate reader with a 450 nm filter. A supernatant sample containing antibody that bound to GM-CSF, but not to KLH and CAB, was considered positive. There was a robust response observed in cultures immunized to the GM-CSF-KLH as compared to controls. While anti-GM-CSF responses were observed in PBMCs for a small fraction of donors, the percentage of positive clones was greatly increased when PBMC were immunized in vitro with GM-CSF complexed with KLH. Positive cells were pooled and used for hybridoma production.
  • activated lymphocytes To prepare activated lymphocytes, cells were pooled and cultured in T flasks at 0.5 - 1 x 10 6 cells/ml in culture medium supplemented with 10% FBS one day prior to the fusion.
  • mouse myeloma NSO cells were transfected with human PMS2-134 expression vector as described in Nicolaides et al. (1998) Mol. Cell. Biol. 18(3):1635-1641. The cells were cultured in RPMI 1640 supplemented with 10% FBS and 2 mM glutamine (Complete Medium) and the culture was kept in log phase.
  • hybridomas derived from GM-CSF-immunized lymphocytes were tested. Briefly, GM-CSF, KLH, or CAB at 0.5 ug/ml in 0.05 M carbonate-bicarbonate buffer was immobilized onto the EIA plate. After blocking with 1% bovine serum albumin in PBS containing 0.05% Tween 20, the cell culture supernate was added to the wells. Antibodies bound were detected with peroxidase-labeled goat anti-human IgG or anti-human IgM.
  • TMB was used for color development. Normal human IgG (nhlgG) and IgM (nhlgM) were used as controls.
  • the plate is read in the Microplate reader with a 450 nm filter. A cell clone that showed reactivity to GM-CSF, but not to CAB was considered positive. The results are shown in Fig. 9.
  • TF-1 cells were seeded at 0.2 x 10 6 /ml in RPMI supplemented with 10% FBS and 0.5 ng/ml recombinant human GM-CSF.
  • TF-1 cells were serum starved for 24 hours in medium containing 0.5% BSA, without rhGM-CSF.
  • Cells were then cultured in the presence of 0.275ng/ml of GM-CSF for 3 days, with or without 4 ug/ml of various antibodies.
  • Cell proliferation was measured using the ATPLite assay (Perkin Elmer). In this assay, ATP was released by lysis of viable cells and utilized by the enzyme luciferase to convert luciferin into oxyluciferin.

Abstract

The invention provides methods for generating high titers of high-affinity antibodies from hybridoma cells produced by fusing myeloma cells with in vitro immunized donor cells. The hybridoma cells or mammalian expression cells with cloned antibody genes from the hybridomas producing the high-affinity antibodies may be mismatch repair defective due to defects of endogenous mismatch repair subunits of through expression of a dominant negative allele of a mismatch repair gene which allows the hybridoma cell to be hypermutable, may be rendered hypermutable by chemical means, or may be naturally mismatch repair deficient. High-affinity antibodies and high titer producer cells producing antibodies may be prepared by the methods of the invention.

Description

METHODS OF GENERATING HIGH-PRODUCTION OF ANTIBODIES FROM HYBRIDOMAS CREATED BY IN VITRO IMMUNIZATION
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This Application claims the benefit of U.S. Provisional Application No. 60/427,165 filed November 15, 2002 and U.S. Provisional Application No. 60/501,650 filed September 10, 2003, the disclosures of which are hereby incorporated by reference in their entirety.
FIELD OF THE INVENTION [0002] The invention relates to the generation of hybridoma cells that produce high-affinity antibodies in high titers. More specifically, the invention relates to the use of an in vitro ii riunization method in conjunction with hybridoma technology using dominant negative mismatch repair genes or chemical inhibitors of mismatch repair to produce high titers of antigen specific antibodies of the IgG subclass, that bind to the antigen with high affinity.
BACKGROUND OF THE RELATED ART
[0003] The use of antibodies to block the activity of foreign and/or endogenous polypeptides provides an effective and selective strategy for treating the underlying cause of disease. In particular is the use of monoclonal antibodies (MAb) as effective therapeutics such as the FDA approved ReoPro (Glaser, (1996) Nat. Biotechnol 14:1216-1217), an anti-platelet MAb from Centocor; Herceptin (Weiner, (1999) Semin. Oncol. 26:43-51), an anti-Her2/neu MAb from Genentech; and Synagis (SaezLlorens, et al. (1998) Pediat. Infect Dis. J. 17:787-791), an anti- respiratory syncytial virus MAb produced by Medimmune.
[0004] Standard methods for generating MAbs against candidate protein targets are known by those skilled in the art. Briefly, rodents such as mice or rats are injected with a purified antigen in the presence of adjuvant to generate an immune response (Shield, et al. (1996) Am. J. Kidney Dis. 27: 855-864). Rodents with positive immune sera are sacrificed and splenocytes are isolated. Isolated splenocytes are fused to melanomas to produce immortalized cell lines that are then screened for antibody production. Positive lines are isolated and characterized for antibody production. The direct use of rodent MAbs as human therapeutic agents were confounded by the fact that human anti-rodent antibody (HARA) responses occurred in a significant number of patients treated with the rodent-derived antibody (Khazaeli, et al, (1994) Immunother. 15:42-52). In order to circumvent the problem of HARA, the grafting of the complementarity determining regions (CDRs), which are the critical motifs found within the heavy and light chain variable regions of the immunoglobulin (Ig) subunits making up the antigen binding domain, onto a human antibody backbone found these chimeric molecules are able to retain their binding activity to antigen while lacking the HARA response (Emery and Harris, "Strategies for humanizing antibodies" In: ANTIBODY ENGINEERING, C.A.K. Borrebaeck (Ed.) Oxford University Press, NY, 1995. pp. 159-183. A common problem that exists during the "humanization" of rodent-derived MAbs (referred to hereafter as HAb) is the loss of binding affinity due to conformational changes in the three- dimensional structure of the CDR domain upon grafting onto the human Ig backbone (U. S. Patent No. 5,530,101 to Queen et al). To overcome this problem, additional HAb vectors are usually needed to be engineered by inserting or deleting additional amino acid residues within the framework region and/or within the CDR coding region itself in order to recreate high affinity HAbs (U. S. Patent No. 5,530,101 to Queen et al). This process is a very time consuming procedure that involves the use of expensive computer modeling programs to predict changes that may lead to a high affinity HAb. In some instances the affinity of the HAb is never restored to that of the MAb, rendering them of little therapeutic use. [0005] Another problem that exists in antibody engineering is the generation of stable, high yielding producer cell lines that is required for manufacturing of the molecule for clinical materials. Several strategies have been adopted in standard practice by those skilled in the art to circumvent this problem. One method is the use of Chinese Hamster Ovary (CHO) cells transfected with exogenous Ig fusion genes containing the grafted human light and heavy chains to produce whole antibodies or single chain antibodies, which are a chimeric molecule containing both light and heavy chains that form an antigen-binding polypeptide (Reff, M. E. (1993) Curr. Opin. Biotechnol. 4:573-576). Another method employs the use of human lymphocytes derived from transgenic mice containing a human grafted immune system or transgenic mice containing a human Ig gene repertoire. Yet another method employs the use of monkeys to produce primate MAbs, which have been reported to lack a human anti-monkey response (Neuberger and Gruggermann (1997) Nature 386:25-26). In all cases, the generation of a cell line that is capable of generating sufficient amounts of high affinity antibody poses a major limitation for producing sufficient materials for clinical studies. Because of these limitations, the utility of other recombinant systems such as plants are currently being explored as systems that will lead to the stable, high-level production of humanized antibodies (Fiedler and Conrad (1995) Bio/Technology 13:1090-1093).
[0006] One strategy to overcome the problem of human reactions against foreign antibodies is to stimulate human immunoglobulin-producing cells in vitro. Various attempts to stimulate human antibody production in vitro typically have resulted in low affinity antibodies of the IgM subclass (Zafiropoulos et al. (1997) J. Immunological Methods 200:181-190). [0007] A method for generating diverse antibody sequences within the variable domain that results in HAbs and MAbs with high binding affinities to antigens would be useful for the creation of more potent therapeutic and diagnostic reagents respectively. Moreover, the generation of randomly altered nucleotide and polypeptide residues throughout an entire antibody molecule will result in new reagents that are less antigenic and/or have beneficial pharmacokinetic properties. The invention described herein is directed to the use of random genetic mutation throughout an antibody structure in vitro by blocking the endogenous mismatch repair (MMR) activity of a host cell producing immunoglobulins that encode biochemically active antibodies. The invention also relates to methods for repeated in vitro genetic alterations and selection for antibodies with enhanced binding and pharmacokinetic profiles.
[0008] In addition, the ability to develop genetically altered host cells that are capable of secreting increased amounts of antibody also will provide a valuable method for creating cell hosts for product development. The invention described herein is further directed to the creation of genetically altered cell hosts with increased antibody production via the blockade of MMR. The invention facilitates the generation of high affinity antibodies and the production of cell lines with elevated levels of antibody production derived from hybridoma cells. The invention described herein provides methods for generating antigen-specific monoclonal antibodies (mAbs). Other advantages of the present invention are described in the examples and figures described herein.
SUMMARY OF THE INVENTION
[0009] The invention provides methods for producing hybridoma cells producing high-affinity antibodies from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobuliii-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells express a dominant negative allele of a mismatch repair gene; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; thereby producing hybridoma cells producing high-affinity antibodies.
[0010] In some embodiments, the dominant negative allele of a mismatch repair gene comprises a truncation mutation of the PMS2 gene (e.g., a PMS2-134 gene). In some embodiments of the method of the invention, antibodies are screened using an ELISA-based assay or other assays that can measure antibody-antigen binding. In some embodiments, the screening assays screen for hypermutated hybridomas that produce higher affinity antibodies than those produced by the parental hybridomas. In other embodiments, the screening assays screen for hypermutated hybridomas that produce antibodies in higher titers than the parental hybridomas.
[0011] In some embodiments of the method of the invention, the method further comprises inactivation of the dominant negative allele of the mismatch repair gene, thereby stabilizing the genome of said hypermutated hybridoma.
[0012] In some embodiments of the method of the invention, the dominant negative mismatch repair gene is introduced into the hybridoma cell after the fusion of said myeloma with the immunoglobulin-producing cells. In other embodiments, the dominant negative mismatch repair gene is introduced into the myeloma cell prior to the fusion with the immunoglobulin- producing cells.
[0013] The invention also comprises antibodies produced by the hybridoma cells. [0014] The invention also comprises methods for producing hybridoma cells that produce high titers of antibodies from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor blood cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells express a dominant negative allele of a mismatch repair gene; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen of the hypermutated hybridoma cells for antigen-specific antibodies produced in higher titers than that produced by the parental hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce higher titers of antibodies than that produced by the parental hybridoma cells. [0015] In some embodiments, the dominant negative allele of a mismatch repair gene comprises a truncation mutation of the PMS2 gene (e.g., a PMS2-134 gene). In some embodiments of the method of the invention, antibodies are screened using an ELISA-based assay. In some embodiments, the screening assays screen for hypermutated hybridomas that produce higher affinity antibodies than those produced by the parental hybridomas. In other embodiments, the screening assays screen for hypermutated hybridomas that produce antibodies in higher titers than the parental hybridomas.
[0016] In some embodiments of the method of the invention, the method further comprising inactivation of the dominant negative allele of the mismatch repair gene, thereby stabilizing the genome of said hypermutated hybridoma.
[0017] In some embodiments of the method of the invention, the dominant negative mismatch repair gene is introduced into the hybridoma cell after the fusion of said myeloma with the immunoglobulin-producing cells. In other embodiments, the dominant negative mismatch repair gene is introduced into the myeloma cell prior to the fusion with the immunoglobulin- producing cells.
[0018] In some embodiments of the method of the invention, the dominant negative allele of the mismatch repair gene is subsequently inactivated in order to restabilize the genome of the cell.
[0019] The dominant negative allele of the mismatch repair gene may be introduced into the myeloma cell prior to fusion with the immunoglobulin producing cells. Thus, the resulting hybridoma cells express the same dominant negative allele of the mismatch repair gene as the myeloma cells. Alternatively, the dominant negative allele of the mismatch repair gene may be introduced into the hybridoma cells.
[0020] The invention also comprises antibodies produced by the hybridoma cells. [0021] The invention further provides recombinant myeloma cells comprising a polynucleotide sequence encoding a dominant negative mismatch repair protein. The dominant negative mismatch repair protein may be a dominant negative form of, for example, aPMS2, PMS1, PMSR3, PMSR2, PMSR6, MLH1, GTBP, MSH3, MSH2, MLH3, or MSH1, and PMSR proteins encoded by homologs of the PMSR genes as described in Nicolaides et al. (1995) Genomics 30:195-206 and Horii et al. (1994) Biochem. Biophys. Res. Commun. 204:1257-1264. In some embodiments, the recombinant myeloma cell expresses a polynucleotide encoding a dominant negative allele of a PMS2 gene (e.g., a truncation mutation of the PMS2 gene, such as the PMS2-134 gene). [0022] In some embodiments, the recombinant myeloma cell is a human cell. In other embodiments, the recombinant myeloma cell does not express immunoglobulin genes and/or Epstein-Barr virus. In other embodiments, the myeloma cells are HAT sensitive. [0023] The invention also provides a method for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing said immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for binding of antibodies produced from said hybridoma cells to antigen; (d) cloning immunoglobulin genes from said hybridoma into a mammalian expression cell, wherein said mammalian expression cell expresses a dominant negative allele of a mismatch repair gene; (e) performing a screen for mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from said hybridoma cells; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells.
[0024] In some embodiments, the dominant negative allele of a mismatch repair gene is introduced into said mammalian expression cell prior to introduction of the immunoglobulin genes. In other embodiments, the dominant negative allele of a mismatch repair gene is introduced into said mammalian expression cell after introduction of said immunoglobulin genes. In other embodiments, the dominant negative allele of a mismatch repair gene is introduced into the mammalian expression cell with the immunoglobulin genes simultaneously.
[0025] The invention also comprises antibodies produced by the mammalian expression cells. [0026] The invention also provides a method for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing said immunoglobulin-producing cells with myeloma cells to form hybridoma cells, wherein said hybridoma cells express a dominant negative allele of a mismatch repair gene; (c) incubating said parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from said hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for said antigen than antibodies produced by said parental hybridoma cells; (f) cloning immunoglobulin genes from said hybridoma into a mammalian expression cell; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized human immunoglobulin-producing cells. [0027] In some embodiments, the dominant negative allele of a mismatch repair gene is present in the myeloma cell prior to cell fusion. In other embodiments, the dominant negative allele of the mismatch repair gene is introduced into the hybridoma cell after cell fusion. [0028] The invention also comprises antibodies produced by the mammalian expression cells. [0029] The invention also provides a method for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for binding of antibodies produced from the hybridoma cells to antigen; (d) cloning immunoglobulin genes from the hybridoma into a parental mammalian expression cell, wherein the mammalian expression cell expresses a dominant negative allele of a mismatch repair gene; (e) incubating the parental mammalian expression cell to allow for mutagenesis, thereby forming hypermutated mammalian expression cells; (f) performing a screen of hypermutable mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from the hybridoma cells; and (g) performing a screen of hypermutable mammalian expression cells that secrete higher titers of antibodies than parental mammalian expression cells; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells. [0030] In some embodiments, the dominant negative allele of a mismatch repair gene is introduced into said mammalian expression cell prior to introduction of the immunoglobulin genes. In other embodiments, the dominant negative allele of a mismatch repair gene is introduced into said mammalian expression cell after introduction of said immunoglobulin genes. In other embodiments, the dominant negative allele of a mismatch repair gene is introduced into the mammalian expression cell with the immunoglobulin genes simultaneously.
[0031] The invention also provides antibodies produced by the mammalian expression cells. [0032] The invention also provides recombinant, hypermutable mammalian expression cells comprising a polynucleotide sequence encoding a dominant negative mismatch repair protein. [0033] The mismatch repair gene may be a dominant negative mismatch repair gene, including, but not limited to a dominant negative form of PMS2, PMS1, PMSR3, PMSR2, PMSR6, MLH1, GTBP, MSH3, MSH2, MLH3, or MSH1, and homologs of PMSR genes as described in Nicolaides et al (1995) Genomics 30:195-206 and Horii et al. (1994) Biochem. Biophys. Res. Commun. 204:1257-1264. A non-limiting example includes a dominant negative truncation mutant of PMS2 (e.g., a.PMS2-134 gene).
[0034] The invention also provides methods for producing hybridoma cells producing high- affinity antibodies from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising ύrimunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells; (c) incubating the parental hybridoma cells in the presence of at least one chemical inhibitor of mismatch repair, thereby forming hypermutated hybridoma cells; (d) performing a screen for antigen binding for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for the antigen than antibodies produced by said parental hybridoma cells; thereby producing hybridoma cells producing high-affinity antibodies. [0035] The invention also comprises antibodies produced by the hybridoma cells. [0036] The invention also provides methods for producing hybridoma cells that produce high titers of antibodies from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells; (c) incubating the parental hybridoma cells in the presence of at least one chemical inhibitor of mismatch repair, thereby forming hypermutated hybridoma cells; (d) performing a screen of the hypermutated hybridoma cells for antigen-specific antibodies produced in higher titers than that produced by the parental hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce higher titers of antibodies than that produced by said parental hybridoma cells; thereby producing hybridoma cells producing high titers of antibodies.
[0037] In some embodiments of the method of the invention, the hypermutated hybridoma cells also are screened for the production of higher titers of antibodies than that produced by the parental hybridomas. The screening may be using an ELISA-based assay, or any other means to measure antibody-antigen binding.
[0038] The invention also comprises antibodies produced by the hybridoma cells. [0039] The invention also provides methods for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for antigen binding of antibodies produced from the hybridoma cells; (d) cloning immunoglobulin genes from the hybridoma cells into a mammalian expression cell; (e) incubating the mammalian expression cell in the presence of at least one chemical inhibitor of mismatch repair; (f) performing a screen for mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from the hybridoma cells; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells.
[0040] In some embodiments of the method of the invention the method may further comprise the removal of the chemical inhibitor from the hypermutated mammalian expression cells, thereby stabilizing the genome of said hypermutated mammalian expression cells. [0041] The invention also comprises antibodies produced by the mammalian expression cells [0042] The invention also provides methods for producing mammalian expression cells that produce high titers of high affinity antibodies to a selected antigen from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) incubating the hybridoma cells in the presence of at least one chemical inhibitor of mismatch repair to form hypermutated hybridoma cells; (d) performing a screen for antigen binding for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for the antigen than antibodies produced by the parental hybridoma cells; (f) cloning immunoglobulin genes from the hypermutated hybridoma cells into a mammalian expression cell, thereby forming parental mammalian expression cells; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells. [0043] In some embodiments, the parental mammalian expression cell is further incubated in the presence of at least one chemical inhibitor of mismatch repair, thereby forming a hypermutated mammalian expression cell; and the hypermutated mammalian expression cells are screened for higher production of antibodies than that of the parental mammalian expression cells.
[0044] In some embodiments of the method of the invention the method may further comprise the removal of the chemical inhibitor from the hypermutated hybridoma and/or hypermutated mammalian expression cells, thereby stabilizing the genome of said hypermutated hybridoma cells and/or hypermutated mammalian expression cells. [0045] The invention also comprises antibodies produced by the mammalian expression cells. [0046] The invention also provides a method for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing said immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for binding of antibodies produced from said hybridoma cells to antigen; (d) cloning immunoglobulin genes from said hybridoma into a mammalian expression cell; (e) incubating said mammalian expression cell in the presence of at least one chemical inhibitor of mismatch repair, thereby forming a hypermutated mammalian expression cell; (f) performing a screen for hypermutated mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from said parental mammalian expression cells; and (g) performing a second screen for hypennutated mammalian expression cells that produce higher titers of antibodies than that produced by parental mammalian expression cells; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells.
[0047] In some embodiments of the method of the invention the method may further comprise the removal of the chemical inhibitor from the hypermutated hybridoma and/or hypermutated mammalian expression cells, thereby stabilizing the genome of said hypermutated hybridoma cells and/or hypermutated mammalian expression cells.
[0048] The invention also comprises antibodies produced by the mammalian expression cells. [0049] In another embodiment, the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro, wherein the donor cells are derived from a donor that is naturally deficient in mismatch repair; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; thereby producing hybridoma cells producing high-affinity antibodies. [0050] The method may further comprise introducing a wild-type gene for mismatch repair into said selected hypermutated hybridoma cell to complement the mismatch repair deficiency, thereby restabilizing the genome of said selected hypermutated hybridoma cell. [0051] The invention also comprises antibodies produced by the hybridoma cells. [0052] In another embodiment, the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells, wherein the myeloma cells are naturally deficient in mismatch repair, thereby forming parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby fonning hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; thereby producing hybridoma cells producing high-affinity antibodies.
[0053] The method may further comprise introducing a wild-type gene for mismatch repair into said selected hypermutated hybridoma cell to complement the mismatch repair deficiency, thereby restabilizing the genome of said selected hypermutated hybridoma cell. [0054] The invention also comprises antibodies produced by the hybridoma cells. [0055] In another embodiment, the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells in high titers comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro, wherein the donor cells are derived from a donor that is naturally deficient in mismatch repair; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; (f) performing a second screen for hypermutated hybridoma cells that produce increased titers of antibodies as compared with parental hybridoma cells; (g) selecting hypermutated hybridoma cells that produce antibodies in higher titers than produced by the parental hybridoma cells; thereby producing hybridoma cells producing high titers of high- affinity antibodies.
[0056] The method may further comprise introducing a wild-type gene for mismatch repair into said selected hypermutated hybridoma cell to complement the mismatch repair deficiency, thereby restabilizing the genome of said selected hypermutated hybridoma cell. [0057] The invention also comprises antibodies produced by the hybridoma cells. [0058] In another embodiment, the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells in high titers comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells, wherein the myeloma cells are naturally deficient in mismatch repair, thereby forming parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; (f) performing a second screen for hypermutated hybridoma cells that produce increased titers of antibodies as compared with parental hybridoma cells; (g) selecting hypermutated hybridoma cells that produce antibodies in higher titers than produced by the parental hybridoma cells; thereby producing hybridoma cells producing high titers of high-affinity antibodies. [0059] The method may further comprise introducing a wild-type gene for mismatch repair into said selected hypermutated hybridoma cell to complement the mismatch repair deficiency, thereby restabilizing the genome of said selected hypermutated hybridoma cell. [0060] The invention also comprises antibodies produced by the hybridoma cells. [0061] In another embodiment, the invention comprises a method for producing mammalian expression cells that produce high-affinity antibodies in high titers from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro, wherein the donor cells are derived from a donor that is naturally deficient in mismatch repair; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; (f) cloning immunoglobulin genes from said hypermutated hybridoma into a mammalian expression cell; thereby producing a mammalian expression cell that produces high titers of high-affinity antibodies in high titer from in vitro immunized immunoglobulin- producing cells.
[0062] In some embodiments, the parental mammalian expression cell is further incubated in the presence of at least one chemical inhibitor of mismatch repair, thereby forming a hypermutated mammalian expression cell; and the hypermutated mammalian expression cells are screened for higher production of antibodies than that of the parental mammalian expression cells.
[0063] In some embodiments of the method of the invention the method may further comprise the removal of the chemical inhibitor from the hypermutated mammalian expression cells, thereby stabilizing the genome of said hypermutated mammalian expression cells. [0064] The invention also comprises antibodies produced by the mammalian expression cells. [0065] In another embodiment, the invention comprises a method for producing mammalian expression cells that produce high-affinity antibodies in high titer from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells, wherein the myeloma cells are naturally deficient in mismatch repair, thereby forming parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; and (f) cloning immunoglobulin genes from said hypermutated hybridoma cell into a mammalian expression cell; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells. [0066] In some embodiments, the parental mammalian expression cell is further incubated in the presence of at least one chemical inhibitor of mismatch repair, thereby forming a hypermutated mammalian expression cell; and the hypermutated mammalian expression cells are screened for higher production of antibodies than that of the parental mammalian expression cells. [0067] In some embodiments of the method of the invention the method may further comprise the removal of the chemical inhibitor from the hypermutated mammalian expression cells, thereby stabilizing the genome of said hypermutated mammalian expression cells. [0068] The invention also comprises antibodies produced by the hybridoma cells. [0069] In some embodiments of the methods of the invention, the immunoglobulin-producing cells are mammalian cells, including but not limited to, mouse cells, rat cells, goat cells, cow cells, horse cells, dog cells, cat cells, rabbit cells, bird cells, monkey cells and human cells. In preferred embodiments, the cells are human cells.
[0070] In some embodiments the dominant negative allele of a mismatch repair gene is a dominant negative allele of PMS2, PMS1, PMSR3, PMSR2, PMSR6, MLH1, GTBP, MSH3, MSH2, MLH3, or MSH1, and homologs of PMSR genes as described in Nicolaides et al. (1995) Genomics 30:195-206 and Horii et al (1994) Biochem. Biophys. Res. Commun. 204:1257-1264. However, the mismatch repair genes are not limit to these examples. [0071] In some embodiments of the method of the invention, the immunogenic antigen is conjugated to a mitogenic polypeptide comprising at least a portion of a polypeptide including, but not limited to tetanus toxoid, ovalbumin, bovine serum albumen, thyroglobulin, diptheria toxoid, BCG, and cholera toxin. In some embodiments, the antigen is generated by denaturing the mature protein.
[0072] In some embodiments of the method of the invention, the antibodies produced have an affinity of at least about 1 x 107 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 108 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 109 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 1010 M'1. In other embodiments, the antibodies have an affinity of at least about 1 x 1011 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 1012 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 1013 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 1014 M"1. [0073] In some embodiments, the antibodies are produced in a higher titer than the parental cell lines, such as in an amount of at least about 1.5 fold higher than the parental cell line. In other embodiments, the titer is at least about 1.5-3 fold higher than the parental cell line. In other embodiments, the titer is at least about 3-5 fold higher than the parental cell line. In other embodiments, the titer is at least about 5-7 fold higher than the parental cell line. In other embodiments, the titer is at least about 7-9 fold higher than the parental cell line. In other embodiments, the titer is at least about 9-10 fold higher than the parental cell line. [0074] In some embodiments of the method of the invention, mutation rates are further enhanced by incubating the hybridoma cells with a chemical mutagen, such as, but not limited to N-ethyl-N-nitrosourea, N-methyl-N-nitrosourea, procarbazine hydrochloride, chlorarnbucil, cyclophosphamide, methyl methanesulfonate, ethyl methanesulfonate, diethyl sulfate, acrylamide monomer, triethylene melamin, melphalan, nitrogen mustard, vincristine, dimethylnitrosamine, N-methyl-N'-nitro-nitrosoguanidine, 7,12 dimethylbenz (a) anthracene, ethylene oxide, hexamethylphosphoramide, and bisulfan.
[0075] The chemical inhibitors of mismatch repair used in certain embodiments of the methods of the invention include, but are not limited to, at least one of an anthracene, an ATPase inhibitor, a nuclease inhibitor, an RNA interference molecule, a polymerase inhibitor and an antisense oligonucleotide that specifically hybridizes to a nucleotide encoding a mismatch repair protein. In some embodiments, the chemical inhibitor is an anthracene having the formula:
Figure imgf000016_0001
wherein Ri-Rio are independently hydrogen, hydroxyl, amino, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S- alkenyl, N-alkenyl, O-alkynyl, S-alkynyl, N-alkynyl, aryl, substituted aryl, aryloxy, substituted aryloxy, heteroaryl, substituted heteroaryl, aralkyloxy, arylalkyl, alkylaryl, alkylaryloxy, arylsulfonyl, alkylsulfonyl, alkoxycarbonyl, aryloxycarbonyl, guanidino, carboxy, an alcohol, an amino acid, sulfonate, alkyl sulfonate, CN, NO2, an aldehyde group, an ester, an ether, a crown ether, a ketone, an organosulfur compound, an organometallic group, a carboxylic acid, an organosilicon or a carbohydrate that optionally contains one or more alkylated hydroxyl groups; wherein said heteroalkyl, heteroaryl, and substituted heteroaryl contain at least one heteroatom that is oxygen, sulfur, a metal atom, phosphorus, silicon or nitrogen; and wherein said substituents of said substituted alkyl, substituted alkenyl, substituted alkynyl, substituted aryl, and substituted heteroaryl are halogen, CN, NO2, lower alkyl, aryl, heteroaryl, aralkyl, aralkoxy, guanidino, alkoxycarbonyl, alkoxy, hydroxy, carboxy and amino; and wherein said amino groups are optionally substituted with an acyl group, or 1 to 3 aryl or lower alkyl groups. In certain embodiments, R5 and R^ are hydrogen. In other embodiments, Ri-Rio are independently hydrogen, hydroxyl, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, phenyl, tolyl, hydroxymethyl, hydroxypropyl, or hydroxybutyl. Non-limiting examples of the anthracenes include 1,2-dimethylanthracene, 9,10-dimethylanthracene, 7,8- dimethylanthracene, 9,10-duphenylanthracene, 9,10-dihydroxymethylanthracene, 9- hydroxymethyl- 10-methylanthracene, dimethylanthracene- 1 ,2-diol, 9-hydroxymethyl- 10- methylanthracene-l,2-diol, 9-hydroxymethyl- 10-methylanthracene-3,4-diol, and 9,10-di-m- tolylanthracene.
[0076] The chemical inhibitor may be introduced into the growth medium of the cells. In some embodiments, the chemical inhibitor may be withdrawn from the hypermutated hybridoma cells in order to re-stabilize the genome of the cells.
[0077] The invention also comprises a method for in vitro production of antigen-specific immunoglobulin-producing cells comprising: (a) isolating donor cells from an animal; (b) treating said cells with L-leucyl-L-leucine methy ester hydrobromide; (c) incubating said donor cells with an immunogenic antigen in vitro, at 25-37°C, 5-10% CO2, in medium supplemented with 5-15% serum, and a growth promoting cytokine for 4 days; (d) washing said cells in medium; and (e) culturing said cells in medium supplemented with 5-15% serum an additional 8 days; thereby stimulating the production of antigen-specific immunoglobulin- producing cells.
[0078] In some embodiments, the immunoglobulin-producing cells are human cells. [0079] In some embodiments of the method of the invention, the immunogenic antigen is conjugated to a mitogenic polypeptide comprising at least a portion of a polypeptide including, but not limited to tetanus toxoid, ovalbumin, bovine serum albumen, thyroglobulin, diptheria toxoid, BCG, and cholera toxin. In some embodiments, the antigen is generated by denaturing the mature protein. [0080] In some embodiments of the method of the invention, the antibodies produced have an
7 1 affinity of at least about 1 x 10 M" . In other embodiments, the antibodies have an affinity of at least about 1 x 108 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 109 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 1010 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 1011 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 1012 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 1013 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 1014 M"1. [0081] In some embodiments, the antibodies are produced in a higher titer than the parental cell lines, such as in an amount of at least about 1.5 fold higher than the parental cell line. In other embodiments, the titer is at least about 1.5-3 fold higher than the parental cell line. In other embodiments, the titer is at least about 3-5 fold higher than the parental cell line. In other embodiments, the titer is at least about 5-7 fold higher than the parental cell line. In other embodiments, the titer is at least about 7-9 fold higher than the parental cell line. In other embodiments, the titer is at least about 9-10 fold higher than the parental cell line. [0082] In some embodiments of the method of the invention, mutation rates are further enhanced by incubating the hybridoma cells and/or mammalian expression cells with a chemical mutagen, such as, but not limited to N-ethyl-N-nitrosourea, N-methyl-N-nitrosourea, procarbazine hydrochloride, chlorambucil, cyclophosphamide, methyl methanesulfonate, ethyl methanesulfonate, diethyl sulfate, acrylamide monomer, triethylene melamin, melphalan, nitrogen mustard, vincristine, dimethylnitrosamine, N-methyl-N'-nitiO-nitrosoguanidine, 7,12 dimethylbenz (a) anthracene, ethylene oxide, hexamethylphosphoramide, and bisulfan. [0083] The mammalian expression cells used in the methods of the invention may include, but are not limited to, Chinese Hamster Ovary, baby hamster kidney cells, human embryonic kidney line 293, normal dog kidney cell lines, normal cat kidney cell lines, monkey kidney cells, African green monkey kidney cells, COS cells, and non-tumorigenic mouse myoblast G8 cells, fibroblast cell lines, myeloma cell lines, mouse NTH/3T3 cells, LMTK31 cells, mouse sertoli cells, human cervical carcinoma cells, buffalo rat liver cells, human lung cells, human liver cells, mouse mammary tumor cells, TRI cells, MRC 5 cells, and FS4 cells. [0084] These and other embodiments are described more fully in the next section and include certain non-limiting examples.
BRIEF DESCRIPTION OF THE DRAWINGS
[0085] Figure 1 shows the immune response of PBMCs to antigen stimulation. PBMCs were cultured in the presence or absence of TT for 4 days then washed with medium and cultured in the presence or absence of TT for an additional eight days. Culture supernates were collected and tested for the presence of antibody reactive to TT. Antibodies bound to TT pre-coated on the solid phase were detected with HRP-labeled goat anti-human IgG, or HRP-labeled goat anti-human IgM.
[0086] Fig. 2A shows reactivity of donor serum to TT by detection of donor anti-TT IgG. Fig. 2B shows reactivity of donor serum to TT by detection of donor anti-TT IgM. [0087] Figure 3 shows the frequency of the anti-TT response of PBMCs upon in vitro immunization with TT, or with TT in combination with IL-2, or CD40L. [0088] Figure 4 shows the intensity of the response of PBMCs upon in vitro immunization with TT, or with TT in combination with IL-2, or CD40L. [0089] Figure 5 shows the response of hybridomas expressing anti-TT antibodies.
[0090] Fig. 6A shows the reactivity of unstimulated PBMCs to EGFR. Fig. 6B shows the reactivity of PBMCs to EGFR after immunization with EGFR-TT. Fig. 6C shows the reactivity of unstimulated PBMCs to EGFR-TT. Fig. 6D shows the reactivity of PBMCs to
EGFR-TT after immunization with EGFR-TT.
[0091] Figure 7 shows the response of hybridomas expressing antibodies against human
EGFR. Antibodies bound to EGFR or BSA (control) pre-coated on the solid phase were detected with HRP-labeled goat anti-human IgG or HRP-labeled goat anti-human IgM.
[0092] Figure 8 shows the IgG and IgM responses of cells immunized with tumor cells in vitro.
[0093] Figure 9 shows reactivity of clones to GM-CSF, chick ovalbumin (CAB), or keyhole limpet hemocyanin.
[0100] Figure 10 shows inhibitory effect of anti-GM-CSF antibodies on proliferation of TF-1 cells. Shown are the effects of a GM-CSF-specific, blocking antibody; a GM-CSF-specific, non-blocking antibody; and a non-specific antibody.
DETAILED DESCRIPTION OF THE INVENTION
[0101] The referenced patents, patent applications, and scientific literature, including accession numbers to GenBank database sequences, referred to herein are hereby incorporated by reference in their entirety. Any conflict between any reference cited herein and the specific teachings of this specification shall be resolved in favor of the latter. Likewise, any conflict between an art-understood definition of a word or phrase and a definition of the word or phrase as specifically taught in this specification shall be resolved in favor of the latter. [0102] Standard reference works setting forth the general principles of recombinant DNA technology known to those of skill in the art include, but are not limited to Ausubel et al. CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York (1998); Sambrook et al. MOLECULAR CLONING: A LABORATORY MANUAL, 2D ED., Cold Spring Harbor Laboratory Press, Plainview, New York (1989); Kaufman et al., Eds., HANDBOOK OF MOLECULAR AND CELLULAR METHODS IN BIOLOGY AND MEDICINE, CRC Press, Boca Raton (1995); McPherson, Ed., DIRECTED MUTAGENESIS: A PRACTICAL APPROACH, ΓRL Press, Oxford (1991).
[0103] The invention provides various embodiments of a method for producing antibody- producing cells and antibodies from in vitro immunized cells with high affinity, and/or increased production. In some embodiments, the cells that produce the antibodies are hybridoma cells, formed by fusing myeloma cells with the lymphoid cells that have been immunized against an antigen in vitro. In other embodiments, the cells that produce the antibodies are mammalian cells that have been transfected with immunoglobulin genes cloned from lymphoid cells that have been immunized against an antigen in vitro. In some embodiments, the method employs both hybridoma cells and mammalian cells. Some basic embodiments of the method of the invention may be described as follows. [0104] In one embodiment, the invention provides a method for generating hybridoma cells producing high-affinity antibodies from in vitro immunized, immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells express a dominant negative allele of a mismatch repair gene; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for the antigen than antibodies produced by the parental hybridoma cells; thereby producing hybridoma cells producing high-affinity antibodies.
[0105] In another embodiment, the invention provides methods of producing hybridoma cells that produce high titers of antibodies from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells express a dominant negative allele of a mismatch repair gene; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen of the hypermutated hybridoma cells for antigen-specific antibodies produced in higher titers than that produced by the parental hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce higher titers of antibodies than that produced by the parental hybridoma cells; thereby producing hybridoma cells that produce high titers of antibodies.
[0106] In another embodiment, the invention provides a method for producing hybridoma cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing said immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for binding of antibodies produced from said hybridoma cells to antigen; (d) cloning immunoglobulin genes from said hybridoma into a mammalian expression cell, wherein said mammalian expression cell expresses a dominant negative allele of a mismatch repair gene; and (e) performing a screen for mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from said hybridoma cells; thereby producing hybridoma cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells.
[0107] In another embodiment, the invention provides a method for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells are produced by: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing said immuiioglobulin-producing cells with myeloma cells to form hybridoma cells, wherein said hybridoma cells express a dominant negative allele of a mismatch repair gene; (c) incubating said parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from said hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for said antigen than antibodies produced by said parental hybridoma cells; and (f) cloning immunoglobulin genes from said hybridoma into a mammalian expression cell; thereby producing high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells.
[0108] In yet another embodiment, the invention provides mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin- producing cells are produced by: (a) combining donor cells comprising immunoglobulin- producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin- producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for binding of antibodies produced from the hybridoma cells to antigen; (d) cloning immunoglobulin genes from the hybridoma into a parental mammalian expression cell, wherein the mammalian expression cell expresses a dominant negative allele of a mismatch repair gene; (e) incubating the parental mammalian expression cell to allow for mutagenesis, thereby forming hypermutated mammalian expression cells; (f) performing a screen of hypermutable mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from the hybridoma cells; and (g) performing a screen of hypermutable mammalian expression cells that secrete higher titers of antibodies than parental mammalian expression cells; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin- producing cells.
[0109] In yet another embodiment, the invention provides a method of producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells are produced by: (a) combining donor cells comprising immunoglobulin- producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin- producing cells with myeloma cells to form parental hybridoma cells; (c) incubating the parental hybridoma cells in the presence of at least one chemical inhibitor of mismatch repair, thereby forming hypermutated hybridoma cells; (d) performing a screen for antigen binding for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for the antigen than antibodies produced by said parental hybridoma cells; thereby producing hybridoma cells that produce high-affinity antibodies.
[0110] In still another embodiment, the invention provides a method of producing hybridoma cells that produce high titers of antibodies from in vitro immunized immunoglobulin- producing cells are produced by: (a) combining donor cells comprising immunoglobulin- producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin- producing cells with myeloma cells to form parental hybridoma cells; (c) incubating the parental hybridoma cells in the presence of at least one chemical inhibitor of mismatch repair, thereby forming hypermutated hybridoma cells; (d) performing a screen of the hypermutated hybridoma cells for antigen-specific antibodies produced in higher titers than that produced by the parental hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce higher titers of antibodies than that produced by said parental hybridoma cells; thereby producing hybridoma cells producing high titers of antibodies.
[0111] In another embodiment, the invention provides methods for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells are produced by: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for antigen binding of antibodies produced from the hybridoma cells; (d) cloning immunoglobulin genes from the hybridoma cells into a mammalian expression cell; (e) incubating the mammalian expression cell in the presence of at least one chemical inhibitor of mismatch repair; and (f) performing a screen for mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from the hybridoma cells; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells. [0112] In yet another embodiment, the invention provides a method for producing mammalian expression cells that produce high affimty antibodies to a selected antigen from in vitro immunized immunoglobulin-producing cells are produced in high titers by: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) incubating the hybridoma cells in the presence of at least one chemical inhibitor of mismatch repair to form hypermutated hybridoma cells; (d) performing a screen for antigen binding for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for the antigen than antibodies produced by the parental hybridoma cells; and (f) cloning immunoglobulin genes from the hypermutated hybridoma cells into a mammalian expression cell, thereby forming parental mammalian expression cells; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin- producing cells.
[0113] In yet another embodiment, the invention also provides methods for producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing said immunoglobulin-producing cells with myeloma cells to form hybridoma cells; (c) performing a screen for binding of antibodies produced from said hybridoma cells to antigen; - (d) cloning immunoglobulin genes from said hybridoma into a mammalian expression cell; (e) incubating said mammalian expression cell in the presence of at least one chemical inhibitor of mismatch repair, thereby forming a hypermutated mammalian expression cell; (f) performing a screen for hypermutated mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from said parental mammalian expression cells; and (g) performing a second screen for hypermutated mammalian expression cells that produce higher titers of antibodies that produced by parental mammalian expression cells; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells. [0114] In another embodiment, the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro, wherein the donor cells are derived from a donor that is naturally deficient in mismatch repair; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; thereby producing hybridoma cells producing high-affinity antibodies.
[0115] In another embodiment, the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells, wherein the myeloma cells are naturally deficient in mismatch repair, thereby forming parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; and (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; thereby producing hybridoma cells producing high-affinity antibodies.
[0116] In another embodiment, the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells in high titers comprising: (a) combimng donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro, wherein the donor cells are derived from a donor that is naturally deficient in mismatch repair; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; (f) performing a second screen for hypermutated hybridoma cells that produce increased titers of antibodies as compared with parental hybridoma cells; and (g) selecting hypermutated hybridoma cells that produce antibodies in higher titers than produced by the parental hybridoma cells; thereby producing hybridoma cells producing high titers of mgh-affinity antibodies.
[0117] In another embodiment, the invention comprises a method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin- producing cells in high titers comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells, wherein the myeloma cells are naturally deficient in mismatch repair, thereby forming parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; (f) performing a second screen for hypermutated hybridoma cells that produce increased titers of antibodies as compared with parental hybridoma cells; and (g) selecting hypermutated hybridoma cells that produce antibodies in higher titers than produced by the parental hybridoma cells; thereby producing hybridoma cells producing high titers of high-affinity antibodies.
[0118] In another embodiment, the invention comprises a method for producing mammalian expression cells that produce high-affinity antibodies in high titers from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro, wherein the donor cells are derived from a donor that is naturally deficient in mismatch repair; (b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypennutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; and (f) cloning immunoglobulin genes from said hypermutated hybridoma into a mammalian expression cell; thereby producing a mammalian expression cell that produce high titers of high-affinity antibodies in high titer from in vitro immunized immunoglobulin-producing cells. [0119] In another embodiment, the invention comprises a method for producing mammalian expression cells that produce high-affinity antibodies in high titer from in vitro immunized immunoglobulin-producing cells comprising: (a) combimng donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro; (b) fusing the immunoglobulin-producing cells with myeloma cells, wherein the myeloma cells are naturally deficient in mismatch repair, thereby forming parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; and (f) cloning immunoglobulin genes from said hypermutated hybridoma cell into a mammalian expression cell; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells. [0120] The invention also provides hybridoma cells, expression cells produced by any of the methods of the invention, as well as antibodies produced by any of the hybridoma cells and expression cells of the invention.
[0121] In still another embodiment, antigen-specific immunoglobulin-producing cells are produced by: (a) isolating donor cells from an animal; (b) treating said cells with L-leucyl-L- leucine methy ester hydrobromide; (c) incubating said donor cells with an immunogenic antigen in vitro, at 25-37°C, 5-10% CO2, in medium supplemented with 5-15% serum, and a growth promoting cytokine for 4 days; (d) washing said cells in medium; and (e) culturing said cells in medium supplemented with 5-15% serum an additional 8 days; thereby stimulating the production of antigen-specific immunoglobulin-producing cells.
[0122] The blood cells used in the methods of the invention may be derived from any animal that produces antibodies. Preferably, the donor cells are derived from mammals, including, but not limited to humans, monkeys, mice, rats, guinea pigs, hamsters, gerbils, birds, rabbits, sheep, goats, pigs, horses, and cows. The source of blood is not necessarily limited, but may be whole blood or fractions containing lymphocytes. The blood may be donor or cord blood, for example. In some embodiments, the blood cells are preferably human donor cells. [0123] The myeloma cells used to create the hybridoma cells in the method of the invention may be derived from any species known to have suitable myeloma cells. For example, but not by way of limitation, the myeloma cells may be conveniently derived from humans or mice. Suitable examples of myeloma cells include, but are not limited to the HuNSl myeloma as described in U.S. Patent No. 4,720,459 to Winkelhake, and deposited with the American Type Culture Collection (ATCC) as CRL 8644; GM4672; RPMI 8226; and murine myeloma cell lines (e.g., P3-NSl/l-Ag4-l; P3-x63-Ag8.653; Sp2/O-Agl4; NS/O, NS/1, SP2 and S194). [0124] The mammalian expression cells suitable for use in certain embodiments of the method of the invention include, but are not limited to Chinese Hamster Ovary cells (CHO cells, Urlaub and Chasin (1980) Proc. Natl. Acad. Sci. USA, 11: 4216), baby hamster kidney (BHK cells), human embryonic kidney line 293 (HeLa cells, Graham et al, (1977) J. Gen Virol, 36: 59), normal dog kidney cell line (e.g., MDCK, ATCC CCL 34), normal cat kidney cell line (CRFK cells), monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587), COS (e.g., COS-7) cells, and non-tumorigenic mouse myoblast G8 cells (e.g., ATCC CRL 1246), fibroblast cell lines (e.g., human, murine or chicken embryo fibroblast cell lines), myeloma cell lines, mouse NIH/3T3 cells, LMTK cells, mouse sertoli cells (TM4, Mather, (1980) Biol. Reprod., 23:243-251); human cervical carcinoma cells (HELA, ATCC CCL 2); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor cells (MMT 060562, ATCC CCL51), TRI cells (Mather et al. (1982) Annals N.Y. Acad. Sci. 383:44-68); MRC 5 cells; FS4 cells; and the human hepatoma line (Hep G2). [0125] As an alternative to mammalian expression cells, other non-mammalian cells may be used to express the cloned immunoglobulin genes. Such non-mammalian cells include, but are not limited to insect cells (e.g., Spodoptera frugiperda cells and the like). Vectors and non- mammalian host cells are well known in the art and are continually being optimized and developed. Any host cell system capable of expressing antibodies may be used in the methods of the invention.
[0126] As used herein, "dominant negative allele of a mismatch repair gene" refers to an allele of a mismatch repair gene that, when expressed, exerts a dominant phenotype in the cell or organism that leads to an inhibition of the mismatch repair system, even in the presence of a wild-type allele. Cells expressing a dominant negative allele of a mismatch repair gene are hypermutable and accumulate mutations at a higher rate than wild-type cells. Examples of nucleic acid sequences encoding mismatch repair proteins useful in the method of the invention include, but are not limited to the following: PMS1 (SEQ LD NO:l); PMS2 (SEQ LO NO:3); PMS2-134 (SEQ TD NO:5); PMSR2 (SEQ ID NO:7); PMSR3 (SEQ ID NO:9); MLHl (SEQ ID NO:ll); MLH3 (SEQ ID NO:13); MSH2 (SEQ ID NO:15); MSH3 (SEQ ID NO:17); MSH4 (SEQ ID NO: 19); MSH5 (SEQ LD NO:21); MSH6 (SEQ ID NO:23); PMSR6 (SEQ ID NO:25); PMSL9 (SEQ ID NO:27); yeast MLHl (SEQ LD NO:29); mouse PMS2 (SEQ LD NO:31); mouse PMS2-134 (SEQ LD NO:33); Arabidopsis thaliana PMS2 (SEQ ID NO:35); A. thaliana PMS2-134 (SEQ ID NO:37) A. thaliana PMS1 (SEQ ID NO:39); A. thaliana MSH7 (SEQ ID NO:41) A. thaliana MSH2 (SEQ JD NO:43); A. thaliana MSH3 (SEQ ID NO:45); A. thaliana MSH6-1 (SEQ ID NO:47); and Oryza satvia MLHl (SEQ ID NO:49). The corresponding amino acid sequences for the listed nucleic acid sequences are: PMS1 (SEQ ID NO:2); PMS2 (SEQ LD NO:4); PMS2-134 (SEQ ID NO:6); PMSR2 (SEQ ID NO:8); PMSR3 (SEQ ID NO:10); MLHl (SEQ 3D NO:12); MLH3 (SEQ LD NO:14); MSH2 (SEQ LD NO: 16); MSH3 (SEQ ID NO: 18); MSH4 (SEQ ID NO:20); MSH5 (SEQ ID NO:22); MSH6 (SEQ ID NO:24); PMSR6 (SEQ ID NO:26); PMSL9 (SEQ ID NO:28); yeast MLHl (SEQ LD NO:30); mouse PMS2 (SEQ ID NO:32); mouse PMS2-134 (SEQ ID NO:34); Arabidopsis thaliana PMS2 (SEQ ID NO:36); A. thaliana PMS2-134 (SEQ ID NO:38); A. thaliana PMS1 (SEQ ID NO:40); A. thaliana MSH7 (SEQ ID NO:42) A. thaliana MSH2 (SEQ ID NO:44); A. thaliana MSH3 (SEQ LD NO:46); A. thaliana MSH6-1 (SEQ ID NO:48); and Oryza satvia MLHl (SEQ ID NO:50).
[0127] As used herein, "high titer" refers to an titer of at least about 1.5 fold higher than the parental cell line. In some embodiments, the titer is at least about 1.5-3 fold higher, 3-5 fold higher, 5-7 fold higher, 7-9 fold higher, or 9-10 fold higher than the parental cell line. [0128] As used herein, "high affinity" refers to a high antibody binding affinity, that may be calculated according to standard methods by the formula Ka = 8/3 (It-Tt) where "It" is the total molar concentration of inhibitor uptake at 50% tracer and "Tt" is the total molar concentration of tracer. See Muller (1980) J. Immunol. Meth. 34:345-352. Binding affinity may also be calculated using the formula B/T = n'NA 'W108 [(V-Vm)K+Q'W] (See Antoni and Mariani (1985) J. Immunol. Meth. 83:61-68). As used herein, "high affinity" is at least about 1 x 107 M"1. In some embodiments, the antibodies have an affinity of at least about 1 x 10s M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 109 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 1010 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 1011 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 1012 M . In other embodiments, the antibodies have an affinity of at least about 1 x 1013 M"1. In other embodiments, the antibodies have an affinity of at least about 1 x 1014 M"1. [0129] As used herein, "antigen-specific" refers to an interaction between the CDR regions of the immunoglobulin molecule with an epitope of the antigen wherein the CDR regions of the immunoglobulin molecule binds to the epitope. [0130] As used herein, "cured" refers to a state of the cells wherein the dominant negative mismatch repair gene has been eliminated from the cell or wherein the expression of the dominant negative allele has been turned off, leading to a stabilized genome, producing stable biological products such as immunoglobulins.
[0131] In some embodiments of the methods of the invention, mismatch repair is inhibited by introducing a dominant negative allele of a mismatch repair gene into a cell. [0132] In other embodiments of the methods of the invention, mismatch repair is inhibited by exposing cells that express an antibody to a compound that inhibits mismatch repair. In some embodiments, the compound is an ATPase inhibitor. Suitable ATPase inliibitors include, but not limited to ATP analogs that are capable of blocking the ATPase activity necessary for mismatch repair in the cell. Examples of ATP analogs that may be used in the methods of the invention, include, but are not limited to non-hydrolyzable forms of ATP, such as AMP-PNP and ATPγS, which block mismatch repair activity (Galio et al. (1999) Nucl. Acids Res. 27:2325-2331; Allen et al. (1997) EMBO J. 16:4467-4476; Bjornson et al. (2000) Biochem. 39:3176-3183). Other suitable ATPase inhibitors may be identified using mismatch repair reporter cells that may be screened with candidate ATPase inhibitors to identify those compounds which effectively block ATPase activity in the cells.
[0133] In other embodiments of the methods of the invention, mismatch repair is inhibited by exposing cells that express an antibody to a nuclease inhibitor. The nuclease inhibitors are capable of blocking exonuclease activity in the mismatch repair biochemical pathway. Mismatch repair reporter cells may be screened with candidate nuclease inhibitors to identify compounds that effectively block the exonuclease activity of the mismatch repair system. Suitable nuclease inhibitors which may be used in the methods of the invention include, but are not limited to analogs of N-ethylmaleimide, an endonuclease inhibitor (Huang et al. (1995) Arch. Biochem. Biophys. 316:485); heterodimeric adenosine-chain-acridine compounds, exonuclease III inhibitors (Belmont et al. (2000) Bioorg. Med. Chem Lett. 10:293-295); as well as antibiotic compounds such as heliquinomycin, which have helicase inhibitory activity (Chino et al. (1998) J. Antϊbiot (Tokyo) 51:480-486). Other suitable nuclease inhibitors may be identified using mismatch repair reporter cells that may be screened with candidate nuclease inhibitors to identify those compounds which effectively block nuclease activity in the cells.
[0134] In other embodiments of the methods of the invention, mismatch repair is inhibited by exposing the cells producing antibodies to DNA polymerase inhibitors. DNA polymerase inhibitors are capable of blocking the polymerization of DNA which is required for functional mismatch repair. Examples of suitable DNA polymerase inhibitors include, but are not limited to actinomycin D (Martin et al. (1990) J. Immunol. 145:1859); aphidicolin (Kuwakado et al. (1993) Biochem. Pharmacol. 46:1909); l-(2'-deoxy-2'-fluoro-beta-L-arabinofuranosyl)-5- methyluracil (L-FMAU) (Kukhanova et al. (1998) Biochem. Pharmacol. 55:1181-1187); and 2'3'-dideoxyribonucleoside 5'-rriphosphates (ddNTPs) (Ono et al. (1984) Biomed. Pharmacother. 38:382-389). Other suitable DNA polymerase inhibitors may be identified using mismatch repair reporter cells that may be screened with candidate DNA polymerase inhibitors to identify those compounds which effectively block DNA polymerase activity in the cells.
[0135] In other embodiments of the methods of the invention, mismatch repair is inhibited by exposing the cells producing antibody to an anthracene. As used herein the term "anthracene" refers to the compound anthracene. However, when refereed to in the general sense, such as "anthracenes," "an anthracene" or "the anthracene," such terms denote any compound that contains the fused triphenyl core structure of anthracene, i.e.,
Figure imgf000030_0001
regardless of extent of substitution. The anthracene may be substituted or unsubstituted. [0136] As used herein, "alkyl" refers to a hydrocarbon containing from 1 to about 20 carbon atoms. Alkyl groups may straight, branched, cyclic, or combinations thereof. Alkyl groups thus include, by way of illustration only, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, cyclopentyl, cyclopentylmethyl, cyclohexyl, cyclohexylmethyl, and the like. Also included within the definition of "alkyl" are fused and/or polycyclic aliphatic cyclic ring systems such as, for example, adamantane. As used herein the term "alkenyl" denotes an alkyl group having at least one carbon-carbon double bond. As used herein the term "alkynyl" denotes an alkyl group having at least one carbon-carbon triple bond.
[0137] In some prefened embodiments, the alkyl, alkenyl, alkynyl, aryl, aryloxy, and heteroaryl substituent groups described above may bear one or more further substituent groups; that is, they may be "substituted". In some preferred embodiments these substituent groups can include halogens (for example fluorine, chlorine, bromine and iodine), CN, NO2, lower alkyl groups, aryl groups, heteroaryl groups, aralkyl groups, aralkyloxy groups, guanidino, alkoxycarbonyl, alkoxy, hydroxy, carboxy and amino groups. In addition, the alkyl and aryl portions of aralkyloxy, arylalkyl, arylsulfonyl, alkylsulfonyl, alkoxycarbonyl, and aryloxycarbonyl groups also can bear such substituent groups. Thus, by way of example only, substituted alkyl groups include, for example, alkyl groups fluoro-, chloro-, bromo- and iodoalkyl groups, aminoalkyl groups, and hydroxyalkyl groups, such as hydroxymethyl, hydroxyethyl, hydroxypropyl, hydroxybutyl, and the like. In some prefened embodiments such hydroxyalkyl groups contain from 1 to about 20 carbons.
[0138] As used herein the term "aryl" means a group having 5 to about 20 carbon atoms and which contains at least one aromatic ring, such as phenyl, biphenyl and naphthyl. Prefened aryl groups include unsubstituted or substituted phenyl and naphthyl groups. The term "aryloxy" denotes an aryl group that is bound through an oxygen atom, for example a phenoxy group.
[0139] In general, the prefix "hetero" denotes the presence of at least one hetero (i.e., non- carbon) atom, which is in some preferred embodiments independently one to three O, N, S, P, Si or metal atoms. Thus, the term "heteroaryl" denotes an aryl group in which one or more ring carbon atom is replaced by such a heteroatom. Prefened heteroaryl groups include pyridyl, pyrimidyl, pynolyl, furyl, thienyl, and imidazolyl groups.
[0140] The term "aralkyl" (or "arylalkyl") is intended to denote a group having from 6 to 15 carbons, consisting of an alkyl group that bears an aryl group. Examples of aralkyl groups include benzyl, phenethyl, benzhydryl and naphthylmethyl groups.
[0141] The term "alkylaryl" (or "alkaryl") is intended to denote a group having from 6 to 15 carbons, consisting of an aryl group that bears an alkyl group. Examples of aralkyl groups include methylphenyl, ethylphenyl and methylnaphthyl groups.
[0142] The term "arylsulfonyl" denotes an aryl group attached through a sulfonyl group, for example phenylsulfonyl. The term "alkylsulfonyl" denotes an alkyl group attached through a sulfonyl group, for example methylsulfonyl.
[0143] The term "alkoxycarbonyl" denotes a group of formula -C(=O)-O-R where R is alkyl, alkenyl, or alkynyl, where the alkyl, alkenyl, or alkynyl portions thereof can be optionally substituted as described herein.
[0144] The term "aryloxycarbonyl" denotes a group of formula -C(=O)-O-R where R is aryl, where the aryl portion thereof can be optionally substituted as described herein. [0145] The terms "arylalkyloxy" or "aralkyloxy" are equivalent, and denote a group of formula -O-R^R7, where R; is R is alkyl, alkenyl, or alkynyl which can be optionally substituted as described herein, and wherein R7/ denotes a aryl or substituted aryl group. [0146] The terms "alkylaryloxy" or "alkaryloxy" are equivalent, and denote a group of formula -O-R'-R77, where R; is an aryl or substituted aryl group, and R/ is alkyl, alkenyl, or alkynyl which can be optionally substituted as described herein. [0147] As used herein, the term "aldehyde group" denotes a group that bears a moiety of formula -C(=O)-H. The term "ketone" denotes a moiety containing a group of formula -R-
C(=O)-R=, where R and R= are independently alkyl, alkenyl, alkynyl, aryl, heteroaryl, aralkyl, or alkaryl, each of which may be substituted as described herein.
[0148] As used herein, the term "ester" denotes a moiety having a group of formula -R-
C(=O)-O-R= or -R-O-C(=O)-R= where R and R= are independently alkyl, alkenyl, alkynyl, aryl, heteroaryl, aralkyl, or alkaryl, each of which may be substituted as described herein.
[0149] The term "ether" denotes a moiety having a group of formula -R-O-R= or where R and
R= are independently alkyl, alkenyl, alkynyl, aryl, heteroaryl, aralkyl, or alkaryl, each of which may be substituted as described herein.
[0150] The term "crown ether" has its usual meaning of a cyclic ether containing several oxygen atoms. As used herein the term "organosulfur compound" denotes aliphatic or aromatic sulfur containing compounds, for example thiols and disulfides. The term
"organometallic group" denotes an organic molecule containing at least one metal atom.
[0151] The term "organosilicon compound" denotes aliphatic or aromatic silicon containing compounds, for example alkyl and aryl silanes.
[0152] The term "carboxylic acid" denotes a moiety having a carboxyl group, other than an amino acid.
[0153] Suitable anthracenes that may be used in the method of the invention comprise compounds having the formula:
Figure imgf000032_0001
wherein R Rio are independently hydrogen, hydroxyl, amino, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S- alkenyl, N-alkenyl, O-alkynyl, S-alkynyl, N-alkynyl, aryl, substituted aryl, aryloxy, substituted aryloxy, heteroaryl, substituted heteroaryl, aralkyloxy, arylalkyl, alkylaryl, alkylaryloxy, arylsulfonyl, alkylsulfonyl, alkoxycarbonyl, aryloxycarbonyl, guanidino, carboxy, an alcohol, an amino acid, sulfonate, alkyl sulfonate, CN, NO2, an aldehyde group, an ester, an ether, a crown ether, a ketone, an organosulfur compound, an organometallic group, a carboxylic acid, an organosilicon or a carbohydrate that optionally contains one or more alkylated hydroxyl groups; wherein said heteroalkyl, heteroaryl, and substituted heteroaryl contain at least one heteroatom that is oxygen, sulfur, a metal atom, phosphorus, silicon or nitrogen; and wherein said substituents of said substituted alkyl, substituted alkenyl, substituted alkynyl, substituted aryl, and substituted heteroaryl are halogen, CN, NO , lower alkyl, aryl, heteroaryl, aralkyl, aralkoxy, guanidino, alkoxycarbonyl, alkoxy, hydroxy, carboxy and amino; and wherein said amino groups are optionally substituted with an acyl group, or 1 to 3 aryl or lower alkyl groups. In some embodiments, the R5 and R^ are hydrogen. In other embodiments, Ri-Rio are independently hydrogen, hydroxyl, methyl, ethyl, propyl, isopropyl, butyl, isoburyl, phenyl, tolyl, hydroxymethyl, hydroxypropyl, or hydroxybutyl. Suitable anthracenes for use in the methods of the invention include, but are not limited to 1,2-dimethylanthracene, 9,10- dimethylanthracene, 7,8-dimethylanthracene, 9,10-duphenylanthracene, 9,10- dihydroxymethylanthracene, 9-hydroxymethyl- 10-methylanthracene, dimethylanthracene- 1 ,2- diol, 9-hydroxymethyl- 10-methylanthracene- 1 ,2-diol, 9-hydroxymethyl- 10-methylanthracene- 3,4-diol, and 9,10-di-m-tolylanthracene.
[0154] Other suitable anthracenes may be identified using mismatch repair reporter cells that may be screened with candidate anthracenes to identify those compounds which effectively block mismatch repair activity in the cells. In some embodiments, the chemical inhibitor of mismatch repair is an RNA interference molecule that is homologous to a mismatch repair gene of the invention. The technique for generating sequence-specific RNA interference molecules is well-known in the art and may be found in, for example, Sharp et al. (2000) Science 287 r:2431-2433; Marx (2000) Science 288:1370-1372; Grishok et al. (2001) Science 287:2494-2497; and Fire et al. (1998) Nature 391:806-811, the disclosures of which are specifically incorporated by reference in their entirety.
[0155] In other embodiments of the method of the invention, mismatch repair is inhibited by exposing the cells producing antibody to "antisense compounds" which specifically hybridize with one or more nucleic acids encoding a mismatch repair gene. As used herein, the terms "target nucleic acid" and "nucleic acid encoding a mismatch repair gene" encompass DNA encoding a mismatch repair gene, RNA (including pre-mRNA and mRNA) transcribed from such DNA, and also cDNA derived from such RNA. The specific hybridization of an antisense compound with its target nucleic acid interferes with the normal function of the nucleic acid, such as replication and transcription. The functions of RNA disrupted by antisense compounds include such functions as, for example, translocation of the RNA to the site of protein translation, translation of protein from the RNA, and splicing of the RNA to yield one or more mRNA species. The antisense compound thereby inhibits the expression or function of a mismatch repair gene. [0156] It is prefened to target specific nucleic acids for antisense inhibition of mismatch repair in order to reversibly disrupt the function of a given mismatch repair gene. "Targeting" an antisense compound to a particular nucleic acid, in the context of this invention, is a multistep process, beginning with the identification of a nucleic acid sequence whose function is to be modulated. As disclosed herein, there are several mismatch repair genes that may be targeted by an antisense strategy. Among the various mismatch repair genes that may be targeted are PMS2, PMSl, PMSR3, PMSR2, PMSR6, MLHl, GTBP, MSH3, MSH2, MLH3, or MSH1, and homologs of PMSR genes as described in Nicolaides et al. (1995) Genomics 30:195-206 and Horii er a/. (1994) Rtoc/ϊem. Biophys. Res. Commun. 204:1257-1264, including DNA or RNA. The next step of targeting involves the determination of a site or sites within this gene for the antisense interaction to occur, such that inhibition of the function of the mismatch repair gene occurs. In one embodiment, an intragenic site is targeted. An "intragenic site" is a region encompassing the translation initiation or termination codon of the open reading frame (ORF) of the gene. Since, as is known in the art, the translation initiation codon is typically 5'-AUG (in transcribed mRNA molecules; 5'-ATG in the conesponding DNA molecule), the translation initiation codon is also refened to as the "AUG codon," the "start codon" or the "AUG start codon." A minority of genes have a translation initiation codon having the RNA sequence 5'-GUG, 5'-UUG or 5'-CUG, and 5*-AUA, 5'-ACG and 5'- CUG have been shown to function in vivo. Thus, the terms "translation initiation codon" and "start codon" can encompass many codon sequences, even though the initiator amino acid in each instance is typically methionine in eukaryotes. It is also known in the art that eukaryotic and prokaryotic genes may have two or more alternative start codons, any one of which may be preferentially utilized for translation initiation in a particular cell type or tissue, or under a particular set of conditions, hi the context of the invention, "start codon" and "translation initiation codon" refer to the codon or codons that are used in vivo to initiate translation of an mRNA molecule transcribed from a gene encoding a mismatch repair gene, regardless of the sequence(s) of such codons.
[0157] It is also known in the art that a translation termination codon (or "stop codon") of a gene may have one of three sequences, i.e., 5'-UAA, 5'-UAG and 5'-UGA (the corresponding DNA sequences are 5'-TAA, 5'-TAG and 5'-TGA, respectively). The terms "start codon region" and "translation initiation codon region" refer to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (t'.e., 5' or 3') from a translation initiation codon. Similarly, the terms "stop codon region" and "translation termination codon region" refer to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5' or 3') from a translation termination codon.
[0158] The open reading frame (ORF) or "coding region," which is known in the art to refer to the region between the translation initiation codon and the translation termination codon, is also a region which may be targeted effectively. Other target regions include the 5' untranslated region (5'UTR), known in the art to refer to the portion of an mRNA in the 5' direction from the translation initiation codon, and thus including nucleotides between the 5' cap site and the translation initiation codon of an mRNA or conesponding nucleotides on the gene, and the 3' untranslated region (3'UTR), known in the art to refer to the portion of an mRNA in the 3' direction from the translation termination codon, and thus including nucleotides between the translation tennination codon and 3' end of an mRNA or conesponding nucleotides on the gene. The 5' cap of an mRNA comprises an N7-methylated guanosine residue joined to the 5'-most residue of the mRNA via a 5'-5' triphosphate linkage. The 5' cap region of an mRNA is considered to include the 5' cap structure itself as well as the first 50 nucleotides adjacent to the cap. The 5' cap region may also be a prefened target region.
[0159] Although some eukaryotic mRNA transcripts are directly translated, many contain one or more regions, known as "introns," which are excised from a transcript before it is translated. The remaining (and therefore translated) regions are known as "exons" and are spliced together to form a continuous mRNA sequence. mRNA splice sites, i.e., intron-exon junctions, may also be prefened target regions, and are particularly useful in situations where abenant splicing is implicated in disease, or where an overproduction of a particular mRNA splice product is implicated in disease. Abenant fusion junctions due to reanangements or deletions are also prefened targets. It has also been found that introns can also be effective, and therefore prefened, target regions for antisense compounds targeted, for example, to DNA or pre-mRNA.
[0160] Once one or more target sites have been identified, oligonucleotides are chosen which are sufficiently complementary to the target, i.e., hybridize sufficiently well and with sufficient specificity, to give the desired effect.
[0161] In the context of this invention, "hybridization" means hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleoside or nucleotide bases. For example, adenine and thymine are complementary nucleobases which pair through the formation of hydrogen bonds. "Complementary," as used herein, refers to the capacity for precise pairing between two nucleotides. For example, if a nucleotide at a certain position of an oligonucleotide is capable of hydrogen bonding with a nucleotide at the same position of a DNA or RNA molecule, then the oligonucleotide and the DNA or RNA are considered to be complementary to each other at that position. The oligonucleotide and the DNA or RNA are complementary to each other when a sufficient number of conesponding positions in each molecule are occupied by nucleotides which can hydrogen bond with each other. Thus, "specifically hybridizable" and "complementary" are terms which are used to indicate a sufficient degree of complementarity or precise pairing such that stable and specific binding occurs between the oligonucleotide and the DNA or RNA target. It is understood in the art that the sequence of an antisense compound need not be 100% complementary to that of its target nucleic acid to be specifically hybridizable. An antisense compound is specifically hybridizable when binding of the compound to the target DNA or RNA molecule interferes with the normal function of the target DNA or RNA to cause a loss of utility, and there is a sufficient degree of complementarity to avoid non-specific binding of the antisense compound to non-target sequences under conditions in which specific binding is desired, i.e., under physiological conditions in the case of in vivo assays or therapeutic treatment, and in the case of in vitro assays, under conditions in which the assays are performed. Complementarity of the antisense oligonucleotide is preferably 100%, however, degeneracy may be introduced into the oligonucleotide such that the complementarity, in some embodiments, is 80-85%, 85-90%, 90- 95% or 95-100%.
[0162] Antisense and other compounds of the invention which hybridize to the target and inhibit expression of the target are identified through experimentation, and the sequences of these compounds are herein below identified as prefened embodiments of the invention. The target sites to which these prefened sequences are complementary comprise the region of PMS2, for example, which inhibits the translation of the C-terminal portion of the PMS2 protein, effectively forming a truncation mutant. The region targeted comprises a portion of the PMS2 gene that encodes the 134 amino acid of PMS2, for example. [0163] In the context of this invention, the term "oligonucleotide" refers to an oligomer or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) or mimetics thereof. This term includes oligonucleotides composed of naturally-occurring nucleobases, sugars and covalent internucleoside (backbone) linkages as well as oligonucleotides having non-naturally- occurring portions which function similarly. Such modified or substituted oligonucleotides are often prefened over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for nucleic acid target and increased stability in the presence of nucleases.
[0164] While antisense oligonucleotides are a prefened form of antisense compound, the present invention comprehends other oligomeric antisense compounds, including but not limited to oligonucleotide mimetics such as are described below. The antisense compounds in accordance with this invention preferably comprise from about 8 to about 50 nucleobases (i.e., from about 8 to about 50 linked nucleosides). Particularly prefened antisense compounds are antisense oligonucleotides, even more preferably those comprising from about 12 to about 30 nucleobases. Antisense compounds include ribozymes, external guide sequence (EGS) oligonucleotides (oligozymes), and other short catalytic RNAs or catalytic oligonucleotides which hybridize to the target nucleic acid and modulate its expression. In some embodiments, the oligonucleotides are at least about 15 nucleotides in length and may be at least 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 or more nucleotides in length. [0165] In some embodiments, the antisense oligonucleotides comprise a sequence that is complementary to a portion of the mismatch repair sequence shown in SEQ ID NO:l; SEQ LD NO:3; SEQ LD NO:5; SEQ LD NO:7; SEQ ID NO:9; SEQ ID NO.ll; SEQ ID NO:13; SEQ ID NO:15; SEQ J-D NO:17; SEQ J-D NO:19; SEQ ID NO:21; SEQ JD NO:23; SEQ LD NO:25; SEQ J-D NO:27; SEQ ID NO:29; SEQ ID NO:31; SEQ J-D NO:33; SEQ ID NO:35; SEQ ID NO:37; SEQ ID NO:39; SEQ ID NO:41; SEQ ID NO:43; SEQ ID NO:45; SEQ ID NO:47; or SEQ ID NO:49. In certain embodiments, the oligonucleotide is at least 15-50 nucleotides in length with 85-100% complementarity.
[0166] As is known in the art, a nucleoside is a base-sugar combination. The base portion of the nucleoside is normally a heterocychc base. The two most common classes of such heterocychc bases are the purines and the pyrimidines. Nucleotides are nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside. For those nucleosides that include a pentofuranosyl sugar, the phosphate group can be linked to either the 2', 3 Or 5' hydroxyl moiety of the sugar. In forming oligonucleotides, the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound. In turn, the respective ends of this linear polymeric structure can be further joined to form a circular structure, however, open linear structures are generally prefened. Within the oligonucleotide structure, the phosphate groups are commonly refened to as fonning the internucleoside backbone of the oligonucleotide. The normal linkage or backbone of RNA and DNA is a 3' to 5' phosphodiester linkage. [0167] Specific examples of prefened antisense compounds useful in this invention include oligonucleotides containing modified backbones or non-natural internucleoside linkages. As defined in this specification, oligonucleotides having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone. For the purposes of this specification, and as sometimes referenced in the art, modified oligonucleotides that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleosides. [0168] Prefened modified oligonucleotide backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates, 5'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates and boranophosphates having normal 3'-5' linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a 3' to 3', 5' to 5' or 2' to 2' linkage. Prefened oligonucleotides having inverted polarity comprise a single 3' to 3' linkage at the 3'-most internucleotide linkage, i.e., a single inverted nucleoside residue which may be a basic (the nucleobase is missing or has a hydroxyl group in place thereof). Various salts, mixed salts and free acid forms are also included. [0169] Representative United States patents that teach the preparation of the above phosphorus-containing linkages include, but are not limited to, U.S. Pat. Nos. 3,687,808; 4,469,863; 4,476,301; 5,023,243; 5,177,196; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,306; 5,550,111; 5,563,253; 5,571,799; 5,587,361; 5,194,599; 5,565,555; 5,527,899; 5,721,218; 5,672,697 and 5,625,050, each of which is herein incorporated by reference.
[0170] Prefened modified oligonucleotide backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocychc internucleoside linkages. These include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; riboacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH2 component parts.
[0171] Representative United States patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Pat. Nos. 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; 5,792,608; 5,646,269 and 5,677,439, each of which is herein incorporated by reference. [0172] In other prefened oligonucleotide mimetics, both the sugar and the internucleoside linkage, i.e., the backbone, of the nucleotide units are replaced with novel groups. The base units are maintained for hybridization with an appropriate nucleic acid target compound. One such oligomeric compound, an oligonucleotide mimetic that has been shown to have excellent hybridization properties, is refened to as a peptide nucleic acid (PNA). In PNA compounds, the sugar-backbone of an oligonucleotide is replaced with an amide containing backbone, in particular an aminoethylglycine backbone. The nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone. Representative United States patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Pat. Nos. 5,539,082; 5,714,331; and 5,719,262, each of which is herein incorporated by reference. Further teaching of PNA compounds can be found in Nielsen et al, (1991) Science 254:1497-1500.
[0173] Most prefened embodiments of the invention are oligonucleotides with phosphorothioate backbones and oligonucleosides with heteroatom backbones, and in particular -CH2 -NH-O-CH2-, -CH2~N(CH3)-O~CH2~ [known as a methylene (methylimino) or MMI backbone], -CH2-O~N(CH3)-CH2 --, ~CH2~N(CH3)~N(CH3)-CH2 ~ and ~O~N(CH3)~CH ~CH2— [wherein the native phosphodiester backbone is represented as -O-P-O~CH2 -] of the above referenced U.S. Pat. No. 5,489,677, and the amide backbones of the above referenced U.S. Pat. No. 5,602,240. Also prefened are oligonucleotides having morpholino backbone structures of the above-referenced U.S. Pat. No. 5,034,506.
[0174] Modified oligonucleotides may also contain one or more substituted sugar moieties. Prefened oligonucleotides comprise one of the following at the 2' position: OH; F; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted to o alkyl or C2 to C10 alkenyl and alkynyl. Particularly prefened are O[(CH2)n O]mCH3, O(CH2)nOCH3, O(CH2)„NH2, O(CH2)nCH3, O(CH2)nONH2, and O(CH2)nON[(CH2)nCH3)]2, where n and m are from 1 to about 10. Other prefened oligonucleotides comprise one of the following at the 21 position: to Cio lower alkyl, substituted lower alkyl, alkenyl, alkynyl, alkaryl, aralkyl, O-alkaryl or O- aralkyl, SH, SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, SO2 CH3, ONO2, NO2, N3, NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties. A prefened modification includes 2' -methoxyethoxy (2'-O~CH2CH2OCH3, also known as 2'-O-(2-methoxyethyl) or 2'-MOE) (Martin et al. (1995) Helv. Chim. Acta 78:486- 504) i.e., an alkoxyalkoxy group. A further prefened modification includes 2'- dimethylaminooxyethoxy, i.e., a O(CH2)2ON(CH3)2 group, also known as 2'-DMAOE, as described in examples hereinbelow, and 2'-dimethylaminoethoxyethoxy (also known in the art as 2'-O-dimethylaminoethoxyethyl or 2'-DMAEOE), i.e., 2'-O~CH2 ~O-CH2~N(CH2)2, also described in examples hereinbelow.
[0175] A further prefened modification includes Locked Nucleic Acids (LNAs) in which the 2'-hydroxyl group is linked to the 3 ' or 4' carbon atom of the sugar ring thereby forming a bicyclic sugar moiety. The linkage is preferably a methelyne (~CH2~)n group bridging the 2' oxygen atom and the 3' or 4' carbon atom wherein n is 1 or 2. LNAs and preparation thereof are described in WO 98/39352 and WO 99/14226.
[0176] Other prefened modifications include 2'-methoxy (2'-O~CH3), 2'-aminopropoxy ( - OCH2CH2CH2NH2), 2'-allyl (2'-CH2--CH=CH2), 2'-O-allyl (2'-O~CH2-CH=CH2) and 2'- fluoro (2'-F). The 2'-modification may be in the arabino (up) position or ribo (down) position. A prefened 2'-arabino modification is 2'-F. Similar modifications may also be made at other positions on the oligonucleotide, particularly the 3' position of the sugar on the 3' terminal nucleotide or in 2'-5' linked oligonucleotides and the 5' position of 5' terminal nucleotide. Oligonucleotides may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. Representative United States patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S. Pat. Nos. 4,981,957; 5,118,800;
5,319,080; 5,359,044 5,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811; 5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,053; 5,639,873; 5,646,265; 5,658,873; 5,670,633; 5,792,747 and 5,700,920, each of which is herein incorporated by reference in its entirety. [0177] Oligonucleotides may also include nucleobase (often refened to in the art simply as "base") modifications or substitutions. As used herein, "unmodified" or "natural" nucleobases include the piirine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U). Modified nucleobases include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2- thiocytosine, 5-halouracil and cytosine, 5-propynyl (~C≡C~CH2) uracil and cytosine and other alkynyl derivatives of pyrimidine bases, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8- substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5- substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 2-F-adenine, 2- amino-adenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3- deazaguanine and 3-deazaadenine. Further modified nucleobases include tricyclic pyrimidines such as phenoxazine cytidine(lH-pyrimido[5,4-b][l,4]benzoxazin-2(3H)-one), phenothiazine cytidine (lH-pyrimido[5,4-b][l,4]benzothiazin-2(3H)-one), G-clamps such as a substituted phenoxazine cytidine (e.g., 9-(2-aminoethoxy)-H-pyrimido[5,4-b][l,4]benzoxazin-2(3H)-one), carbazole cytidine (2H-pyrimido[4,5-b]indol-2-one), pyridoindole cytidine (H- pyrido[3',2':4,5]pynolo[2,3-d]pyrimidin-2-one). Modified nucleobases may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example 7- deaza-adenine, 7-deazaguanosine, 2-aminopyridine and 2-pyridone. Further nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in THE CONCISE ENCYCLOPEDIA OF POLYMER SCIENCE AND ENGINEERING, Kroschwitz, (Ed.) John Wiley & Sons, 1990, pages 858-859, those disclosed by Englisch et al. (1991) Angewandte Chemie (International Edition) 30:613, and those disclosed by Sanghvi, A TISENSE RESEARCH AND APPLICATIONS, Crooke and Lebleu (Eds.), CRC Press, 1993, pages 289-302. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2°C. (Sanghvi, ANTISENSE RESEARCH AND APPLICATIONS, Crooke and Lebleu (Eds.), CRC Press, 1993, pp. 276-278) and are presently prefened base substitutions, even more particularly when combined with 2'-O-methoxyethyl sugar modifications. [0178] Representative United States patents that teach the preparation of certain of the above noted modified nucleobases as well as other modified nucleobases include, but are not limited to, the above noted U.S. Pat. No. 3,687,808, as well as U.S. Pat. Nos. 4,845,205; 5,130,302; 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,457,187; 5,459,255; 5,484,908; 5,502,177; 5,525,711; 5,552,540; 5,587,469; 5,594,121, 5,596,091; 5,614,617; 5,645,985; 5,750,692; 5,830,653; 5,763,588; 6,005,096; and 5,681,941, each of which is herein incorporated by reference in its entirety.
[0179] Another modification of the oligonucleotides of the invention involves chemically linking to the oligonucleotide one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide. The compounds of the invention can include conjugate groups covalently bound to functional groups such as primary or secondary hydroxyl groups. Conjugate groups of the invention include intercalators, reporter molecules, polyamines, polyamides, polyethylene glycols, polyethers, groups that enhance the pharmacodynamic properties of oligomers, and groups that enhance the pharmacokinetic properties of oligomers. Typical conjugates groups include cholesterols, lipids, phosphohpids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes. Groups that enhance the pharmacodynamic properties, in the context of this invention, include groups that improve oligomer uptake, enhance oligomer resistance to degradation, and/or strengthen sequence-specific hybridization with RNA. Groups that enhance the pharmacokinetic properties, in the context of this invention, include groups that improve oligomer uptake, distribution, metabolism or excretion. Representative conjugate groups are disclosed in International Patent Application PCT/US92/09196, filed Oct. 23, 1992 the entire disclosure of which is incorporated herein by reference. Conjugate moieties include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al. (1989) Proc. Natl. Acad. Sci. USA 86:6553-6556), cholic acid (Manoharan et al. (1994) Bioorg. Med. Chem. Let. 4:1053-1060), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al. (1992) Ann. N.Y. Acad. Sci. 660:306-309; Manoharan et al. (1993) Bioorg. Med. Chem. Let. 3:2765-2770), a thiocholesterol (Oberhauser et al. (1992) Nucl. Acids Res. 20:533-538), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et al. (1991) EMBO J. 10:1111- 1118; Kabanov et α/. (1990) FEBS Lett. 259:327-330; Svinarchuk et al. (1993) Biochimie 75:49-54), a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethyl-ammonium 1,2-di-O- hexadecyl-rac-glycero-3-H-phosphonate (Manoharan et al. (1995) Tetrahedron Lett. 36:3651- 3654; Shea et al. (1990) Nucl. Acids Res. 18:3777-3783), a polyamine or a polyethylene glycol chain (Manoharan et al. (1995) Nucleosides & Nucleotides 14:969-973), or adamantane acetic acid (Manoharan et al. (1995) Tetrahedron Lett. 36:3651-3654), apalmityl moiety (Mishra et al. (1995) Biochim. Biophys. Ada 1264:229-237), or an octadecylamine or hexylamino- carbonyl-oxycholesterol moiety (Crooke et al. (1996) J. Pharmacol. Exp. Ther. 277:923-937. [0180] Representative United States patents that teach the preparation of such oligonucleotide conjugates include, but are not limited to, U.S. Pat. Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538; 5,578,717, 5,580,731; 5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4,587,044; 4,605,735; 4,667,025; 4,762,779; 4,789,737; 4,824,941; 4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082,830; 5,112,963; 5,214,136; 5,082,830; 5,112,963; 5,214,136; 5,245,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241, 5,391,723; 5,416,203, 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810; 5,574,142; 5,585,481; 5,587,371; 5,595,726; 5,597,696; 5,599,923; 5,599,928 and 5,688,941, each of which is herein incoφorated by reference in its entirety.
[0181] It is not necessary for all positions in a given compound to be liniformly modified, and in fact more than one of the aforementioned modifications may be incorporated in a single compound or even at a single nucleoside within an oligonucleotide. The present invention also includes antisense compounds which are chimeric compounds. "Chimeric" antisense compounds or "chimeras," in the context of this invention, are antisense compounds, particularly oligonucleotides, which contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of an oligonucleotide compound. These oligonucleotides typically contain at least one region wherein the oligonucleotide is modified so as to confer upon the oligonucleotide increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid. An additional region of the oligonucleotide may serve as a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids. By way of example, RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of oligonucleotide inhibition of gene expression. Consequently, comparable results can often be obtained with shorter oligonucleotides when chimeric oligonucleotides are used, compared to phosphorothioate deoxyoligonucleotides hybridizing to the same target region. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.
[0182] Chimeric antisense compounds of the invention may be formed as composite structures of two or more oligonucleotides, modified oligonucleotides, oligonucleosides and/or oligonucleotide mimetics as described above. Such compounds have also been refened to in the art as hybrids or gapmers. Representative United States patents that teach the preparation of such hybrid structures include, but are not limited to, U.S. Pat. Nos. 5,013,830; 5,149,797; 5,220,007; 5,256,775; 5,366,878; 5,403,711; 5,491,133; 5,565,350; 5,623,065; 5,652,355; 5,652,356; and 5,700,922, each of which is herein incorporated by reference in its entirety. [0183] As used herein "donor cells comprising immunoglobulin-producing cells" or "donor cells comprising immunoglobulin-producing cells" sometimes refened to simply as "donor cells" or "donor blood cells" refers to cells that are capable of producing antibodies when immunized with an antigenic compound. Examples of sources of such donor cells suitable for use in the invention include, but are not limited to spleen cells, lymph node cells, bone manow cells, and immortalizing tumor infiltrating lymphocytes.
[0184] As used herein, the tenn "amino acid" denotes a molecule containing both an amino group and a carboxyl group. In some prefened embodiments, the amino acids are α-, β-, γ- or δ-amino acids, including their stereoisomers and racemates. As used herein the term "L-amino acid" denotes an α-amino acid having the L configuration around the α-carbon, that is, a carboxylic acid of general formula CH(COOH)(NH2)-(side chain), having the L-configuration. The term "D-amino acid" similarly denotes a carboxylic acid of general formula CH(COOH)(NH2)-(side chain), having the D-configuration around the α-carbon. Side chains of L-amino acids include naturally occurring and non-naturally occurring moieties. Non- naturally occurring (i.e., unnatural) amino acid side chains are moieties that are used in place of naturally occurring amino acid side chains in, for example, amino acid analogs. See, for example, Lehninger, BIOCHEMISTRY, Second Edition, Worth Publishers, Inc., 1975, pages 72- 77 (incorporated herein by reference). Amino acid substituents may be attached through their carbonyl groups through the oxygen or carbonyl carbon thereof, or through their amino groups, or through functionalities residing on their sidechain portions. [0185] As used herein "polynucleotide" refers to a nucleic acid molecule and includes genomic DNA cDNA, RNA, mRNA and the like.
[0186] As used herein "inhibitor of mismatch repair" refers to an agent that interferes with at least one function of the mismatch repair system of a cell and thereby renders the cell more susceptible to mutation.
[0187] As used herein "hypermutable" refers to a state in which a cell in vitro or in vivo is made more susceptible to mutation through a loss or impairment of the mismatch repair system. [0188] As used herein "agents," "chemicals," and "inhibitors" when used in connection with inhibition of MMR refers to chemicals, oligonucleotides, RNA interference molecules, analogs of natural substrates, and the like that interfere with normal function of MMR. [0189] As used herein, "about" refers to an amount within a range of +/- 10% of the cited value.
[0190] As used herein, "mitogenic polypeptide" refers to a polypeptide when in combination with the antigen provides stimulation of appropriate cells to increase the immune response against the subject antigen.
[0191] As used herein, "hybridoma" refers to the result of a cell fusion between an immunoglobulin-producing cell and a transformed cell, such as a myeloma cell. [0192] As used herein, "IgG subclass" refers to a category of immunoglobulins comprising IgGl, IgG2, IgG2a, IgG2b, IgG3, and IgG4.
[0193] As used herein, "mismatch repair gene" refers to a gene that encodes one of the proteins of the mismatch repair complex. Although not wanting to be bound by any particular theory of mechanism of action, a mismatch repair complex is believed to detect distortions of the DNA helix resulting from non-complementary pairing of nucleotide bases. The non- complementary base on the newer DNA strand is excised, and the excised base is replaced with the appropriate base which is complementary to the older DNA strand. In this way, cells eliminate many mutations that occur as a result of mistakes in DNA replication. Dominant negative alleles cause a mismatch repair defective phenotype even in the presence of a wild- type allele in the same cell. A non-limiting example of a dominant negative allele of a mismatch repair gene is the human gene hPMS2-134, which carries a truncation mutation at codon 134. The mutation causes the product of this gene to abnormally terminate at the position of the 134th amino acid, resulting in a shortened polypeptide containing the N- terminal 133 amino acids. Such a mutation causes an increase in the rate of mutations which accumulate in cells after DNA replication. Thus, expression of a dominant negative allele of a mismatch repair gene results in impairment of mismatch repair activity, even in the presence of the wild-type allele.
[0194] As used herein, "HAT-sensitive" refers to a lethal effect on cells when cultured in medium containing hypoxanthine, aminopterin and thymidine.
[0195] As used herein, "EBV-negative" refers to lack of infection of Epstein-Ban vims in a cell as measured by production of EBNA protein, or detection of EBV nucleic acids. [0196] As used herein, "Ig-negative" refers to lack of production in a cell of any light or heavy chains of immunoglobulins. [0197] As used herein, "screening" refers to an assay to assess the genotype or phenotype of a cell or cell product including, but not limited to nucleic acid sequence, protein sequence, protein function (e.g., binding, enzymatic activity, blocking activity, cross-blocking activity, neutralization activity, and the like). The assays include ELISA-based assays, Biacore analysis, and the like.
[0198] As used herein, "isolated" refers to a nucleic acid or protein that has been separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials which would interfere with diagnostic or therapeutic uses for the polypeptide, and may include enzymes, hormones, and other proteinaceous or non- proteinaceous solutes. In some embodiments, the nucleic acid or protein is purified to greater than 95% by weight of protein. In other embodiments, the nucleic acid or protein is purified to greater than 99% by weight of protein. Determination of protein purity may be by any means known in the art such as the Lowry method, by SDS-PAGE under reducing or non-reducing conditions using a stain such as a Coomassie blue or silver stain. Purification of nucleic acid may be assessed by any known method, including, but not limited to spectroscopy, agarose or polyacrylamide separation with fluorescent or chemical staining such as methylene blue, for example.
[0199] The invention provides an in vitro immunization method for obtaining antigen-specific immunoglobulin producing cells wherein the cells produce immunoglobulins of the IgG subclass, and cells produced by this method. The in vitro immunization procedure comprises combining donor cells with an immunogenic antigen in culture. In one embodiment, the buffy coat of donor cells is used. The donor may be from any source, including, but not limited to cord blood, venous blood, and the like. The source of the blood cells may be from any animal producing immune cells, particularly mammals. Non-limiting examples of blood cell sources include, mice, rats, humans, monkeys, dogs, cats, horses, pigs, sheep, goats, rabbits, birds, cows, guinea pigs and fish. The blood or buffy coat may be further enriched for lymphocytes by any known method, such as, but not limited to differential centrifugation, filtration, and the like.
[0200] Donor cells such as peripheral blood mononuclear cells (PBMC) may be incubated in L-leucyl-L-lysine methyl ester hydrobromide (LLOMe). While not wishing to be bound by any particular theory of operation, LLOme is believed to lysosomotropic and specifically kills cytotoxic cells in the PBMC pool such as NK cells, cytotoxic T cells, and CD8+ suppressor T cells, while not having an effect on B cells, T helper cells accessory cells and fibroblasts (Bonebaeck (1988) Immunol. Today 9(11):355-359). Generally, the PBMCs may be incubated with LLOMe for a period of 1-30 minutes. In some embodiments, the incubation is performed for 10-20 minutes. In other embodiments, the incubation is performed for 15 minutes. The LLOMe is generally a component of culture medium, such as, for example, RPMI 1640, and is provided in a concentration of about 0.10 to ImM. In some embodiments, LLOMe is provided in an amount of about 0.10 to 0.50 mM. In other embodiments, LLOMe is provided in an amount of about 0.25 mM.
[0201] The antigen may be any antigen provided that it is immunogenic. Whole proteins or peptides may be used. In addition, one may use, for example, membrane preparations (including those from tumors), lymphoma cells, whole cells, single cells, homogenized cells, pathogens, inclusion bodies, cell lysates, protein preparations, and minced tissue (including tumor tissue). Whole proteins may be in native or denatured conformation. Peptides may be conjugated to carrier molecules to provide immunogenicity. While not wishing to be bound by any particular theory of operation, carrier molecules may provide additional T cell epitopes which may be useful in stimulating a more robust in vitro antibody response. Examples of carriers that are suitable for use in the method of the invention include tetanus toxoid, diptheria toxin, thyroglobulin, cholera toxin, BCG, bovine serum albumen (BSA), ovalbumin (OVA), and the like. These carriers are refened to herein as "mitogenic polypeptides." [0202] Antigens may be conjugated to mitogenic polypeptides in any way known in the art. For example, fusion proteins may be generated by expressing a polypeptide in a recombinant expression system comprising the polynucleotide encoding at least a portion of the antigen joined in-frame to a polynucleotide encoding at least a portion of the mitogenic polypeptide. The fusion protein may have the mitogenic polypeptide joined at either the amino- or carboxy terminus of the antigen. In some embodiments, more that one antigen may be expressed as a fusion protein in combination with a mitogenic polypeptide. In other embodiments, more than one mitogenic polypeptide may be expressed as a fusion protein with the antigen or antigens. In other embodiments, more than one mitigenic polypeptide and more than one antigen may be expressed together as a single fusion protein.
[0203] In an alternative embodiment, at least a portion of the mitogenic polypeptide is conjugated to at least a portion of the antigen using chemical cross-linkers. Examples of chemical cross-linkers include, but are not limited to gluteraldehyde, formaldehyde, 1,1 -bis (diazoacetyl)-2-phenylethane, N-hydroxysuccinimide esters (e.g., esters with 4-azidosalicylic acid, homobifunctional imidoesters including disuccinimidyl esters such as 3,3'-dithiobis (succinimidyl-propionate). and bifunctional maleimides such as bis-N-maleimido-l,8-octane). Derivatizing agents such as methyl-3-[(p-azido-phenyl)dithio] propioimidate yield photoactivatable intermediates which are capable of forming cross-links in the presence of light. Alternatively, for example, a lysine residue in the mitogenic polypeptide or antigen may be coupled to a C-terminal or other cysteine residue in the antigen or mitogenic polypeptide, respectively, by treatment with N-γ-maleimidobutyryloxy-succinimide (Kitagawa and Aikawa (1976) J. Biochem. 79, 233-236). Alternatively, a lysine residue in the mitogenic polypeptide or antigen may be conjugated to a glutamic or aspartic acid residue in the antigen or mitogenic polypeptide, respectively, using isobutylchloroformate (Thorell and De Larson (1978) RADIOIMMUNOASSAY AND RELATED TECHNIQUES: METHODOLOGY AND CLINICAL APPLICATIONS, p. 288). Other coupling reactions and reagents have been described in the literature.
[0204] The conditions for the in vitro immunization procedure comprise incubating the cells at about 25-37°C, (preferably 37°C ) supplied with about 5-10% CO2. In some embodiments, the incubation is performed with between about 6-9% CO2. In other embodiments the incubation is performed in about 8% CO2. The cell density is between about 2.5 to 5 x 106 cells/ml in culture medium. In some embodiments, the culture medium is supplemented with about 2- 20% FBS. In other embodiments, the culture medium is supplemented with about 5-15% FBS. In other embodiments, the culture medium is supplemented with about 7-12% FBS. In other embodiments, the culture medium is supplemented with about 10% FBS. [0205] The in vitro stimulation culture medium is supplemented with cytokines to stimulate the cells and increase the immune response. In general IL-2 is supplied in the culture medium. However, other cytokines and additives may also be included to increase the immune response. Such cytokines and factors may include, for example, LL-4 and anti-CD40 antibodies.
[0206] The fusion of myeloma cells with the immunoglobulin-producing cells may be by any method known in the art for the creation of hybridoma cells. These methods include, but are not limited to, the hybridoma technique of Kohler and Milstein, (1975, Nature 256:495-497; and U.S. Patent No. 4,376,110) (see also, Brown et al. (1981) J. Immunol. 127:539-546; Brown et al. (1980) J. Biol. Chem. 255 (ll):4980-4983; Yeh et al. (1976) Proc. Natl. Acad. Sci. USA 76:2927-2931; and Yeh et al. (1982) Int. J. Cancer 29:269-275), the human B-cell hybridoma technique (Kosbor et al, 1983, Immunology Today 4:72; Cole et al, 1983, Proc. Natl. Acad. Sci. USA 80:2026-2030), and the EBV-hybridoma technique (Cole et al, 1985, MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96). The hybridoma producing the mAb of this invention may be cultivated in vitro or in vivo. [0207] The technology for producing monoclonal antibody hybridomas is well-known to those of skill in the art and is described, for example in Kenneth, in MONOCLONAL ANTIBODIES: A NEW DIMENSION IN BIOLOGICAL ANALYSES, Plenum Publishing Corp., New York, N.Y. (1980); Lerner (1981) Yale J. Biol. Med., 54:387-402; Galfre et al. (1977) Nature 266:55052; and Gefter et al. (1977) Somatic Cell Genet. 3:231-236). However, many variations of such methods are possible and would be appreciated by one of skill in the art. Thus, the techniques for generation of hybridomas is not limited to the disclosures of these references. [0208] Any myeloma cell may be used in the method of the invention. Preferably, the myeloma cells are human cells, but the invention is not limited thereto or thereby. In some embodiments, the cells are sensitive to medium containing hypoxanthine, aminopterin, an thymidine (HAT medium). In some embodiments, the myeloma cells do not express iimnunoglobulin genes. In some embodiments the myeloma cells are negative for Epstein- Ban virus (EBV) infection. In prefened embodiments, the myeloma cells are HAT-sensitive, EBV negative and Ig expression negative. Any suitable myeloma may be used. An example of such a myeloma is that described in U.S. Patent No. 4,720,459 to Winkelhake, and deposited with the American Type Culture Collection (ATCC) as CRL 8644. Murine hybridomas may be generated using mouse myeloma cell lines (e.g., the P3-NSl/l-Ag4-l, P3- x63-Ag8.653 or Sp2/O-Agl4 myeloma lines). These murine myeloma lines are available
Figure imgf000049_0001
[0209] In some embodiments of the method of the invention, the hybridoma cells and/or mammalian expression cells may be rendered hypermutable by the introduction of a dominant negative allele of a mismatch repair gene. The dominant negative allele of the mismatch repair gene may be introduced into the hybridoma cell (i.e., after the fusion of immunoglobulin-producing cells with the myeloma cells) or may be introduced into the myeloma cell prior to the fusions. The invention, therefore, also provides hypermutable myeloma cells for use in the generation of hybridoma cells. The dominant negative allele may also be introduced into the mammalian expression cells.
[0210] The dominant negative allele of the mismatch repair gene is in the form of a polynucleotide which may be in the form of genomic DNA, cDNA, RNA, or a chemically synthesized polynucleotide. The polynucleotide can be cloned into an expression vector containing a constitutively active promoter segment (such as, but not limited to, CMV, SV40, EF-1 Dor LTR sequences) or to inducible promoter sequences such as those from tefracycline, or ecdysone/glucocorticoid inducible vectors, where the expression of the dominant negative mismatch repair gene can be regulated. The polynucleotide can be introduced into the cell by transfection.
[0211] Transfection is any process whereby a polynucleotide is introduced into a cell. The process of transfection can be carried out in vitro, e.g., using a suspension of one or more isolated cells in culture. The cell can be any immortalized cell used for creating hybridomas for the production of monoclonal antibodies, or the cell may be the hybridoma itself. The hybridomas may be heterohybridoma cells (e.g. human-mouse cell fusions) or homohybridoma cells (e.g., human-human hybridoma cells and mouse-mouse hybridoma cells).
[0212] In general, transfection will be carried out using a suspension of cells, or a single cell, but other methods can also be applied as long as a sufficient fraction of the treated cells or tissue incorporates the polynucleotide so as to allow transfected cells to be grown and utilized. The protein product of the polynucleotide may be transiently or stably expressed in the cell. Techniques for transfection are well known. Available techniques for introducing polynucleotides include but are not limited to electroporation, transduction, cell fusion, the use of calcium chloride, and packaging of the polynucleotide together with lipid for fusion with the cells of interest. Once a cell has been transfected with the mismatch repair gene, the cell can be grown and reproduced in culture. If the transfection is stable, such that the gene is expressed at a consistent level for many cell generations, then a cell line results. [0213] The dominant negative allele of the mismatch repair gene may be derived from any known mismatch repair gene including, but not limited to PMS2, PMSl, PMSR3, PMSR2, PMSR6, MLHl, GTBP, MSH3, MSH2, MLH3, or MSH1, and homologs of PMSR genes as described in Nicolaides et αl (1995) Genomics 30:195-206 and Horii et αl (1994) Biochem. Biophys. Res. Commun. 204:1257-1264 and the like. "Dominant negative alleles" as used herein, refers to the ability of the allele to confer a hypermutable status to the cell expressing the allele. Any allele which produces such effect can be used in this invention. The dominant negative alleles of a mismatch repair gene can be obtained from the cells of humans, animals, yeast, bacteria, or other organisms. Dominant negative alleles of mismatch repair genes that are suitable for use in the invention have certain functional characteristics associated with structural features. A non-limiting example of a dominant negative mismatch repair gene is the PMS2 truncation mutant, PMS2-134. This gene contains a mutation which truncates the PMS2 protein after amino acid 133. The lack of the C-terminus in the PMS2 protein is believed to interfere with the binding of PMS2 with Screening cells for defective mismatch repair activity can identify such alleles. Cells from animals or humans with cancer can be screened for defective mismatch repair. Cells from colon cancer patients may be particularly useful. Genomic DNA, cDNA, or mRNA from any cell encoding a mismatch repair protein can be analyzed for variations from the wild type sequence. Dominant negative alleles of a mismatch repair gene can also be created artificially, for example, by producing variants of the hPMS2-134 allele or other mismatch repair genes. Various techniques of site-directed mutagenesis can be used. The suitability of such alleles, whether natural or artificial, for use in generating hypermutable cells or animals can be evaluated by testing the mismatch repair activity caused by the allele in the presence of one or more wild-type alleles, to determine if it is a dominant negative allele.
[0214] Dominant negative alleles of such genes, when introduced into cells or transgenic animals, increase the rate of spontaneous mutations by reducing the effectiveness of DNA repair and thereby render the cells or animals hypermutable. This means that the spontaneous mutation rate of such cells or animals is elevated compared to cells or animals without such alleles. The degree of elevation of the spontaneous mutation rate can be at least 2-fold, 5-fold, 10-fold, 20-fold, 50-fold, 100-fold, 200-fold, 500-fold, or 1000-fold that of the normal cell or animal. The hypermutable hybridoma cells will accumulate new mutations in gene(s) to produce new output traits within the hybridoma. The hybridoma cells can be screened for desired characteristics and cell lines bearing these characteristics may be expanded. Furthermore, the hybridoma cells may be "cured" of the mismatch repair defect by eliminating the dominant negative mismatch repair gene in the cell or by turning of its expression, leading to stable biological products consisting of altered genes, RNAs, or polypeptides. [0215] The dominant negative alleles of the mismatch repair gene may be introduced as part of a vector. The polynucleotide encoding the dominant negative mismatch repair protein allele may be operably linked to a promoter that functions in the cell to drive expression of the dominant negative allele of the mismatch repair gene. Other elements of the vector may include an origin of replication, one or more selectable markers, such as a drug resistance gene that allows the cells to grow in the presence of a growth inhibitory compound. [0216] In embodiments of the invention that utilize myeloma cells or donor immunoglobulin- producing cells that are naturally deficient in mismatch repair, the invention may further comprise the step of restoring genetic stability of the hybridoma by introducing a wild-type mismatch repair gene into the cell to complement the deficiency and restore genetic stability. [0217] Another aspect of the invention is the use of cells lacking MMR (either due to defects in endogenous mismatch repair genes, or due to the introduction of a dominant negative MMR gene) and chemical mutagens to cause an enhanced rate of mutations in a host's genome. The lack of MMR activity has been known to make cells more resistant to the toxic effects of DNA damaging agents. This invention comprises making proficient MMR cells mismatch repair defective via the expression of a dominant negative MMR gene allele and then enhancing the genomic hypermutability with the use of a DNA mutagen. Chemical mutagens are classifiable by chemical properties, e.g., alkylating agents, cross-linking agents, etc. The following chemical mutagens are useful, as are others not listed here, according to the invention and may be used to further enhance the rate of mutation in any of the embodiments of the method of the invention: N-ethyl-N-nitrosourea (ENU), N-methyl-N-nitrosourea (MNU), procarbazine hydrochloride, chlorambucil, cyclophosphamide, methyl methanesulfonate (MMS), ethyl methanesulfonate (EMS), diethyl sulfate, acrylamide monomer, triethylene melamin (TEM), melphalan, nitrogen mustard, vincristine, dimethylnitrosamine, N-methyl-N'-nitro- nitrosoguanidine (MNNG), 7,12 dimethylbenz (a) anthracene (DMBA), ethylene oxide, hexamethylphosphoramide, bisulfan. In a prefened aspect of the invention, a mutagenesis technique is employed that confers a mutation rate in the range of 1 mutation out of every 100 genes; 1 mutation per 1,000 genes. The use of such combination (MMR deficiency and chemical mutagens will allow for the generation of a wide anay of genome alterations (such as but not limited to expansions or deletions of DNA segments within the context of a gene's coding region, a gene's intronic regions, or 5'or 3' proximal and/or distal regions, point mutations, altered repetitive sequences) that are preferentially induced by each particular agent.
[0218] Mutations can be detected by analyzing for alterations in the genotype of the cells or animals, for example by examining the sequence of genomic DNA, cDNA, messenger RNA, or amino acids associated with the gene of interest. Mutations can also be detected by screening the phenotype of the gene. An altered phenotype can be detected by identifying alterations in electrophoretic mobility, spectroscopic properties, or other physical or structural characteristics of a protein encoded by a mutant gene. One can also screen for altered function of the protein in situ, in isolated form, or in model systems. One can screen for alteration of any property of the cell or animal associated with the function of the gene of interest, such as but not limited to measuring protein secretion, chemical-resistance, pathogen resistance, etc. [0219] In some embodiments of the method of the invention, inducible vectors that control the expression of a dominant negative and normally functioning MMR gene are used. This strategy restores DNA stability once a host cell or organism exhibiting a new output trait, altered gene, RNA or polypeptide has been generated via trait selection with or without the combination of chemical mutagens to establish a genetically stable version of this cell or organism. In the case of MMR defective cells as a result of ectopically expressing a dominant negative MMR gene allele, the MMR activity is decreased or completely eliminated by removing the inducer molecule from the cell culture or organism's environment. In addition, the expression of a dominant negative MMR gene can be suppressed by knocking out the MMR gene allele using methods that are standard to those skilled in the art of DNA knockout technology in germ or somatic cells (Waldman et al. (1995) Cancer Res. 55:5187-5190). [0220] The chiral position of the side chains of the anthracenes is not particularly limited and may be any chiral position and any chiral analog. The anthracenes may also comprise a stereoisomeric form of the anthracenes and include any isomeric analog. [0221] Examples of hosts are but not limited to cells or whole organisms from human, primate, mammal, rodent, plant, fish, reptiles, amphibians, insects, fungi, yeast or microbes of prokaryotic origin.
[0222] A more detailed disclosure of particular embodiments of the invention follows in the specific examples, however, the invention is not limited thereto or thereby.
EXAMPLES Example 1: Generation of Hybridomas Secreting Human Monoclonal Antibodies to Tetanus Toxin (TT)
A. Generation and Assaying of TT-specific B lymphocytes
[0223] Isolation of lymphocytes from Donor. Lymphocytes were isolated from whole blood by centrifugation through Ficoll-Paque according to the manufacturer's instructions. Isolated lymphocytes were incubated with 0.25mM Leu-Leu methyl ester hydrobromide (LLOMe) prepared in RPMI 1640 medium containing 2% fetal bovine serum (FBS) for 15 minutes at room temperature. The cells were then washed three times with culture medium. [0224] In vitro stimulation of isolated lymphocytes. The cells were incubated at 37°C in a incubator, supplied with 8% CO2, at a density between 2.5 to 5 x 10 cells/ml in culture medium supplemented with 10% FBS and TT and IL-2 at various concentrations. After four days of culture, the cells were washed four times with medium and the culture was continued for additional eight days.
[0225] Measurement of the B cell response. Lymphocyte culture supernatants were collected on day 12 of the culture and tested in an ELISA for the presence of anti-TT antibodies. Briefly, TT or BSA at 0.5 μg/ml in 0.05 M carbonate-bicarbonate buffer was immobilized onto an EIA plate. After blocking with 1% bovine serum albumin (BSA) in PBS containing 0.05% Tween 20, the supernatant was added to the wells. Antibodies bound to TT were detected with peroxidase-labeled goat anti-human IgG or anti-human IgM. TMB was used for color development. The plate was read using a Microplate reader with a 450 nm filter. A supernatant sample that had antibody bound to TT, but not to BSA, and in which the signal was two times the assay background was considered positive. The positive cells were pooled, and used for hybridoma production (Fig. 1).
[0226] Notably, peripheral blood mononuclear cells (PBMCs) from some donors contain a fraction of B cells that secret TT-specific antibodies in culture. This is due to the fact that about 90%) of the population in the United States has been vaccinated against TT. Such sera also has a titer of higher than 1000 (Fig. 2). However, the percentage of positive events is greatly increased when PBMCs are immunized in vitro with TT (Fig. 3). The intensity of the PBMC response is also enhanced with the stimulation of TT alone or in combination with IL-2 or CD40L (Fig. 4). Similar effects were observed with other antigens (data not shown). [0227] Generation of hybridomas secreting human antibodies. To prepare activated lymphocytes, cells were pooled and cultured in T flasks at 0.5 - 1 x 106 cells/ml in culture medium supplemented with 10% FBS one day prior to the fusion. To prepare the fusion partner, mouse myeloma NS0 cells were transfected with human PMS2-134 expression vector as described in Nicolaides et al. (1998) Mol. Cell. Biol. 18(3):1635-1641. The cells were cultured in RPMI 1640 supplemented with 10% FBS and 2 mM glutamine (Complete Medium) and the culture was kept in log phase.
[0228] Next, lymphocytes were harvested and counted. An equal number of myeloma cells was harvested. Both types of cells were combined and washed three times with RPMI 1640 medium. Polyethylene glycol (PEG) was added dropwise to the loosened cell pellet, and the PEG was subsequently diluted out slowly with 25 ml of RPMI medium in a course of 2.5 minutes. After diluting out the PEG, fused cells were suspended in Complete Medium supplemented with HAT and 20% FBS, and seeded onto 96-well plates. [0229] Screening and characterization of antigen-specific hybridoma clones. When the hybridoma cells grew to semi-confluence, supernatants were collected and subjected to an ELISA for antigen-specific reactivity. As an example, hybridomas derived from TT- immunized lymphocytes were tested. Briefly, TT or BSA at 0.5 ug/ml in 0.05 M carbonate- bicarbonate buffer was immobilized onto the EIA plate. After blocking with 1% bovine serum albumen in PBS containing 0.05%ι Tween 20, the cell culture supernate was added to the wells. Antibodies bound to TT were detected with peroxidase-labeled goat anti-human IgG or anti-human IgM. TMB was used for color development. The plate was read in the Microplate reader with a 450 nm filter. A cell clone that showed reactivity to TT but not to BSA was considered positive (Fig. 5). Positive clones were expanded and subcloned by limiting dilution to generate monoclonal cells.
Example 2: Generation of hybridomas secreting human monoclonal antibodies to epidermal growth factor receptor (EGFR) (self antigen)
A. Generation of EGFR-specific B lymphocytes
[0230] Preparation of Antigen. Human epidermal growth factor receptor (EGFR), purified from A431 cells, was purchased from Sigma. Previous studies found that immune responses to this antigen were very weak, most likely due to tolerance. In order to enhance immunization, we conjugated the EGFR to tetanus toxin C (EGFR-TT) and the conjugate was used as immunogen for in vitro immunization in order to overcome any immunotolerance.
[0231] Preparation of EGFR-TT conjugate. 100 ug of purified EGFR was reconstituted in
100 ul of sterile MilliQ-grade water. 1 mg of purified, lyophilized recombinant tetanus toxin
C fragment (TT-C) was dissolved in sterile MilliQ-grade water to yield a 2 mg/ml TT-C solution. Crosslinking was performed in 50 mM sodium carbonate buffer pH 9.0 at equimolar ratios of EGFR to TT-C, using glutaraldehyde at a final concentration of 0.5% for 3 hours at room temperature, followed by 4°C overnight. Glutaraldehyde was quenched by addition of a fresh 100 mg/ml solution of sodium borohydride in 50 mM sodium carbonate pH 9.0, under open atmosphere for 1 hour at 4°C. Crosslinked products were dialyzed against Ca2+-, Mg2+- free phosphate-buffered saline overnight at 4°C, using 3.5K MWCO Slide- A-Lyzer cassettes.
The reaction was monitored by Western blotting, using commercial anti-EGFR (mAb- 15) and anti-TT-C (Roche) monoclonal antibodies. By this method, greater than 70%) of the components are crosslinked, and appear as immunoreactive species of greater MW than the starting material (data not shown).
[0232] In vitro stimulation of peripheral blood mononuclear cells (PBMC). LLOMe- pretreated PBMC were incubated at a density of 3 x 106 cells/ml in culture medium supplemented with 10% FBS and a stimuli mixture. The stimuli mixture was composed of
EGFR-TT at a concentration of 50 ng/ml with or without recombinant human IL-2 at 20 IU, mouse anti-human CD40 antibody as CD40L at 0.5 ug/ml (used to enhance IgG class switching). After four days of culture, the cells were re-fed with complete medium, in the absence of added stimulus, every three or four days. Culture supernatants were collected on days 12-18 and tested for EGFR-specific antibodies.
[0233] Detection of EGFR-specific antibody response. The PBMC response to the stimulation was examined in a EGFR-specific ELISA. Briefly, EGFR, TT, or BSA at 0.5 ug/ml in 0.05 M carbonate-bicarbonate buffer, pH 9.6, was immobilized onto EIA plates. After blocking the plates with 5% non-fat dry milk in PBS containing 0.05% Tween 20, the supernatant was added to the wells. Antibodies from the supernatant bound to immobilized antigens were detected with peroxidase-labeled goat anti-human IgG+IgM (H+L). TMB substrate kit was used for color development. The plates were read in a Microplate reader with a 450 nm filter. A supernatant sample containing antibody that bound to EGFR, but not to TT and BSA, was considered positive. A robust response was observed in cultures immunized to the EGFR-TT as compared to controls. While anti-EGFR responses were observed in PBMCs for a small fraction of donors, the percentage of positive clones was greatly increased when PBMCs were immunized in vitro with EGFR complexed with TT (Fig. 6). Positive cells were pooled and used for hybridoma production.
[0234] Generation of Hybridomas. To prepare activated lymphocytes, cells were pooled and cultured in T flasks at 0.5 - 1 x 106 cells/ml in culture medium supplemented with 10% FBS one day prior to the fusion. To prepare the fusion partner, mouse myeloma NS0 cells were transfected with human PMS2-134 expression vector as described in Nicolaides et al. (1998) Mol. Cell. Biol. 18(3): 1635-1641. The cells were cultured in RPMI 1640 supplemented with 10% FBS and 2 mM glutamine (Complete Medium) and the culture was kept in log phase. [0235] Screening and characterization of antigen-specific hybridoma clones. When the hybridoma cells grew to semi-confluence, supernatants were collected and subjected to an ELISA for antigen-specific reactivity. As an example, hybridomas derived from EGFR- immunized lymphocytes were tested. Briefly, EGFR, TT TNFRl or BSA at 0.5 ug/ml in 0.05 M carbonate-bicarbonate buffer was immobilized onto the EIA plate. After blocking with 1% bovine serum albumen in PBS containing 0.05% Tween 20, the cell culture supernate was added to the wells. Antibodies bound were detected with peroxidase-labeled goat anti-human IgG or anti-human IgM. TMB was used for color development. Normal human IgG (nhlgG) and IgM (nhlgM) were used as controls. The plate was read in the Microplate reader with a 450 nm filter. A cell clone that showed reactivity to EGFR, but not to BSA was considered positive (Fig. 7).
Example 3
A. Isolation of PBMC from whole blood
[0236] Approximately 200ml of whole blood mixed with 200ml PBS"7" was centrifuged through Ficoll-Paque at 2000rpm for 30min. Serum was aspirated, the interface layer containing lymphocytes was collected and diluted 1 :3 with PBS"7" and centrifuged at 2000 rpm for lOmin. The supernatant fluid was aspirated and the pellet was resuspended in 10 ml PBS"7". The cell suspension was split into two 50 ml conical tubes and PBS"7" was added to each tube to adjust the volume to 35 ml each. The tubes were centrifuged at 800 rpm for 7 minutes to remove the platelets. After aspirating the supernatant fluid, the pellet was resuspended in 10 ml ACK Lysing Buffer and incubated for 5 minutes at room temperature. Following lysis, 35 ml PBS" " was added to the tubes and the tubes were centrifuged at 1000 rpm for 7 minutes. The cells were then washed with 45 ml RPMI medium.
B. Preparation of Dendritic Cells
[0237] Cells were centrifuged at 1000 rpm for 7 minutes and resuspended at 1 x 108 cells per 40 ml cRPMI for a density of 2.5 x 106 cells/ml. The cells were incubated at 37°C/8% CO2 for 2 hours. Non-adherent cells were removed for further treatment (see Step C), and the adherent cells were carefully rinsed twice with PBS"7". Adherent cells were cultured in cRPMI supplemented with 400 JU/ml IL-4 and 50 ng/ml GM-CSF.
C. LLOMe treatment and cryopreservation of non-adherent culture
[0238] The non-adherent cell culture was centrifuged at 1000 rpm for 7 minutes. The supernatant fluid was aspirated and the pellet was resuspended in 10 ml RPMI supplemented with 2% FBS and freshly thawed 85 μg/ml LLOMe. The cells were incubated for 15 minutes at room temperature. The cells were washed twice with cRPMI and resuspended in 45 ml cRPMI. The cells were transfened to an upright T25 flask at a density of 5 x 10 cells/ml in cRPMI supplemented with 2 μg/ml PHA and incubated at 37°C/8% CO2 for 24 hours. The non-adherent cells were harvested, centrifuged at 1000 rpm for 7 minutes, and the cell pellet was resuspended in 5 ml cold cRPMI/5%DMSO. The tubes containing the cells were wrapped in paper towels and stored at -80° C until needed.
D. Tumor Immunization
[0239] On day 6 of the procedure for isolation of dendritic cells, tumor cells were thawed in 2.5 ml pre-warmed medium at 37°C. The flask of dendritic cells was rinsed twice with 10 ml PBS"7". The dendritic cells were incubated with gentle rocking in 5 ml Cell Dissociation Buffer (Invitrogen Cat. No. 13151-014), and the solution was collected (scraping the remaining cells from the flask. The flask was rinsed with 10 ml cRPMI and the medium was collected. The cells were centrifuged at 1000 rpm for 7 minutes and the pellet was resuspended at 4 x 106 cells/ml cRPMI. Cells were distributed in a culture plate at a density of 1 x 106 cells/well. A tumor sample was chopped into fine pieces of approximately 1-3 mm3. An aliquot of the tumor suspension was transfened to all but 1 well, titrating the amount of tumor per well. An aliquot of 0.25 ml cRPMI was added to the control well. The total volume in the wells was 0.5ml/well. The dendritic cells and tumor cells were co-cultured at 37°C/8% CO2 for 24 hours.
E. Co-culture of PBMC with DC
[0240] Frozen PBMC were thawed by adding 40 ml cRPMI/30 JU/ml IL-2/600 JU/ml IL- 4/0.75 μg/ml CD-40L pre-warmed to 50°C to the frozen cells. When thawed, the cells were incubated for 1-2 hours at 37°C. The cells were centrifuged at 1000 rpm for 7 minutes and the pellet was resuspended in 5 ml of a 2X cocktail of cRPMI/ 60 JU/ml IL-2/1200 IU/ml IL- 4/1.5 μg/ml CD-40L. The cell suspension was divided among wells in a tissue culture plate at 0.5 ml/well of suspension and diluted with 0.5 ml medium for a final concentration of 30 IU/ml IL-2, 600 IU/ml IL-4, and 0.75 μg/ml CD-40L. Cells were fed with cRPMI supplemented with 20 IU/ml IL-2, 400 JU/ml IL-4, 100 IU/ml IL-10, and 0.5μg/ml CD-40L.
F. Fusion
[0241] Tumor-immunized PBMCs were then fused with A6 myeloma cells to generate hybridomas. Briefly, lymphocytes were harvested from 75% tumor and 100% tumor wells, rinsed with 1 ml RPMI, transfer to conical tubes, and the volume was adjusted to 5 ml with cRPMI. The cells were centrifuged through Ficoll-Paque, and the supernatant fluid was aspirated. Interfaces containing cells from all tubes were combined and the cells were rinsed with cRPMI. The cells were then resuspended in 7.5 ml cRPMI. Viable cells were assessed by trypan blue exclusion. A6 cell viability was also assessed by trypan blue exclusion. A6 cells and tumor-immunized lymphocytes were centrifuged separately at 1200 m for lOminutes. The supernatant fluids were aspirated and the cells were washed with 10ml DPBS "7Jtube. Each cell line was washed three times with 2 ml cold Mannitol Fusion Medium (MFM) (0.3M Mannitol, 0.18mM MgCl2, 0.18mM CaCl2, ImM Hepes) and the cells were combined and resuspended in MFM at a density of 3 x 106 A6 cells and 3 x 106 PBMCs in 200 μl for a total of 6 x 106 cells in 200 μl. BTX 450 microslides were sterilized with 65μL 100% EtOH and pre-wetted with 65 μl MFM. A 40μl aliquot of cell suspension was distributed evenly onto a BTX 450-1 microslide. To fuse the cells, the ECM 2001 conditions were set as follows: alignment conditions, 20V for 30 seconds; pulse conditions, 150V for 30 μseconds (IX); compression conditions, 20V for 9 seconds. After fusion, the cells were transfened to one well of a 24 well plate containing 1 ml phenol red-free cRPMI. The fusion steps were repeated for the remaining cell suspensions, rinsing slide between fusions with 65 μL MFM. The culture plate containing fused cell cultures was incubated overnight at 37°C/8%CO2. The fused cells were cloned and assessed by ELISA for IgG and IgM production. The results are shown in Fig. 8.
Example 4: In vitro immunization
Purified GM-CSF from a commercial source is administered in vitro to peripheral blood mononuclear cells (PBMC).
A. Generation and Assaying of GM-CSF-specific B lymphocytes
Isolation of lymphocytes from Peripheral Blood.
[0242] Lymphocytes are isolated from whole blood by centrifugation through Ficoll-Paque according to the manufacturer's instructions. Isolated lymphocytes are incubated with 0.25 mM Leu-Leu methyl ester hydrobromide (LLOMe) prepared in RPMI 1640 medium containing 2% fetal bovine serum (FBS) for 15 minutes at room temperature. The cells are then washed three times with culture medium.
In vitro stimulation of isolated lymphocytes.
[0243] The cells are incubated at 37 C in an incubator, supplied with 8% CO2, at a density between 2.5 to 5 x 106 cells/ml in culture medium supplemented with 10% FBS and GM-CSF and IL-2 at various concentrations. After four days of culture, the cells are washed four times with medium and the culture was continued for additional eight days.
Measurement of the B cell response.
[0244] Lymphocyte culture supematants are collected on day 12 of the culture and tested in an ELISA for the presence of anti-GM-CSF antibodies. Briefly, GM-CSF or BSA at 0.5 μg/ml in 0.05 M carbonate-bicarbonate buffer is immobilized onto an EIA plate. After blocking with 1% bovine serum albumin (BSA) in PBS containing 0.05% Tween 20, the supernatant is added to the wells. Antibodies bound to GM-CSF are detected with peroxidase-labeled goat anti-human IgG or anti-human IgM. TMB is used for color development. The plate is read using a Microplate reader with a 450 nm filter. A supernatant sample that had antibody bound to GM-CSF, but not to BSA, and in which the signal was two times the assay background is considered positive. The positive cells are pooled, and used for hybridoma production.
Generation of hybridomas secreting human antibodies.
[0245] To prepare activated lymphocytes, cells are pooled and cultured in T flasks at 0.5 - 1 x 106 cells/ml in culture medium supplemented with 10% FBS one day prior to the fusion. To prepare the fusion partner, mouse myeloma NS0 cells are transfected with human PMS2-134 expression vector as described in Nicolaides et al. (1998) Mol. Cell. Biol. 18(3):1635-1641. The cells are cultured in RPMI 1640 supplemented with 10% FBS and 2 mM glutamine (Complete Medium) and the culture is kept in log phase.
[0246] Next, lymphocytes are harvested and counted. An equal number of myeloma cells is harvested. Both types of cells are combined and washed three times with RPMI 1640 medium. Polyethylene glycol (PEG) is added dropwise to the loosened cell pellet, and the PEG is subsequently diluted out slowly with 25 ml of RPMI medium in a course of 2.5 minutes. After diluting out the PEG, fused cells are suspended in Complete Medium supplemented with HAT and 20% FBS, and seeded onto 96-well plates.
Screening and characterization of antigen-specific hybridoma clones.
[0247] When the hybridoma cells grew to semi-confluence, supematants are collected and subjected to an ELISA for antigen-specific reactivity. As an example, hybridomas derived from TT-immunized lymphocytes are tested. Briefly, TT or BSA at 0.5 ug/ml in 0.05 M carbonate-bicarbonate buffer is immobilized onto the EIA plate. After blocking with 1% bovine serum albumin in PBS containing 0.05% Tween 20, the cell culture supernate is added to the wells. Antibodies bound to GM-CSF are detected with peroxidase-labeled goat anti- human IgG or anti-human IgM. TMB is used for color development. The plate is read in the Microplate reader with a 450 nm filter. A cell clone that showed reactivity to GM-CSF but not to BSA is considered positive. Positive clones are expanded and subcloned by limiting dilution to generate monoclonal cells.
Example 5: Generation of hybridomas secreting human monoclonal antibodies to GM- CSF-KLH
A. Generation of GM-CSF-specific B lymphocytes Preparation of Antigen.
[0248] Human GM-CSF was purchased from a vendor. In order to enhance immunization, GM-CSF was conjugated to keyhole limpet hemocyanin (KLH) (GM-CSF-KLH) and the conjugate was used as immunogen for in vitro immunization in order to overcome any immunotolerance.
Preparation of GM-CSF-KLH conjugate.
[0249] Purified GM-CSF was reconstituted in sterile MilliQ-grade water to yield a lmg/ml solution. Purified, lyophilized recombinant KLH was dissolved in sterile MilliQ-grade water to yield a 1 mg/ml KLH solution. A 0.2% solution of glutaraldehyde in PBS was prepared. Crosslinking was performed by combining 25 ul of 1 mg/ml KLH, 25 ul of 1 mg/ml GM-CSF, and 50 ul 0.2% glutaraldehyde in a microcentrifuge tube wrapped in aluminum foil at room temperature, with shaking for 1 hour. Following cross-linking, 25 ul of 1 M glycine was added to the tube and the solution was incubated an additional 1 hour at room temperature with shaking. Crosslinked products were dialyzed against three changes of 300 ml PBS. The reaction was monitored by Western blotting, using a commercial anti-GM-CSF and anti-KLH monoclonal antibodies. By this method, greater than 80% of the components are crosslinked, and appeared as immunoreactive species of greater MW than the starting material (data not shown).
In vitro stimulation of peripheral blood mononuclear cells (PBMC).
[0250] LLOMe-pretreated PBMC were incubated at a density of 3 x 106 cells/ml in culture medium supplemented with 10%o FBS and a stimuli mixture. The stimuli mixture was composed of GM-CSF-KLH at a concentration of 50 ng/ml with or without recombinant human IL-2 at 20 IU, mouse anti-human CD40 antibody as CD40L at 0.5 ug/ml (used to enhance IgG class switching). After four days of culture, the cells were re-fed with complete medium, in the absence of added stimulus, every three or four days. Culture supematants were collected on days 12-18 and tested for GM-CSF-specific antibodies.
Detection of GM-CSF-specific antibody response. [0251] The PBMC response to the stimulation was examined in a GM-CSF-specific ELISA. Briefly, GM-CSF, KLH, or chick ovalbumin (CAB) at 0.5 ug/ml in 0.05 M carbonate- bicarbonate buffer, pH 9.6, was immobilized onto EIA plates. After blocking the plates with 1%) BSA containing 0.05% Tween 20, the supernatant were added to the wells. Antibodies from the supernatant bound to immobilized antigens were detected with peroxidase-labeled goat anti-human IgG+IgM (H+L). TMB substrate kit was used for color development. The plates were read in a Microplate reader with a 450 nm filter. A supernatant sample containing antibody that bound to GM-CSF, but not to KLH and CAB, was considered positive. There was a robust response observed in cultures immunized to the GM-CSF-KLH as compared to controls. While anti-GM-CSF responses were observed in PBMCs for a small fraction of donors, the percentage of positive clones was greatly increased when PBMC were immunized in vitro with GM-CSF complexed with KLH. Positive cells were pooled and used for hybridoma production.
Generation of Hybridomas.
[0252] To prepare activated lymphocytes, cells were pooled and cultured in T flasks at 0.5 - 1 x 106 cells/ml in culture medium supplemented with 10% FBS one day prior to the fusion. To prepare the fusion partner, mouse myeloma NSO cells were transfected with human PMS2-134 expression vector as described in Nicolaides et al. (1998) Mol. Cell. Biol. 18(3):1635-1641. The cells were cultured in RPMI 1640 supplemented with 10% FBS and 2 mM glutamine (Complete Medium) and the culture was kept in log phase.
Screening and characterization of antigen-specific hybridoma clones.
[0253] When the hybridoma cells grew to semi-confluence, supematants were collected and subjected to an ELISA for antigen-specific reactivity. As an example, hybridomas derived from GM-CSF-immunized lymphocytes were tested. Briefly, GM-CSF, KLH, or CAB at 0.5 ug/ml in 0.05 M carbonate-bicarbonate buffer was immobilized onto the EIA plate. After blocking with 1% bovine serum albumin in PBS containing 0.05% Tween 20, the cell culture supernate was added to the wells. Antibodies bound were detected with peroxidase-labeled goat anti-human IgG or anti-human IgM. TMB was used for color development. Normal human IgG (nhlgG) and IgM (nhlgM) were used as controls. The plate is read in the Microplate reader with a 450 nm filter. A cell clone that showed reactivity to GM-CSF, but not to CAB was considered positive. The results are shown in Fig. 9.
Example 6
Inhibition of Proliferation Assays
[0254] TF-1 cells were seeded at 0.2 x 106/ml in RPMI supplemented with 10% FBS and 0.5 ng/ml recombinant human GM-CSF. TF-1 cells were serum starved for 24 hours in medium containing 0.5% BSA, without rhGM-CSF. Cells were then cultured in the presence of 0.275ng/ml of GM-CSF for 3 days, with or without 4 ug/ml of various antibodies. Cell proliferation was measured using the ATPLite assay (Perkin Elmer). In this assay, ATP was released by lysis of viable cells and utilized by the enzyme luciferase to convert luciferin into oxyluciferin. Light was emitted (luminescence) as a result of the reaction, and the intensity of the emission was ultimately proportional to the ATP content and thus to the cell number. Counts per second (CPS) were obtained by reading the reactions with a luminometer and the percentage of inhibition was calculated according to the formula: 100 - (CPS no Ab : CPS with Ab) x 100%). The results are shown in Fig. 10.
[0255] The foregoing examples are merely illustrative of the invention and are not to be construed to limit the scope of the invention in any way. The scope of the invention is defined by the appended claims.

Claims

What is claimed is:
1. A method for producing hybridoma cells producing Mgh-affinity antibodies from in vitro immunized immunoglobulin-producing cells comprising:
(a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro;
(b) fusing said immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein said hybridoma cells express a dominant negative allele of a mismatch repair gene;
(c) incubating said parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells;
(d) performing a screen for binding of antibodies to antigen for antibodies produced from said hypermutated hybridoma cells; and
(e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for said antigen than antibodies produced by said parental hybridoma cells; thereby producing hybridoma cells producing high-affinity antibodies.
2. The method of claim 1 wherein said dominant negative allele of a mismatch repair gene comprises a dominant negative allele of a gene selected from the group consisting of PMS2, PMSl, PMSR3, PMSR2, PMSR6, MLHl, GTBP, MSH3, MSH2, MLH3, oxMSHl, and homologs of PMSR genes.
3. The method of claim 1 wherein said dominant negative allele of a mismatch repair gene comprises a dominant negative allele of a PMS2 gene.
4. The method of claim 1 further comprismg a screen for hypermutated hybridomas that also produce antibodies in higher titers than said parental hybridomas.
5. The method of claim 1 further comprising inactivation of said dominant negative allele of said mismatch repair gene, thereby stabilizing the genome of said hypermutated hybridoma.
6. The method of claim 4 further comprising inactivation of said dominant negative allele of said mismatch repair gene, thereby stabilizing the genome of said hypermutated hybridoma.
7. The method ofclaim 1 wherein said high affinity antibodies have an affinity of at least about 1 x 107 M"1 to about 1 x 1014 M"1.
8. The method of claim 4 wherein said higher titer of said antibodies is at least about 1.5- 8 fold greater that the titer produced by said parental hybridoma cell.
9. The method ofclaim 1 or 4 further comprising the step of inactivating said dominant negative allele of a mismatch repair gene by knocking out said dominant negative allele or removing an inducer of said dominant negative allele.
10. The method of claim 1 further comprising incubating said parental hybridoma cells with a chemical mutagen.
11. The method of claim 1 wherein the dominant negative mismatch repair gene is introduced into said hybridoma cell after the fusion of said myeloma with said immunoglobulin-producing cells.
12. The method ofclaim 1 wherein said myeloma cells express a dominant negative mismatch repair gene which is also expressed in said hybridoma cells.
13. A hybridoma cell producing high affinity antibodies produced by the method of claim 1, 4, 5, 6, 7, or 10.
14. An antibody produced by a hybridoma cell of claim 13.
15. A method for producing hybridoma cells that produce high titers of antibodies from in vitro immunized immunoglobulin-producing cells comprising:
(a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro;
(b) fusing said immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein said hybridoma cells express a dominant negative allele of a mismatch repair gene;
(c) incubating said parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells; (d) performing a screen of said hypermutated hybridoma cells for antibodies produced in higher titers than that produced by said parental hybridoma cells; and
(e) selecting hypermutated hybridoma cells that produce higher titers of antibodies than that produced by said parental hybridoma cells; thereby producing hybridoma cells that produce high titers of antibodies.
16. The method of claim 15 wherein said dominant negative allele of a mismatch repair gene is selected from the group consisting of a dominant negative allele of PMS2, PMSl, PMSR3, PMSR2, PMSR6, MLHl, GTBP, MSH3, MSH2, MLH3, or MSHl , and homologs of PMSR.
17. The method of claim 15 wherein said dominant negative allele of a mismatch repair gene comprises a dominant negative allele of a PMS2 gene.
18. The method of claim 15 further comprising inactivation of said dominant negative allele of said mismatch repair gene, thereby stabilizing the genome of said hypermutated hybridoma.
19. The method of claim 15 wherein said higher titer of said antibodies is at least about 1.5-8 fold greater that the titer produced by said parental hybridoma cell.
20. The method of claim 18 wherein said dominant negative allele of a mismatch repair gene is inactivated by knocking out said dominant negative allele or removing an inducer of said dominant negative allele.
21. The method of claim 15 further comprising incubating said parental hybridoma cells with a chemical mutagen.
22. The method ofclaim 15 wherein the dominant negative mismatch repair gene is introduced into said hybridoma cell after the fusion of said myeloma with said immunoglobulin-producing cells.
23. The method ofclaim 15 wherein said myeloma cells express a dominant negative mismatch repair gene which is also expressed in said hybridoma cells.
24. A hybridoma cell producing high affinity antibodies produced by the method of claim 15, 18, or 21.
25. An antibody produced by a hybridoma cell of claim 24.
26. A recombinant myeloma cell comprising a polynucleotide sequence encoding a dominant negative mismatch repair protein.
27. The recombinant myeloma cell of claim 26 wherein said mismatch repair protein is selected from the group consisting of PMS2, PMSl, PMSR3, PMSR2, PMSR6, MLHl, GTBP, MSH3, MSH2, MLH3, or MSHl, and homologs of PMSR genes.
28. The recombinant myeloma cell ofclaim 26 wherein said dominant negative allele of a mismatch repair gene comprises a dominant negative allele of a PMS2 gene.
29. The recombinant myeloma cell of claim 26 wherein said myeloma cell does not express immunoglobulin genes.
30. The recombinant myeloma cell of claim 29 wherein said myeloma cell is HAT sensitive.
31. The recombinant myeloma cell of claim 30 wherein said myeloma cell is EBV- negative.
32. A method for producing mammalian expression cells that produce high titers of high- affinity antibodies from in vitro immunized immunoglobulin-producing cells comprising:
(a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro;
(b) fusing said immunoglobulin-producing cells with myeloma cells to form hybridoma cells;
(c) performing a screen for binding of antibodies produced from said hybridoma cells to antigen; (d) cloning immunoglobulin genes from said hybridoma into a mammalian expression cell, wherein said mammalian expression cell expresses a dominant negative allele of a mismatch repair gene;
(e) performing a screen for mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from said hybridoma cells; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized human immunoglobulin-producing cells.
33. The method of claim 32 wherein said dominant negative allele of a mismatch repair gene is introduced into said mammalian expression cell prior to introduction of said immunoglobulin genes.
34. The method of claim 32 wherein said dominant negative allele of a mismatch repair gene is introduced into said mammalian expression cell after introduction of said immunoglobulin genes.
35. The method of claim 32 wherein said dominant negative allele of a mismatch repair gene is introduced into said mammalian expression cell simultaneously said immunoglobulin genes.
36. The method of claim 32 wherein said mismatch repair gene is selected from the group consisting of PMS2, PMSl, PMSR3, PMSR2, PMSR6, MLHl, GTBP, MSH3, MSH2, MLH3, or MSHl, and homologs of PMSR genes.
37. The method of claim 32 wherein said dominant negative allele of a mismatch repair gene comprises a dominant negative allele of a PMS2 gene.
38. The method of claim 32 wherein said high affinity antibodies have an affinity of at least about 1 x 107 M"1 to about 1 x 1014 M"1.
39. A mammalian expression cell produced by the method of claim 32.
40. An antibody produced by a mammalian expression cell of claim 39.
41. A method for producing mammalian expression cells that produce high titers of high- affinity antibodies from in vitro immunized immunoglobulin-producing cells comprising:
(a) combining donor cells comprising i-mmunoglobulin-producing cells with an immunogenic antigen in vitro;
(b) fusing said immunoglobulin-producing cells with myeloma cells to form hybridoma cells, wherein said hybridoma cells express a dominant negative allele of a mismatch repair gene;
(c) incubating said parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells;
(d) performing a screen for binding of antibodies to antigen for antibodies produced from said hypermutated hybridoma cells;
(e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for said antigen than antibodies produced by said parental hybridoma cells;
(f) cloning immunoglobulin genes from said hybridoma into a mammalian expression cell; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulm-producing cells.
42. The method of claim 41 wherein said dominant negative allele of a mismatch repair gene is expressed in said myeloma cell and in said hybridoma cell.
43. The method of claim 41 wherein said dominant negative allele of a mismatch repair gene is introduced into said hybridoma cell after said fusion.
44. The method of claim 41 wherein said mismatch repair gene is selected from the group consisting of PMS2, PMSl, PMSR3, PMSR2, PMSR6, MLHl, GTBP, MSH3, MSH2, MLH3, or MSHl, and homologs of PMSR genes.
45. The method of claim 41 wherein said dominant negative allele of a mismatch repair gene comprises a dominant negative allele of a PMS2 gene.
46. The method of claim 41 wherein said high affinity antibodies have an affinity of at least about 1 x 107 M"1 to about 1 x 1014 M"1.
47. A mammalian expression cell produced by the method of claim 41.
48. An antibody produced by a mammalian expression cell of claim 47.
49. A method for producing mammalian expression cells that produce high titers of high- affinity antibodies from in vitro immunized immunoglobulin-producing cells comprising:
(a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro;
(b) fusing said immunoglobulin-producing cells with myeloma cells to form hybridoma cells;
(c) performing a screen for binding of antibodies produced from said hybridoma cells to antigen;
(d) cloning immunoglobulin genes from said hybridoma into a parental mammalian expression cell, wherein said mammalian expression cell expresses a dominant negative allele of a mismatch repair gene;
(e) incubating said parental mammalian expression cell to allow for mutagenesis, thereby forming hypermutated mammalian expression cells;
(f) performing a screen of hypennutable mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from said hybridoma cells; and
(g) performing a screen of hypermutable mammalian expression cells that secrete higher titers of antibodies than parental mammalian expression cells; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells.
50. The method of claim 49 wherein said dominant negative allele of a mismatch repair gene is introduced into said mammalian expression cell prior to introduction of said immunoglobulin genes.
51. The method of claim 49 wherein said dominant negative allele of a mismatch repair gene is introduced into said mammalian expression cell after introduction of said immunoglobulin genes.
52. The method of claim 49 wherein said dominant negative allele of a mismatch repair gene is introduced into said mammalian expression cell simultaneously said immunoglobulin genes.
53. The method of claim 49 wherein said mismatch repair gene is selected from the group consisting of PMS2, PMSl, PMSR3, PMSR2, PMSR6, MLHl, GTBP, MSH3, MSH2, MLH3, o MSHl, and homologs of PMSR genes.
54. The method of claim 49 wherein said dominant negative allele of a mismatch repair gene comprises a dominant negative allele of a PMS2 gene.
55. The method of claim 49 wherein said high affinity antibodies have an affinity of at least about 1 x 107 M"1 to about 1 x 1014 M"1.
56. The method of claim 49 wherein said higher titer of said antibodies is at least about 1.5-8 fold greater that the titer produced by said parental hybridoma cell.
57. Mammalian expression cells produced by the method of claim 49.
58. Antibodies produced by the mammalian expression cells of claim 57.
59. A recombinant, hypermutable mammalian expression cell comprising a polynucleotide sequence encoding a dominant negative mismatch repair protein.
60. The recombinant, hypermutable mammalian expression cell ofclaim 59 wherein said mismatch repair protein is selected from the group consisting of PMS2, PMSl, PMSR3, PMSR2, PMSR6, MLHl, GTBP, MSH3, MSH2, MLH3, oxMSHl, and homologs of PMSR genes.
61. The recombinant, hypermutable mammalian expression cell of claim 59 wherein said dominant negative allele of a mismatch repair gene comprises a dominant negative allele of a PMS2 gene.
62. A method for producing hybridoma cells producing high-affinity antibodies from in vitro immunized immunoglobulin-producing cells comprising:
(a) combimng donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro;
(b) fusing said immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells;
(c) incubating said parental hybridoma cells in the presence of at least one chemical inhibitor of mismatch repair, thereby forming hypermutated hybridoma cells;
(d) performing a screen for binding of antibodies to antigen for antibodies produced from said hypermutated hybridoma cells; and
(e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for said antigen than antibodies produced by said parental hybridoma cells; thereby producing hybridoma cells producing high-affinity antibodies.
63. The method of claim 62 wherein said chemical inhibitor of mismatch repair is selected from the group consisting of an anthracene, ATPase inhibitor, a nuclease inhibitor, an RNA interference molecule, a polymerase inhibitor and an antisense oligonucleotide that specifically hybridizes to a nucleotide encoding a mismatch repair protein.
64. The method of claim 62 wherein said inhibitor is an anthracene having the formula:
Figure imgf000072_0001
wherein Ri-Rio are independently hydrogen, hydroxyl, amino, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S- alkenyl, N-alkenyl, O-alkynyl, S-alkynyl, N-alkynyl, aryl, substituted aryl, aryloxy, substituted aryloxy, heteroaryl, substituted heteroaryl, aralkyloxy, arylalkyl, alkylaryl, alkylaryloxy, arylsulfonyl, alkylsulfonyl, alkoxycarbonyl, aryloxycarbonyl, guanidino, carboxy, an alcohol, an amino acid, sulfonate, alkyl sulfonate, CN, NO2, an aldehyde group, an ester, an ether, a crown ether, a ketone, an organosulfur compound, an organometallic group, a carboxylic acid, an organosilicon or a carbohydrate that optionally contains one or more alkylated hydroxyl groups; wherein said heteroalkyl, heteroaryl, and substituted heteroaryl contain at least one heteroatom that is oxygen, sulfur, a metal atom, phosphorus, silicon or nitrogen; and wherein said substituents of said substituted alkyl, substituted alkenyl, substituted alkynyl, substituted aryl, and substituted heteroaryl are halogen, CN, NO2, lower alkyl, aryl, heteroaryl, aralkyl, aralkoxy, guanidino, alkoxycarbonyl, alkoxy, hydroxy, carboxy and amino; and wherein said amino groups are optionally substituted with an acyl group, or 1 to 3 aryl or lower alkyl groups.
65. The method ofclaim 64 wherein Ri-Rio are independently hydrogen, hydroxyl, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, phenyl, tolyl, hydroxymethyl, hydroxypropyl, or hydroxybutyl.
66. The method of claim 62 further comprising a screen for hypermutated hybridomas that also produce antibodies in higher titers than said parental hybridomas.
67. The method of claim 62 further comprising the step of removing said chemical inhibitor from said growth medium following hypermutation, thereby stabilizing the genome of said hypermutated hybridoma.
68. The method ofclaim 66 further comprising the step of removing said chemical inhibitor from said growth medium following hypermutation, thereby stabilizing the genome of said hypermutated hybridoma.
69. The method of claim 62 wherein said high affinity antibodies have an affinity of at least about 1 x 107 M"1 to about 1 x 1014 M"1.
70. The method of claim 66 wherein said higher titer of said antibodies is at least about 1.5-8 fold greater that the titer produced by said parental hybridoma cell.
71. A hybridoma cell produced by the method of claim 62, 66, 67, or 68.
72. An antibody produced by a hybridoma cell of claim 71.
73. A method for producing hybridoma cells that produce high titers of antibodies from in vitro immunized immunoglobulin-producing cells comprising:
(a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro;
(b) fusing said immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells;
(c) incubating said parental hybridoma cells in the presence of at least one chemical inhibitor of mismatch repair, thereby forming hypermutated hybridoma cells;
(d) performing a screen of said hypermutated hybridoma cells for antigen-specific antibodies produced in higher titers than that produced by said parental hybridoma cells; and
(e) selecting hypermutated hybridoma cells that produce higher titers of antibodies than that produced by said parental hybridoma cells; thereby producing hybridoma cells that produce high titers of antibodies.
74. The method ofclaim 73 wherein said chemical inhibitor is selected from the group consisting of an anthracene, ATPase inhibitor, a nuclease inhibitor, an RNA interference molecule, a polymerase inhibitor and an antisense oligonucleotide that specifically hybridizes to a nucleotide encoding a mismatch repair protein.
75. The method of claim 74 wherein said inhibitor is an anthracene having the formula:
Figure imgf000074_0001
wherein Ri-Rio are independently hydrogen, hydroxyl, amino, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S- alkenyl, N-alkenyl, O-alkynyl, S-alkynyl, N-alkynyl, aryl, substituted aryl, aryloxy, substituted aryloxy, heteroaryl, substituted heteroaryl, aralkyloxy, arylalkyl, alkylaryl, alkylaryloxy, arylsulfonyl, alkylsulfonyl, alkoxycarbonyl, aryloxycarbonyl, guanidino, carboxy, an alcohol, an amino acid, sulfonate, alkyl sulfonate, CN, NO2, an aldehyde group, an ester, an ether, a crown ether, a ketone, an organosulfur compound, an organometallic group, a carboxylic acid, an organosilicon or a carbohydrate that optionally contains one or more alkylated hydroxyl groups; wherein said heteroalkyl, heteroaryl, and substituted heteroaryl contain at least one heteroatom that is oxygen, sulfur, a metal atom, phosphorus, silicon or nitrogen; and wherein said substituents of said substituted alkyl, substituted alkenyl, substituted alkynyl, substituted aryl, and substituted heteroaryl are halogen, CN, NO , lower alkyl, aryl, heteroaryl, aralkyl, aralkoxy, guanidino, alkoxycarbonyl, alkoxy, hydroxy, carboxy and amino; and wherein said amino groups are optionally substituted with an acyl group, or 1 to 3 aryl or lower alkyl groups.
76. The method of claim 75 wherein Ri-Rio are independently hydrogen, hydroxyl, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, phenyl, tolyl, hydroxymethyl, hydroxypropyl, or hydroxybutyl.
77. The method of claim 73 further comprising the step of removing said chemical inhibitor from said growth medium following hypermutation, thereby stabilizing the genome of said hypermutated hybridoma.
78. The method of claim 73 wherein said higher titer of said antibodies is at least about 1.5-8 fold greater that the titer produced by said parental hybridoma cell.
79. A hybridoma cell produced by the method of claim 73 or 77.
80. An antibody produced by a hybridoma of claim 79.
81. A method for producing mammalian expression cells that produce high titers of high- affinity antibodies from in vitro immunized immunoglobulin-producing cells comprising:
(a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro;
(b) fusing said immunoglobulin-producing cells with myeloma cells to form hybridoma cells;
(c) performing a screen for binding of antibodies produced from said hybridoma cells to antigen;
(d) cloning immunoglobulin genes from said hybridoma into a mammalian expression cell; (e) incubating said mammalian expression cell in the presence of at least one chemical inhibitor of mismatch repair;
(f) performing a screen for mammalian expression cells that secrete antibodies with higher affinity for antigen as compared to antibodies produced from said hybridoma cells; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells.
82. The method ofclaim 81 wherein said chemical inhibitor of mismatch repair is selected from the group consisting of an anthracene, ATPase inhibitor, a nuclease inhibitor, an RNA interference molecule, a polymerase inhibitor and an antisense oligonucleotide that specifically hybridizes to a nucleotide encoding a mismatch repair protein.
83. The method ofclaim 82 wherein said inhibitor is an anthracene having the formula:
Figure imgf000076_0001
wherein R1-R10 are independently hydrogen, hydroxyl, amino, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S- alkenyl, N-alkenyl, O-alkynyl, S-alkynyl, N-alkynyl, aryl, substituted aryl, aryloxy, substituted aryloxy, heteroaryl, substituted heteroaryl, aralkyloxy, arylalkyl, alkylaryl, alkylaryloxy, arylsulfonyl, alkylsulfonyl, alkoxycarbonyl, aryloxycarbonyl, guanidino, carboxy, an alcohol, an amino acid, sulfonate, alkyl sulfonate, CN, NO , an aldehyde group, an ester, an ether, a crown ether, a ketone, an organosulfur compound, an organometallic group, a carboxylic acid, an organosilicon or a carbohydrate that optionally contains one or more alkylated hydroxyl groups; wherein said heteroalkyl, heteroaryl, and substituted heteroaryl contain at least one heteroatom that is oxygen, sulfur, a metal atom, phosphorus, silicon or nitrogen; and wherein said substituents of said substituted alkyl, substituted alkenyl, substituted alkynyl, substituted aryl, and substituted heteroaryl are halogen, CN, NO2, lower alkyl, aryl, heteroaryl, aralkyl, aralkoxy, guanidino, alkoxycarbonyl, alkoxy, hydroxy, carboxy and amino; and wherein said amino groups are optionally substituted with an acyl group, or 1 to 3 aryl or lower alkyl groups.
84. The method of claim 83 wherein Ri-Ri o are independently hydrogen, hydroxyl, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, phenyl, tolyl, hydroxymethyl, hydroxypropyl, or hydroxyburyl.
85. The method of claim 81 further comprising screen, prior to collection of said antibodies from said hypermutated hybridoma cells, for hypermutated hybridomas that also produce antibodies in higher titers than said parental hybridomas.
86. The method of claim 81 wherein said high affinity antibodies have an affinity of at least about 1 x 107 M"1 to about 1 x 1014 M"1.
87. The method of claim 81 wherein said higher titer of said antibodies is at least about 1.5-8 fold greater that the titer produced by said parental hybridoma cell.
88. The method ofclaim 81 further comprising removing said chemical inhibitor, thereby stabilizing the genome of said hypermutated mammalian expression cells.
89. A mammalian expression cell produced by the method ofclaim 81, 85, or 88.
90. An antibody produced by a mammalian expression cell ofclaim 89.
91. A method for producing mammalian expression cells that produce high titers of high affinity antibodies to a selected antigen from in vitro immunized immunoglobulin-producing cells comprising:
(a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro;
(b) fusing said immunoglobulin-producing cells with myeloma cells to form hybridoma cells;
(c) incubating said hybridoma cells in the presence of at least one chemical inhibitor of mismatch repair to form hypermutated hybridoma cells;
(d) performing a screen for binding of antigen for antibodies produced from said hypermutated hybridoma cells; (e) selecting hypermutated hybridoma cells that produce antibodies with greater affinity for said antigen than antibodies produced by said parental hybridoma cells;
(f) cloning immunoglobulin genes from said hypermutated hybridoma cells into a mammalian expression cell, thereby forming parental mammalian expression cells; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells.
92. The method ofclaim 91 wherein said chemical inhibitor of mismatch repair is selected from the group consisting of an anthracene, ATPase inhibitor, a nuclease inhibitor, an RNA interference molecule, a polymerase inhibitor and an antisense oligonucleotide that specifically hybridizes to a nucleotide encoding a mismatch repair protein.
93. The method of claim 91 wherein said inhibitor is an anthracene having the formula:
Figure imgf000078_0001
wherein Ri-Rio are independently hydrogen, hydroxyl, amino, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S- alkenyl, N-alkenyl, O-alkynyl, S-alkynyl, N-alkynyl, aryl, substituted aryl, aryloxy, substituted aryloxy, heteroaryl, substituted heteroaryl, aralkyloxy, arylalkyl, alkylaryl, alkylaryloxy. arylsulfonyl, alkylsulfonyl, alkoxycarbonyl, aryloxycarbonyl, guanidino, carboxy, an alcohol, an amino acid, sulfonate, alkyl sulfonate, CN, NO2, an aldehyde group, an ester, an ether, a crown ether, a ketone, an organosulfur compound, an organometallic group, a carboxylic acid, an organosilicon or a carbohydrate that optionally contains one or more alkylated hydroxyl groups; wherein said heteroalkyl, heteroaryl, and substituted heteroaryl contain at least one heteroatom that is oxygen, sulfur, a metal atom, phosphorus, silicon or nitrogen; and wherein said substituents of said substituted alkyl, substituted alkenyl, substituted alkynyl, substituted aryl, and substituted heteroaryl are halogen, CN, NO2, lower alkyl, aryl, heteroaryl, aralkyl, aralkoxy, guanidino, alkoxycarbonyl, alkoxy, hydroxy, carboxy and amino; and wherem said amino groups are optionally substituted with an acyl group, or 1 to 3 aryl or lower alkyl groups.
94. The method of claim 92 wherein R io are independently hydrogen, hydroxyl, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, phenyl, tolyl, hydroxymethyl, hydroxypropyl, or hydroxybutyl.
95. The method ofclaim 91 wherein said high affinity antibodies have an affinity of at least about 1 x 107 M"1 to about 1 x 1014 M"1.
96. The method ofclaim 91 further comprising the steps of: incubating said mammalian expression cell in the presence of at least one chemical inhibitor of mismatch repair, thereby forming a hypermutated mammalian expression cell; and screening for hypermutated mammalian expression cells that produce a higher titer of antibodies that said parental mammalian expression cells.
97. The method ofclaim 91 further comprising removing said chemical inhibitor, thereby stabilizing the genome of said hypermutated hybridoma cells.
98. The method of claim 96 further comprising removing said chemical inhibitor, thereby stabilizing the genome of said hypermutated mammalian expression cells.
99. The method ofclaim 96 wherein said higher titer of said antibodies is at least about 1.5-8 fold greater that the titer produced by said parental hybridoma cell.
100. A mammalian expression cell produced by the method ofclaim 91, 95, 96, or 97.
101. An antibody produced by a mammalian expression cell of claim 100.
102. A method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin-producing cells in high titers comprising:
(a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro, wherein said donor cells are derived from a donor that is naturally deficient in mismatch repair; (b) fusing said immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein said parental hybridoma cells are deficient in mismatch repair;
(c) incubating the said parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells;
(d) performing a screen for binding of antibodies to antigen for antibodies produced from said hypermutated hybridoma cells;
(e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by said parental hybridoma cells;
(f) performing a second screen for hypermutated hybridoma cells that produce increased titers of antibodies as compared with said parental hybridoma cells;
(g) selecting hypermutated hybridoma cells that produce antibodies in higher titers than produced by said parental hybridoma cells; thereby producing hybridoma cells producing high titers of high-affinity antibodies.
103. The method ofclaim 102 further comprising the step of introducing a wild-type gene for mismatch repair into said selected hypennutated hybridoma cell to complement the mismatch repair deficiency, thereby restabilizing the genome of said selected hypermutated hybridoma cell.
104. The method of claim 102 further comprising incubating said parental hybridoma cells with a chemical mutagen.
105. A hybridoma cell produced by the method of claim 102, 103 , or 104.
106. An antibody produced by a hybridoma cell of claim 105.
107. A method for producing hybridoma cells that produce high-affinity antibodies from in vitro immunized immunoglobulin-producing cells in high titers comprising:
(a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro;
(b) fusing said immunoglobulin-producing cells with myeloma cells, wherein the myeloma cells are naturally deficient in mismatch repair, thereby forming parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair; (c) incubating said parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells;
(d) performing a screen for binding of antibodies to antigen for antibodies produced from said hypermutated hybridoma cells;
(e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by said parental hybridoma cells;
(f) performing a second screen for hypermutated hybridoma cells that produce increased titers of antibodies as compared with parental hybridoma cells;
(g) selecting hypermutated hybridoma cells that produce antibodies in higher titers than produced by said parental hybridoma cells; thereby producing hybridoma cells producing high titers of high-affinity antibodies.
108. The method ofclaim 107 further comprising introducing a wild-type gene for mismatch repair into said selected hypermutated hybridoma cell to complement the mismatch repair deficiency, thereby restabilizing the genome of said selected hypermutated hybridoma cell.
109. The method ofclaim 107 further comprising incubating said parental hybridoma cells with a chemical mutagen. t
110. The method of claim 107 wherein said high affinity antibodies have an affinity of at least about 1 x 107 M"1 to about 1 x 1014 M"1.
111. The method of claim 107 wherein said higher titer of said antibodies is at least about 1.5-8 fold greater that the titer produced by said parental hybridoma cell.
112. A hybridoma cell produced by the method ofclaim 107, 108, or 109.
113. An antibody produced by a hybridoma cell of claim 112.
114. A method for producing mammalian expression cells that produce high-affinity antibodies in high titers from in vitro immunized immunoglobulin-producing cells comprising: (a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro, wherein the donor cells are derived from a donor that is naturally deficient in mismatch repair;
(b) fusing the immunoglobulin-producing cells with myeloma cells to form parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair;
(c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells;
(d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells;
(e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells;
(f) cloning immunoglobulin genes from said hypermutated hybridoma into a mammalian expression cell; thereby producing a mammalian expression cell that produce high titers of high-affinity antibodies in high titer from in vitro immunized immunoglobulin-producing cells.
115. The method of claim 114 wherein said high affinity antibodies have an affinity of at least about 1 x 107 M"1 to about 1 x 1014 M"1.
116. The method ofclaim 114 wherein said higher titer of said antibodies is at least about 1.5-8 fold greater that the titer produced by said parental hybridoma cell.
117. The method of claim 114 further comprising the steps of: incubating the mammalian expression cells in the presence of at least one chemical inhibitor of mismatch repair, thereby forming hypermutated mammalian expression cells; and screening said hypermutated mammalian expression cells for higher production of antibodies than that of the parental mammalian expression cells.
118. The method of claim 117 wherein said chemical inhibitor of mismatch repair is selected from the group consisting of an anthracene, ATPase inhibitor, a nuclease inhibitor, an RNA interference molecule, a polymerase inhibitor and an antisense oligonucleotide that specifically hybridizes to a nucleotide encoding a mismatch repair protein.
119. The method of claim 117 wherein said inhibitor is an anthracene having the formula:
Figure imgf000083_0001
wherein Ri-Rio are independently hydrogen, hydroxyl, amino, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S- alkenyl, N-alkenyl, O-alkynyl, S-alkynyl, N-alkynyl, aryl, substituted aryl, aryloxy, substituted aryloxy, heteroaryl, substituted heteroaryl, aralkyloxy, arylalkyl, alkylaryl, alkylaryloxy, arylsulfonyl, alkylsulfonyl, alkoxycarbonyl, aryloxycarbonyl, guanidino, carboxy, an alcohol, an amino acid, sulfonate, alkyl sulfonate, CN, NO2, an aldehyde group, an ester, an ether, a crown ether, a ketone, an organosulfur compound, an organometallic group, a carboxylic acid, an organosilicon or a carbohydrate that optionally contains one or more alkylated hydroxyl groups; wherein said heteroalkyl, heteroaryl, and substituted heteroaryl contain at least one heteroatom that is oxygen, sulfiir, a metal atom, phosphorus, silicon or nitrogen; and wherein said substituents of said substituted alkyl, substituted alkenyl, substituted alkynyl, substituted aryl, and substituted heteroaryl are halogen, CN, NO2, lower alkyl, aryl, heteroaryl, aralkyl, aralkoxy, guanidino, alkoxycarbonyl, alkoxy, hydroxy, carboxy and amino; and wherein said amino groups are optionally substituted with an acyl group, or 1 to 3 aryl or lower alkyl groups.
120. The method of claim 119 wherein Ri -Ri o are independently hydrogen, hydroxyl, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, phenyl, tolyl, hydroxymethyl, hydroxypropyl, or hydroxybutyl.
121. The method of claim 117 further comprising the step of removing said chemical inhibitor of mismatch repair from the hypermutated mammalian expression cells, thereby stabilizing the genome of said hypermutated mammalian expression cells.
122. A mammalian expression cell produced by the method of claim 114, 117, 118, or 121.
123. An antibody produced by a mammalian expression cell of claim 122.
124. A method for producing mammalian expression cells that produce high-affinity antibodies in high titer from in vitro immunized immunoglobulin-producing cells comprising:
(a) combining donor cells comprising immunoglobulin-producing cells with an immunogenic antigen in vitro;
(b) fusing the immunoglobulin-producing cells with myeloma cells, wherein the myeloma cells are naturally deficient in mismatch repair, thereby forming parental hybridoma cells, wherein the hybridoma cells are deficient in mismatch repair;
(c) incubating the parental hybridoma cells to allow for mutagenesis, thereby forming hypermutated hybridoma cells;
(d) performing a screen for binding of antibodies to antigen for antibodies produced from the hypermutated hybridoma cells;
(e) selecting hypermutated hybridoma cells that produce antibodies with enhanced affinity for the antigen than antibodies produced by the parental hybridoma cells; and
(f) cloning immunoglobulin genes from said hypermutated hybridoma cell into a mammalian expression cell; thereby producing mammalian expression cells that produce high titers of high-affinity antibodies from in vitro immunized immunoglobulin-producing cells.
125. The method ofclaim 124 wherein said high affinity antibodies have an affinity of at least about 1 x 107 M"1 to about 1 x 1014 M"1.
126. The method of claim 124 wherein said higher titer of said antibodies is at least about 1.5-8 fold greater that the titer produced by said parental hybridoma cell.
127. The method ofclaim 124 further comprising the steps of: incubating the mammalian expression cells in the presence of at least one chemical inhibitor of mismatch repair, thereby forming hypermutated mammalian expression cells; and screening said hypermutated mammalian expression cells for higher production of antibodies than that of the parental mammalian expression cells.
128. The method of claim 127 wherein said chemical inhibitor of mismatch repair is selected from the group consisting of an anthracene, ATPase inhibitor, a nuclease inhibitor, an RNA interference molecule, a polymerase inhibitor and an antisense oligonucleotide that specifically hybridizes to a nucleotide encoding a mismatch repair protein.
129. The method of claim 127 wherein said inhibitor is an anthracene having the formula:
Figure imgf000085_0001
wherein Ri-Rio are independently hydrogen, hydroxyl, amino, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S- alkenyl, N-alkenyl, O-alkynyl, S-alkynyl, N-alkynyl, aryl, substituted aryl, aryloxy, substituted aryloxy, heteroaryl, substituted heteroaryl, aralkyloxy, arylalkyl, alkylaryl, alkylaryloxy, arylsulfonyl, alkylsulfonyl, alkoxycarbonyl, aryloxycarbonyl, guanidino, carboxy, an alcohol, an amino acid, sulfonate, alkyl sulfonate, CN, NO , an aldehyde group, an ester, an ether, a crown ether, a ketone, an organosulfur compound, an organometallic group, a carboxylic acid, an organosilicon or a carbohydrate that optionally contains one or more alkylated hydroxyl groups; wherein said heteroalkyl, heteroaryl, and substituted heteroaryl contain at least one heteroatom that is oxygen, sulfur, a metal atom, phosphorus, silicon or nitrogen; and wherein said substituents of said substituted alkyl, substituted alkenyl, substituted alkynyl, substituted aryl, and substituted heteroaryl are halogen, CN, NO2, lower alkyl, aryl, heteroaryl, aralkyl, aralkoxy, guanidino, alkoxycarbonyl, alkoxy, hydroxy, carboxy and amino; and wherein said amino groups are optionally substituted with an acyl group, or 1 to 3 aryl or lower alkyl groups.
130. The method of claim 129 wherein Ri-Rio are independently hydrogen, hydroxyl, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, phenyl, tolyl, hydroxymethyl, hydroxypropyl, or hydroxybutyl.
131. The method of claim 127 further comprising the step of removing said chemical inhibitor of mismatch repair from the hypermutated mammalian expression cells, thereby stabilizing the genome of said hypermutated mammalian expression cells.
132. A mammalian expression cell produced by the method ofclaim 124, 127, or 131.
133. An antibody produced by a mammalian expression cell of claim 132.
134. A method for in vitro production of antigen-specific immunoglobulin-producing cells comprising:
(a) isolating donor cells from an animal;
(b) treating said cells with L-leucyl-L-leucine methy ester hydrobromide;
(c) incubating said donor cells with an immunogenic antigen in vitro, at 25-37°C, 5- 10%) CO2, in medium supplemented with 5-15% serum, and a growth promoting cytokine for 4 days;
(d) washing said cells in medium; and
(e) culturing said cells in medium supplemented with 5-15% serum an additional 8 days; thereby stimulating the production of antigen-specific immunoglobulin-producing cells.
135. The method of claim 62, 73, 81, 91, 117, or 127 wherein said chemical inhibitor of mismatch repair is an antisense molecule comprising at least 15 consecutive nucleotides of a sequence encoding a protein selected from the group consisting of SEQ ID NO:2; SEQ ID NO:4; SEQ JD NO:6; SEQ JD NO:8; SEQ ID NO: 10; SEQ JD NO: 12; SEQ JD NO: 14; SEQ JD NO: 16; SEQ ID NO: 18; SEQ ID NO:20; SEQ ID NO:22; SEQ JD NO:24; SEQ J_D NO:26; SEQ JD NO:28; SEQ LD NO:30; SEQ ID NO:32; SEQ ID NO:34; SEQ JD NO:36; SEQ ID NO:38; SEQ ID NO:40; SEQ TD NO:42; SEQ JD NO:44; SEQ JD NO:46; SEQ JD NO:48; and SEQ JD NO:50.
136. The method ofclaim 62, 73, 81, 91, 117, or 127 wherein said chemical inhibitor of mismatch repair is an antisense molecule comprising at least 15 consecutive nucleotides of a sequence selected from the group consisting of SEQ ID NO:l; SEQ JD NO:3; SEQ JD NO:5; SEQ ID NO:7; SEQ JD NO:9; SEQ ID NO:ll; SEQ ID NO:13; SEQ JD NO:15; SEQ ID NO:17; SEQ ID NO:19; SEQ JD NO:21; SEQ ID NO:23; SEQ ID NO:25; SEQ JD NO:27; SEQ ID NO:29; SEQ JD NO:31; SEQ JD NO:33; SEQ JD NO:35; SEQ ID NO:37; SEQ ID NO:39; SEQ JD NO:41; SEQ JD NO:43; SEQ JD NO:45; SEQ JD NO:47; and SEQ TD NO:49.
137. A hybridoma cell producing high affinity antibodies produced by the method of claim 9.
138. An antibody produced by a hybridoma cell of claim 137.
PCT/US2003/036702 2002-11-15 2003-11-14 Methods of generating high-production of antibodies from hybridomas created by in vitro immunization WO2004046330A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
JP2004570421A JP4555089B2 (en) 2002-11-15 2003-11-14 Method for producing high production amount of antibody from hybridoma created by in vitro immunization
DE60329526T DE60329526D1 (en) 2002-11-15 2003-11-14 METHOD OF GENERATING HIGH ANTIBODY PRODUCTION OF HYBRIDOMAS RESULTING FROM IN VITRO IMMUNIZATION
AU2003295576A AU2003295576B2 (en) 2002-11-15 2003-11-14 Methods of generating high-production of antibodies from hybridomas created by in vitro immunization
AT03786774T ATE444359T1 (en) 2002-11-15 2003-11-14 METHOD FOR GENERATING HIGH ANTIBODIES PRODUCTION FROM HYBRIDOMAS CREATED BY IN VITRO IMMUNIZATION
CA2506127A CA2506127C (en) 2002-11-15 2003-11-14 Methods of generating high-production of antibodies from hybridomas created by in vitro immunization
EP20030786774 EP1572971B1 (en) 2002-11-15 2003-11-14 Methods of generating high-production of antibodies from hybridomas created by in vitro immunization

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US42716502P 2002-11-15 2002-11-15
US60/427,165 2002-11-15
US50165003P 2003-09-10 2003-09-10
US60/501,650 2003-09-10

Publications (2)

Publication Number Publication Date
WO2004046330A2 true WO2004046330A2 (en) 2004-06-03
WO2004046330A3 WO2004046330A3 (en) 2006-09-28

Family

ID=32329149

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/036702 WO2004046330A2 (en) 2002-11-15 2003-11-14 Methods of generating high-production of antibodies from hybridomas created by in vitro immunization

Country Status (8)

Country Link
US (2) US7754450B2 (en)
EP (1) EP1572971B1 (en)
JP (1) JP4555089B2 (en)
AT (1) ATE444359T1 (en)
AU (1) AU2003295576B2 (en)
CA (1) CA2506127C (en)
DE (1) DE60329526D1 (en)
WO (1) WO2004046330A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007092939A3 (en) * 2006-02-08 2008-03-27 Morphotek Inc Antigenic gm-csf peptides and antibodies to gm-csf
EP2221384A1 (en) * 2007-12-03 2010-08-25 Kabushiki Kaisya Advance Method for production of antibody
EP2289937A1 (en) * 2009-08-24 2011-03-02 Wilfried Stücker Production of monoclonal antibodies in vitro
WO2014036404A3 (en) * 2012-08-31 2015-07-30 Synthetic Genomics, Inc. Crowding agent-induced nucleic acid transfer into a recipient host cell
AU2013201228B2 (en) * 2006-02-08 2016-01-21 Eisai, Inc. Antigenic gm-csf peptides and antibodies to gm-csf
US9751939B2 (en) 2005-05-18 2017-09-05 Morphosys Ag Anti-GM-CSF antibodies
CN110776564A (en) * 2019-10-30 2020-02-11 西北农林科技大学 Two-strain anti-newcastle disease virus nano antibody and expression preparation method and application thereof

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6982169B2 (en) * 2001-01-15 2006-01-03 Morphotek, Inc. Chemical inhibitors of mismatch repair
ATE444359T1 (en) 2002-11-15 2009-10-15 Morphotek Inc METHOD FOR GENERATING HIGH ANTIBODIES PRODUCTION FROM HYBRIDOMAS CREATED BY IN VITRO IMMUNIZATION
WO2007121396A2 (en) * 2006-04-17 2007-10-25 Morphotek, Inc. Whole genome evolution technology applied to improve protein and antibody yields by cells
WO2008103474A1 (en) 2007-02-20 2008-08-28 Anaptysbio, Inc. Methods of generating libraries and uses thereof
CN103168119B (en) * 2010-06-16 2016-05-11 Cdi实验室公司 For generation of, confirm and use the method and system of monoclonal antibody
US10260089B2 (en) 2012-10-29 2019-04-16 The Research Foundation Of The State University Of New York Compositions and methods for recognition of RNA using triple helical peptide nucleic acids
EP2970470B1 (en) * 2013-03-15 2019-12-04 Amgen Inc. Human pac1 antibodies
US20140273031A1 (en) * 2013-03-15 2014-09-18 The Cleveland Clinic Foundation In-vitro method for monoclonal antibody production

Family Cites Families (165)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US4376110A (en) 1980-08-04 1983-03-08 Hybritech, Incorporated Immunometric assays using monoclonal antibodies
US4469863A (en) 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US5023243A (en) 1981-10-23 1991-06-11 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and method of making same
US4476301A (en) 1982-04-29 1984-10-09 Centre National De La Recherche Scientifique Oligonucleotides, a process for preparing the same and their application as mediators of the action of interferon
JPS5927900A (en) 1982-08-09 1984-02-14 Wakunaga Seiyaku Kk Oligonucleotide derivative and its preparation
FR2540122B1 (en) 1983-01-27 1985-11-29 Centre Nat Rech Scient NOVEL COMPOUNDS COMPRISING A SEQUENCE OF OLIGONUCLEOTIDE LINKED TO AN INTERCALATION AGENT, THEIR SYNTHESIS PROCESS AND THEIR APPLICATION
US4605735A (en) 1983-02-14 1986-08-12 Wakunaga Seiyaku Kabushiki Kaisha Oligonucleotide derivatives
US4948882A (en) 1983-02-22 1990-08-14 Syngene, Inc. Single-stranded labelled oligonucleotides, reactive monomers and methods of synthesis
US4824941A (en) 1983-03-10 1989-04-25 Julian Gordon Specific antibody to the native form of 2'5'-oligonucleotides, the method of preparation and the use as reagents in immunoassays or for binding 2'5'-oligonucleotides in biological systems
US4587044A (en) 1983-09-01 1986-05-06 The Johns Hopkins University Linkage of proteins to nucleic acids
US5118800A (en) 1983-12-20 1992-06-02 California Institute Of Technology Oligonucleotides possessing a primary amino group in the terminal nucleotide
US5118802A (en) 1983-12-20 1992-06-02 California Institute Of Technology DNA-reporter conjugates linked via the 2' or 5'-primary amino group of the 5'-terminal nucleoside
US5550111A (en) 1984-07-11 1996-08-27 Temple University-Of The Commonwealth System Of Higher Education Dual action 2',5'-oligoadenylate antiviral derivatives and uses thereof
FR2567892B1 (en) 1984-07-19 1989-02-17 Centre Nat Rech Scient NOVEL OLIGONUCLEOTIDES, THEIR PREPARATION PROCESS AND THEIR APPLICATIONS AS MEDIATORS IN DEVELOPING THE EFFECTS OF INTERFERONS
US5367066A (en) 1984-10-16 1994-11-22 Chiron Corporation Oligonucleotides with selectably cleavable and/or abasic sites
US5430136A (en) 1984-10-16 1995-07-04 Chiron Corporation Oligonucleotides having selectably cleavable and/or abasic sites
US5258506A (en) 1984-10-16 1993-11-02 Chiron Corporation Photolabile reagents for incorporation into oligonucleotide chains
US4828979A (en) 1984-11-08 1989-05-09 Life Technologies, Inc. Nucleotide analogs for nucleic acid labeling and detection
FR2575751B1 (en) 1985-01-08 1987-04-03 Pasteur Institut NOVEL ADENOSINE DERIVATIVE NUCLEOSIDES, THEIR PREPARATION AND THEIR BIOLOGICAL APPLICATIONS
US4720459A (en) 1985-02-14 1988-01-19 Medical College Of Wisconsin Research Foundation, Inc. Myelomas for producing human/human hybridomas
US5405938A (en) 1989-12-20 1995-04-11 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5185444A (en) 1985-03-15 1993-02-09 Anti-Gene Deveopment Group Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US5166315A (en) 1989-12-20 1992-11-24 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5235033A (en) 1985-03-15 1993-08-10 Anti-Gene Development Group Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US4762779A (en) 1985-06-13 1988-08-09 Amgen Inc. Compositions and methods for functionalizing nucleic acids
US5317098A (en) 1986-03-17 1994-05-31 Hiroaki Shizuya Non-radioisotope tagging of fragments
JPS638396A (en) 1986-06-30 1988-01-14 Wakunaga Pharmaceut Co Ltd Poly-labeled oligonucleotide derivative
DE3788914T2 (en) 1986-09-08 1994-08-25 Ajinomoto Kk Compounds for cleaving RNA at a specific position, oligomers used in the preparation of these compounds and starting materials for the synthesis of these oligomers.
US5264423A (en) 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5276019A (en) 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US4904582A (en) 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
CA1340032C (en) 1987-06-24 1998-09-08 Jim Haralambidis Lucleoside derivatives
US5585481A (en) 1987-09-21 1996-12-17 Gen-Probe Incorporated Linking reagents for nucleotide probes
US4924624A (en) 1987-10-22 1990-05-15 Temple University-Of The Commonwealth System Of Higher Education 2,',5'-phosphorothioate oligoadenylates and plant antiviral uses thereof
US5188897A (en) 1987-10-22 1993-02-23 Temple University Of The Commonwealth System Of Higher Education Encapsulated 2',5'-phosphorothioate oligoadenylates
US5525465A (en) 1987-10-28 1996-06-11 Howard Florey Institute Of Experimental Physiology And Medicine Oligonucleotide-polyamide conjugates and methods of production and applications of the same
DE3738460A1 (en) 1987-11-12 1989-05-24 Max Planck Gesellschaft MODIFIED OLIGONUCLEOTIDS
EP0348458B1 (en) 1987-11-30 1997-04-09 University Of Iowa Research Foundation Dna molecules stabilized by modifications of the 3'-terminal phosphodiester linkage and their use as nucleic acid probes and as therapeutic agents to block the expression of specifically targeted genes
US5403711A (en) 1987-11-30 1995-04-04 University Of Iowa Research Foundation Nucleic acid hybridization and amplification method for detection of specific sequences in which a complementary labeled nucleic acid probe is cleaved
US5082830A (en) 1988-02-26 1992-01-21 Enzo Biochem, Inc. End labeled nucleotide probe
EP0406309A4 (en) 1988-03-25 1992-08-19 The University Of Virginia Alumni Patents Foundation Oligonucleotide n-alkylphosphoramidates
US5278302A (en) 1988-05-26 1994-01-11 University Patents, Inc. Polynucleotide phosphorodithioates
US5109124A (en) 1988-06-01 1992-04-28 Biogen, Inc. Nucleic acid probe linked to a label having a terminal cysteine
US5216141A (en) 1988-06-06 1993-06-01 Benner Steven A Oligonucleotide analogs containing sulfur linkages
US5175273A (en) 1988-07-01 1992-12-29 Genentech, Inc. Nucleic acid intercalating agents
US5262536A (en) 1988-09-15 1993-11-16 E. I. Du Pont De Nemours And Company Reagents for the preparation of 5'-tagged oligonucleotides
US5194599A (en) 1988-09-23 1993-03-16 Gilead Sciences, Inc. Hydrogen phosphonodithioate compositions
US5512439A (en) 1988-11-21 1996-04-30 Dynal As Oligonucleotide-linked magnetic particles and uses thereof
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5599923A (en) 1989-03-06 1997-02-04 Board Of Regents, University Of Tx Texaphyrin metal complexes having improved functionalization
US5457183A (en) 1989-03-06 1995-10-10 Board Of Regents, The University Of Texas System Hydroxylated texaphyrins
US5391723A (en) 1989-05-31 1995-02-21 Neorx Corporation Oligonucleotide conjugates
US5256775A (en) 1989-06-05 1993-10-26 Gilead Sciences, Inc. Exonuclease-resistant oligonucleotides
US4958013A (en) 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides
US5451463A (en) 1989-08-28 1995-09-19 Clontech Laboratories, Inc. Non-nucleoside 1,3-diol reagents for labeling synthetic oligonucleotides
US5134066A (en) 1989-08-29 1992-07-28 Monsanto Company Improved probes using nucleosides containing 3-dezauracil analogs
US5254469A (en) 1989-09-12 1993-10-19 Eastman Kodak Company Oligonucleotide-enzyme conjugate that can be used as a probe in hybridization assays and polymerase chain reaction procedures
US5591722A (en) 1989-09-15 1997-01-07 Southern Research Institute 2'-deoxy-4'-thioribonucleosides and their antiviral activity
US5399676A (en) 1989-10-23 1995-03-21 Gilead Sciences Oligonucleotides with inverted polarity
US5721218A (en) 1989-10-23 1998-02-24 Gilead Sciences, Inc. Oligonucleotides with inverted polarity
US5264562A (en) 1989-10-24 1993-11-23 Gilead Sciences, Inc. Oligonucleotide analogs with novel linkages
DE69033495T2 (en) 1989-10-24 2000-07-20 Isis Pharmaceuticals Inc 2'-MODIFIED NUCLEOTIDES
US5264564A (en) 1989-10-24 1993-11-23 Gilead Sciences Oligonucleotide analogs with novel linkages
US5292873A (en) 1989-11-29 1994-03-08 The Research Foundation Of State University Of New York Nucleic acids labeled with naphthoquinone probe
US5177198A (en) 1989-11-30 1993-01-05 University Of N.C. At Chapel Hill Process for preparing oligoribonucleoside and oligodeoxyribonucleoside boranophosphates
US5130302A (en) 1989-12-20 1992-07-14 Boron Bilogicals, Inc. Boronated nucleoside, nucleotide and oligonucleotide compounds, compositions and methods for using same
US5486603A (en) 1990-01-08 1996-01-23 Gilead Sciences, Inc. Oligonucleotide having enhanced binding affinity
US5587361A (en) 1991-10-15 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5670633A (en) 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US5587470A (en) 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. 3-deazapurines
US5623065A (en) 1990-08-13 1997-04-22 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5578718A (en) 1990-01-11 1996-11-26 Isis Pharmaceuticals, Inc. Thiol-derivatized nucleosides
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5220007A (en) 1990-02-15 1993-06-15 The Worcester Foundation For Experimental Biology Method of site-specific alteration of RNA and production of encoded polypeptides
US5149797A (en) 1990-02-15 1992-09-22 The Worcester Foundation For Experimental Biology Method of site-specific alteration of rna and production of encoded polypeptides
US5214136A (en) 1990-02-20 1993-05-25 Gilead Sciences, Inc. Anthraquinone-derivatives oligonucleotides
AU7579991A (en) 1990-02-20 1991-09-18 Gilead Sciences, Inc. Pseudonucleosides and pseudonucleotides and their polymers
US5321131A (en) 1990-03-08 1994-06-14 Hybridon, Inc. Site-specific functionalization of oligodeoxynucleotides for non-radioactive labelling
US5470967A (en) 1990-04-10 1995-11-28 The Dupont Merck Pharmaceutical Company Oligonucleotide analogs with sulfamate linkages
GB9009980D0 (en) 1990-05-03 1990-06-27 Amersham Int Plc Phosphoramidite derivatives,their preparation and the use thereof in the incorporation of reporter groups on synthetic oligonucleotides
EP0745689A3 (en) 1990-05-11 1996-12-11 Microprobe Corporation A dipstick for a nucleic acid hybridization assay
US5688941A (en) 1990-07-27 1997-11-18 Isis Pharmaceuticals, Inc. Methods of making conjugated 4' desmethyl nucleoside analog compounds
US5623070A (en) 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5541307A (en) 1990-07-27 1996-07-30 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs and solid phase synthesis thereof
US5618704A (en) 1990-07-27 1997-04-08 Isis Pharmacueticals, Inc. Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5677437A (en) 1990-07-27 1997-10-14 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5138045A (en) 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5610289A (en) 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
JPH0874B2 (en) 1990-07-27 1996-01-10 アイシス・ファーマシューティカルス・インコーポレーテッド Nuclease-resistant, pyrimidine-modified oligonucleotides that detect and modulate gene expression
US5218105A (en) 1990-07-27 1993-06-08 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5608046A (en) 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5245022A (en) 1990-08-03 1993-09-14 Sterling Drug, Inc. Exonuclease resistant terminally substituted oligonucleotides
EP0541722B1 (en) 1990-08-03 1995-12-20 Sterling Winthrop Inc. Compounds and methods for inhibiting gene expression
US5177196A (en) 1990-08-16 1993-01-05 Microprobe Corporation Oligo (α-arabinofuranosyl nucleotides) and α-arabinofuranosyl precursors thereof
US5512667A (en) 1990-08-28 1996-04-30 Reed; Michael W. Trifunctional intermediates for preparing 3'-tailed oligonucleotides
US5214134A (en) 1990-09-12 1993-05-25 Sterling Winthrop Inc. Process of linking nucleosides with a siloxane bridge
US5561225A (en) 1990-09-19 1996-10-01 Southern Research Institute Polynucleotide analogs containing sulfonate and sulfonamide internucleoside linkages
WO1992005186A1 (en) 1990-09-20 1992-04-02 Gilead Sciences Modified internucleoside linkages
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
ATE198598T1 (en) 1990-11-08 2001-01-15 Hybridon Inc CONNECTION OF MULTIPLE REPORTER GROUPS ON SYNTHETIC OLIGONUCLEOTIDES
US5672697A (en) 1991-02-08 1997-09-30 Gilead Sciences, Inc. Nucleoside 5'-methylene phosphonates
US5719262A (en) 1993-11-22 1998-02-17 Buchardt, Deceased; Ole Peptide nucleic acids having amino acid side chains
US5714331A (en) 1991-05-24 1998-02-03 Buchardt, Deceased; Ole Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US5539082A (en) 1993-04-26 1996-07-23 Nielsen; Peter E. Peptide nucleic acids
US5371241A (en) 1991-07-19 1994-12-06 Pharmacia P-L Biochemicals Inc. Fluorescein labelled phosphoramidites
US5571799A (en) 1991-08-12 1996-11-05 Basco, Ltd. (2'-5') oligoadenylate analogues useful as inhibitors of host-v5.-graft response
DE59208572D1 (en) 1991-10-17 1997-07-10 Ciba Geigy Ag Bicyclic nucleosides, oligonucleotides, processes for their preparation and intermediates
US5594121A (en) 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
US5276423A (en) 1991-11-12 1994-01-04 Texas Instruments Incorporated Circuit units, substrates therefor and method of making
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
ATE226093T1 (en) 1991-11-26 2002-11-15 Isis Pharmaceuticals Inc INCREASED FORMATION OF TRIPLE AND DOUBLE HELICES FROM OLIGOMERS WITH MODIFIED PYRIMIDINES
TW393513B (en) 1991-11-26 2000-06-11 Isis Pharmaceuticals Inc Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
US5792608A (en) 1991-12-12 1998-08-11 Gilead Sciences, Inc. Nuclease stable and binding competent oligomers and methods for their use
US5359044A (en) 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
US5700922A (en) 1991-12-24 1997-12-23 Isis Pharmaceuticals, Inc. PNA-DNA-PNA chimeric macromolecules
US5565552A (en) 1992-01-21 1996-10-15 Pharmacyclics, Inc. Method of expanded porphyrin-oligonucleotide conjugate synthesis
US5595726A (en) 1992-01-21 1997-01-21 Pharmacyclics, Inc. Chromophore probe for detection of nucleic acid
FR2687679B1 (en) 1992-02-05 1994-10-28 Centre Nat Rech Scient OLIGOTHIONUCLEOTIDES.
EP0564982A3 (en) * 1992-04-04 1995-09-13 Hoechst Ceram Tec Ag Ceramic alumina body with high metallization adherence
US5633360A (en) 1992-04-14 1997-05-27 Gilead Sciences, Inc. Oligonucleotide analogs capable of passive cell membrane permeation
SE9201281L (en) 1992-04-23 1993-10-24 Bioinvent Int Ab Novel human monoclonal antibodies and process for their preparation
US5434257A (en) 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
EP0577558A2 (en) 1992-07-01 1994-01-05 Ciba-Geigy Ag Carbocyclic nucleosides having bicyclic rings, oligonucleotides therefrom, process for their preparation, their use and intermediates
US5272250A (en) 1992-07-10 1993-12-21 Spielvogel Bernard F Boronated phosphoramidate compounds
US5652355A (en) 1992-07-23 1997-07-29 Worcester Foundation For Experimental Biology Hybrid oligonucleotide phosphorothioates
US5574142A (en) 1992-12-15 1996-11-12 Microprobe Corporation Peptide linkers for improved oligonucleotide delivery
US5476925A (en) 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
GB9304618D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Chemical compounds
ATE155467T1 (en) 1993-03-30 1997-08-15 Sanofi Sa ACYCLIC NUCLEOSIDE ANALOGUES AND OLIGONUCLEOTIDE SEQUENCES CONTAINING THEM
EP0691977B1 (en) 1993-03-31 1997-11-26 Sanofi Oligonucleotides with amide linkages replacing phosphodiester linkages
DE4311944A1 (en) 1993-04-10 1994-10-13 Degussa Coated sodium percarbonate particles, process for their preparation and detergent, cleaning and bleaching compositions containing them
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US5457187A (en) 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
US5446137B1 (en) 1993-12-09 1998-10-06 Behringwerke Ag Oligonucleotides containing 4'-substituted nucleotides
WO1995015972A1 (en) 1993-12-09 1995-06-15 Thomas Jefferson University Compounds and methods for site-directed mutations in eukaryotic cells
US5519134A (en) 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US5596091A (en) 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5627053A (en) 1994-03-29 1997-05-06 Ribozyme Pharmaceuticals, Inc. 2'deoxy-2'-alkylnucleotide containing nucleic acid
US5625050A (en) 1994-03-31 1997-04-29 Amgen Inc. Modified oligonucleotides and intermediates useful in nucleic acid therapeutics
US5646269A (en) 1994-04-28 1997-07-08 Gilead Sciences, Inc. Method for oligonucleotide analog synthesis
US5525711A (en) 1994-05-18 1996-06-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pteridine nucleotide analogs as fluorescent DNA probes
US5597696A (en) 1994-07-18 1997-01-28 Becton Dickinson And Company Covalent cyanine dye oligonucleotide conjugates
US5597909A (en) 1994-08-25 1997-01-28 Chiron Corporation Polynucleotide reagents containing modified deoxyribose moieties, and associated methods of synthesis and use
US5580731A (en) 1994-08-25 1996-12-03 Chiron Corporation N-4 modified pyrimidine deoxynucleotides and oligonucleotide probes synthesized therewith
US5792747A (en) 1995-01-24 1998-08-11 The Administrators Of The Tulane Educational Fund Highly potent agonists of growth hormone releasing hormone
US5652356A (en) 1995-08-17 1997-07-29 Hybridon, Inc. Inverted chimeric and hybrid oligonucleotides
US6146894A (en) 1998-04-14 2000-11-14 The Johns Hopkins University Method for generating hypermutable organisms
US6333318B1 (en) * 1998-05-14 2001-12-25 The Salk Institute For Biological Studies Formulations useful for modulating expression of exogenous genes in mammalian systems, and products related thereto
DE60135312D1 (en) 2000-02-11 2008-09-25 Morphotek Inc METHOD FOR THE PRODUCTION OF HYPERMUTIVE BACTERIA
US6825038B2 (en) * 2000-05-12 2004-11-30 The Johns Hopkins University Method for generating hypermutable organisms
ATE423467T1 (en) * 2000-11-07 2009-03-15 Morphotek Inc METHOD FOR PRODUCING GENETICALLY MODIFIED ANTIBODY PRODUCING CELL LINES WITH IMPROVED ANTIBODY PROPERTIES
US6808894B1 (en) 2000-11-07 2004-10-26 Morphotek, Inc. Methods for generating genetically altered antibody producing cell lines with improved antibody characteristics
US7235643B2 (en) * 2000-11-07 2007-06-26 Morphotek, Inc. Antibodies and methods for generating genetically altered antibodies with high affinity
CA2434926C (en) * 2001-01-15 2014-04-01 Morphotek, Inc. Chemical inhibitors of mismatch repair
CA2455686A1 (en) 2001-07-25 2003-02-13 Morphotek Inc. Methods for generating antibiotic resistant microbes and novel antibiotics
ATE548461T1 (en) 2002-01-18 2012-03-15 Morphotek Inc METHOD FOR GENERATING MANIPULATED CELLS FOR LOCUS-SPECIFIC GENE REGULATION AND ANALYSIS
JP4527531B2 (en) * 2002-07-19 2010-08-18 モーフオテク・インコーポレーテツド Method for generating enhanced antibody-producing cell lines with improved growth characteristics
ATE444359T1 (en) 2002-11-15 2009-10-15 Morphotek Inc METHOD FOR GENERATING HIGH ANTIBODIES PRODUCTION FROM HYBRIDOMAS CREATED BY IN VITRO IMMUNIZATION
WO2005011735A1 (en) 2003-07-29 2005-02-10 Morphotek, Inc. Antibodies and methods for generating genetically altered antibodies with enhanced effector function

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BIGGER ET AL.: 'Mutational spectra for polycyclic aromatic hydrocarbons in the supF target gene' MUTATION RESEARCH vol. 450, 2000, pages 75 - 93, XP003001110 *
REYNAUD ET AL.: 'Mismatch repair and immunoglobulin gene hypermutation: did we learn something' IMMUNOLOGY TODAY vol. 20, November 1999, pages 522 - 527, XP004183761 *
See also references of EP1572971A2 *
SPENCER ET AL.: 'Characteristics of Sequences Around the Individual Nucleotide Substitutions in lgVh Genes Suggest Different GC and AT Mutators' JOURNAL OF IMMUNOLOGY vol. 162, 1999, pages 6596 - 6601, XP003001109 *

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9751939B2 (en) 2005-05-18 2017-09-05 Morphosys Ag Anti-GM-CSF antibodies
US11028165B2 (en) 2005-05-18 2021-06-08 Morphosys Ag Nucleic acid encoding anti-GM-CSF antibodies
US10377820B2 (en) 2005-05-18 2019-08-13 Morphosys Ag Methods of treatment using anti-GM-CSF antibodies
EP3181585A3 (en) * 2006-02-08 2017-08-30 Morphotek, Inc. Antigenic gm-csf peptides and antibodies to gm-csf
KR101395515B1 (en) * 2006-02-08 2014-05-14 모르포테크, 인크. Antigenic gm-csf peptides and antibodies to gm-csf
JP2009526082A (en) * 2006-02-08 2009-07-16 モルフォテック、インク. Antigenic GM-CSF peptide and anti-GM-CSF antibody
US7741450B2 (en) 2006-02-08 2010-06-22 Morphotek Inc. Antibodies to GM-CSF
US8318168B2 (en) 2006-02-08 2012-11-27 Morphotek, Inc. Antigenic GM-CSF peptides and antibodies to GM-CSF
AU2007213716B2 (en) * 2006-02-08 2012-12-06 Eisai, Inc. Antigenic GM-CSF peptides and antibodies to GM-CSF
US8623364B2 (en) 2006-02-08 2014-01-07 Morphotek, Inc. Antigenic GM-CSF peptides and antibodies to GM-CSF
EP3181585A2 (en) 2006-02-08 2017-06-21 Morphotek, Inc. Antigenic gm-csf peptides and antibodies to gm-csf
KR101486183B1 (en) * 2006-02-08 2015-01-28 모르포테크, 인크. Antigenic gm-csf peptides and antibodies to gm-csf
AU2007213716C1 (en) * 2006-02-08 2015-05-07 Eisai, Inc. Antigenic GM-CSF peptides and antibodies to GM-CSF
US10023632B2 (en) 2006-02-08 2018-07-17 Morphotek, Inc. Antigenic GM-CSF peptides and antibodies to GM-CSF
WO2007092939A3 (en) * 2006-02-08 2008-03-27 Morphotek Inc Antigenic gm-csf peptides and antibodies to gm-csf
AU2013201228B2 (en) * 2006-02-08 2016-01-21 Eisai, Inc. Antigenic gm-csf peptides and antibodies to gm-csf
US9422367B2 (en) 2006-02-08 2016-08-23 Morphotek, Inc. Antigenic GM-CSF peptides and antibodies to GM-CSF
EP2221384A1 (en) * 2007-12-03 2010-08-25 Kabushiki Kaisya Advance Method for production of antibody
EP2221384A4 (en) * 2007-12-03 2011-04-06 Advance Kk Method for production of antibody
EP2289937A1 (en) * 2009-08-24 2011-03-02 Wilfried Stücker Production of monoclonal antibodies in vitro
WO2011023705A1 (en) * 2009-08-24 2011-03-03 Jansen, Gabriele Production of monoclonal antibodies in vitro
US9206435B2 (en) 2012-08-31 2015-12-08 Synthetic Genomics, Inc. Crowding agent-induced nucleic acid transfer into a recipient host cell
WO2014036404A3 (en) * 2012-08-31 2015-07-30 Synthetic Genomics, Inc. Crowding agent-induced nucleic acid transfer into a recipient host cell
CN110776564A (en) * 2019-10-30 2020-02-11 西北农林科技大学 Two-strain anti-newcastle disease virus nano antibody and expression preparation method and application thereof
CN110776564B (en) * 2019-10-30 2022-02-08 西北农林科技大学 Two-strain anti-newcastle disease virus nano antibody and expression preparation method and application thereof

Also Published As

Publication number Publication date
EP1572971B1 (en) 2009-09-30
JP4555089B2 (en) 2010-09-29
US20040214288A1 (en) 2004-10-28
US20100311169A1 (en) 2010-12-09
AU2003295576B2 (en) 2011-03-17
US7754450B2 (en) 2010-07-13
ATE444359T1 (en) 2009-10-15
DE60329526D1 (en) 2009-11-12
CA2506127A1 (en) 2004-06-03
US8445229B2 (en) 2013-05-21
WO2004046330A3 (en) 2006-09-28
JP2006526983A (en) 2006-11-30
CA2506127C (en) 2013-07-09
AU2003295576A1 (en) 2004-06-15
EP1572971A2 (en) 2005-09-14
EP1572971A4 (en) 2007-07-04

Similar Documents

Publication Publication Date Title
US8445229B2 (en) Methods of generating high-production of antibodies from hybridomas created by in vitro immunization
Han et al. Cellular interaction in germinal centers. Roles of CD40 ligand and B7-2 in established germinal centers.
US20100021996A1 (en) Genetically Altered Anti-body Producing Cell Lines With Improved Antibody Characteristics
CN108136001A (en) Use TGF-β inhibitor and PD-1 inhibitor for treating cancers
JP2019509976A (en) Antibodies specific for glycosylated PD-1 and methods of use thereof
CN108925136A (en) It is specific to glycosylated BTLA(B and T lymphocyte decay factor)Antibody
JP2006503035A (en) Methods for producing antibodies and genetically modified antibodies with high affinity
KR20010052174A (en) Antibodies against human cd40
CN108383909A (en) Anti- SEZ6 antibody and application method
JP2015509947A (en) DLL3 modulator and method of use
TW201309729A (en) Novel modulators and methods of use
KR20140014064A (en) Novel modulators and methods of use
JP2015083016A (en) Method of making hybrid cells that express useful antibodies
CN109415437B (en) Antibodies and molecules that immunospecifically bind to BTN1A1 and therapeutic uses thereof
TW202003567A (en) Single-domain antibodies against LAG-3 and uses thereof
US20100029002A1 (en) Enhancement of hybridoma fusion efficiencies through cell synchronization
CN114728171A (en) anti-TIGIT antibody and application thereof
JP4527531B2 (en) Method for generating enhanced antibody-producing cell lines with improved growth characteristics
CA3123352A1 (en) Method for selecting biological binding molecules
CN113747917A (en) Monoclonal antibody aiming at MHC (major histocompatibility complex) bound human Dickkopf-1 peptide and application thereof
JP7455787B2 (en) Antibodies specific for glycosylated PD-1 and methods of use thereof
US20220185911A1 (en) Therapeutic antibodies for treating lung cancer
RU2776121C2 (en) New anti-pd-l1 antibodies
CN115698070A (en) CD5L binding antibodies and uses thereof

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 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): BW 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
WWE Wipo information: entry into national phase

Ref document number: 2506127

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2004570421

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2003295576

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2003786774

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2003786774

Country of ref document: EP

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