US20060292166A1 - Vaccine composition comprising Flt3-ligand - Google Patents

Vaccine composition comprising Flt3-ligand Download PDF

Info

Publication number
US20060292166A1
US20060292166A1 US11/510,651 US51065106A US2006292166A1 US 20060292166 A1 US20060292166 A1 US 20060292166A1 US 51065106 A US51065106 A US 51065106A US 2006292166 A1 US2006292166 A1 US 2006292166A1
Authority
US
United States
Prior art keywords
flt3
cells
ligand
tumor
antigen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/510,651
Inventor
David Lynch
Luis Borges
Robert Miller
Charles Maliszewski
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Immunex Corp
Original Assignee
Immunex Corp
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 Immunex Corp filed Critical Immunex Corp
Priority to US11/510,651 priority Critical patent/US20060292166A1/en
Publication of US20060292166A1 publication Critical patent/US20060292166A1/en
Abandoned legal-status Critical Current

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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0639Dendritic cells, e.g. Langherhans cells in the epidermis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4615Dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4622Antigen presenting cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464436Cytokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5154Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55516Proteins; Peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/125Stem cell factor [SCF], c-kit ligand [KL]
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/22Colony stimulating factors (G-CSF, GM-CSF)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/24Interferons [IFN]
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/26Flt-3 ligand (CD135L, flk-2 ligand)
    • 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
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/30Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from cancer cells, e.g. reversion of tumour cells

Definitions

  • the present invention relates to a dendritic cell stimulatory factor, to methods of enhancing an immune response in vivo, methods of expanding dendritic cells ex vivo, and to preparations of purified dendritic cells, and to dendritic cell populations useful in the manipulation of T cell-mediated and B-cell mediated immune responses.
  • the objective of vaccination is to provide effective immunity by establishing adequate levels of antibody and a primed population of cells that can rapidly expand on renewed contact with antigen.
  • the first contact with antigen during vaccination must not be injurious to the recipient and thus usually consists of pathogenically-deficient antigen.
  • a frequent difficulty with active immunization protocols is that the vaccine antigen does not possess sufficient immunogenicity to promote a strong immune response, and therefore a sufficient level of protection against subsequent challenge by the same antigen.
  • certain antigens may elicit only weak cell-mediated or antibody responses. For many antigens, both a strong humoral response and a strong cell-mediated response is desirable.
  • Immunopotentiators also known as adjuvants
  • vaccines are compositions of matter that facilitate a strong immune response to a vaccine.
  • the relatively weak immunogenicity of certain novel recombinant antigens has required adjuvants to be more potent.
  • Vaccine adjuvants have different modes of action, affecting the immune response both quantitatively and qualitatively. Such modes of action can be by mobilizing T cells, acting as depots and altering lymphocyte circulation so that these cells remain localized in draining lymph nodes. They may also serve to focus antigen at the site of immunization, thereby allowing antigen specific T cells and B cells to interact more efficiently with antigen-presenting cells.
  • adjuvants may stimulate and affect the behavior of antigen-presenting cells, particularly dendritic cells and macrophages, rendering them more effective for presenting antigen to T cells and B cells.
  • Dendritic cells are a rare and heterogeneous cell population with distinctive morphology and a widespread tissue distribution. A discussion of the dendritic cell system and its role in immunogenicity is provided by Steinman, R. M., Annu. Rev. Immunol., 9:271-296 (1991), incorporated herein by reference. Dendritic cells display an unusual cell surface phenotype and can be characterized by the presence of the cell surface markers CD1, CD4, CD86, CD11c, DEC-205, CD40 or HLA-DR, and the absence of CD14 and other lineage markers.
  • Dendritic cells have a high capacity for sensitizing MHC-restricted T cells and provide an effective pathway for presenting antigens to T cells in situ, both self-antigens during T cell development and foreign antigens during immunity.
  • dendritic cells ex vivo as tumor or infectious disease vaccine adjuvants. See, for example, Romani, et al., J. Exp. Med, 180:83 (1994).
  • the use of dendritic cells as immunostimulatory agents has been limited due to the low frequency of dendritic cells in peripheral blood, the limited accessibility to lymphoid organs and the dendritic cells' terminal state of differentiation.
  • Dendritic cells originate from CD34+ bone marrow progenitors, and the proliferation and maturation of dendritic cells can be enhanced by the cytokines GM-CSF (sargramostim, Leukine®, Immunex Corporation, Seattle, Washington), TNF- ⁇ , c-kit ligand (also known as stem cell factor (SCF), steel factor (SF), or mast cell growth factor (MGF)) and interleukin-4. Therefore, an agent that stimulated the generation of large numbers of functionally mature dendritic cells in vivo or in vitro would be of wide importance.
  • GM-CSF serosis factor
  • SCF stem cell factor
  • SF steel factor
  • MMF mast cell growth factor
  • Flt3-ligand (“flt3-ligand,” “flt3-L,” “Flt3-L”) is known to affect hematopoietic stem and progenitor cells. It was surprisingly found that flt3-ligand can also potently stimulate the generation of downstream or intermediate, cells such as myeloid precursor cells, monocytic cells, macrophages, B cells, and dendritic cells from CD34+ bone marrow progenitors and stem cells.
  • the present invention pertains to a method of mobilizing dendritic cells in vivo, expanding dendritic cells ex vivo and to preparations of dendritic cells. The preparation of purified dendritic cells according to the invention would potentially find use as vaccine adjuvants. Also included within the embodiments of the invention is a method of preparing antigen-specific T cells using the dendritic cells mobilized with flt3-ligand.
  • the invention provides for the use of an effective amount of flt3-ligand to increase or mobilize the numbers of intermediate cells in vivo, for example, in the patient's peripheral blood or spleen. While the invention relates to the generation of large numbers of such downstream and intermediate cells (e.g., myeloid cells, monocytic cells and macrophages) from CD34+ cells using flt3-ligand, the focus is particularly on dendritic cells. By increasing the quantity of the patient's dendritic cells, such cells may themselves be used to present antigen to T cells.
  • the antigen may be one that already exists within the patient, such as a tumor antigen, or a bacterial or viral antigen.
  • Flt3-L may be used, therefore, to increase the numbers of dendritic cells in vivo to boost a patient's immune response against existing antigens.
  • the invention further provides for using combination therapy to enhance a patient's immune response.
  • Such combination therapy includes administering flt3-ligand and one or more therapeutic reagents in amounts sufficiently effective to upregulate the patient's immune response.
  • flt3-ligand may be administered prior to, concurrently with or subsequent to administration of an antigen to a patient for immunization purposes.
  • flt3-ligand can generate large quantities of dendritic cells in vivo to more effectively present the antigen.
  • the overall response is a stronger and improved immune response and more effective immunization to the antigen.
  • the invention also provides a method of generating large quantities of dendritic cells ex vivo.
  • flt3-ligand can be used to expand such cells in vitro (also known as ex vivo expansion) and to drive such CD34+ cells to differentiate into dendritic cells of the lymphoid or myeloid lineage.
  • the resulting collection of dendritic cells can be administered to a patient to provide a stronger and improved immune response to an antigen.
  • the resulting dendritic cells find use as a vaccine adjuvant and can be administered prior to, concurrently with or subsequent to antigen administration.
  • the invention also provides a method of generating large quantities of antigen-presenting dendritic cells ex vivo.
  • flt3-ligand can be used to expand such cells in vitro and to drive such CD34+ cells to differentiate into dendritic cells.
  • the resulting collection of dendritic cells is exposed to an antigen and allowed to process and present the antigen in vitro (this procedure is sometimes referred to in the art as “antigen-pulsing”).
  • An alternate method for preparing dendritic cells that present antigen is to transfect the dendritic cells with a gene encoding an antigen-specific polypeptide. Once the dendritic cells express the antigen, the antigen-presenting dendritic cells can be administered to a patient.
  • the invention also provides for the ex vivo preparation of antigen-specific T cells. Following the procedures described above for preparing large numbers of antigen-presenting dendritic cells ex vivo, the collected antigen-presenting dendritic cells are used to generate antigen-specific T cells from naive T cells that have been collected from a patient. After the antigen has been adequately presented to the T cells generated, the antigen-specific T cells can be administered to the patient.
  • the invention also provides a method of augmenting an immune response in a patient that has an infectious disease wherein the method comprises the step of administering an amount of flt3-ligand sufficient to increase the patient's number of dendritic cells.
  • methods for augmenting an immune response include administering flt3-ligand in combination therapies with additional active compounds, including but not limited to CD40 binding proteins, 4-1BB-L, antibodies to 4-1BB, interferon alpha, RANKL, a CD30 ligand antagonist, and combinations thereof.
  • the invention also provides a method of augmenting an immune response in a patient that has a cancerous or neoplastic disease wherein the method comprises the step of administering an amount of flt3-ligand sufficient to increase the patient's number of dendritic cells.
  • Embodiments of methods for augmenting an immune response include administering flt3-ligand in combination therapies with additional active compounds, including but not limited to CD40 binding proteins, 4-1BB-L, antibodies to 4-1BB, interferon alpha, RANKL, a CD30 ligand antagonist, and combinations thereof. Such method provides a means to enhance the patient's tumor-specific immune response.
  • a method for enhancing a patient's autoimmune tolerance comprising the step of administering an amount of flt3-ligand sufficient to increase the patient's number of dendritic cells. Further included are methods for promoting survival of grafts and transplanted tissues and organs.
  • the methods of the invention can further comprise the use of an effective amount of a cytokine in sequential or concurrent combination with flt3-ligand.
  • cytokines include, but are not limited to, interleukins (“ILs”) IL-3 and IL-4, a colony stimulating factor (“CSF”) selected from the group consisting of granulocyte macrophage colony stimulating factor (“GM-CSF”) or GM-CSF/IL-3 fusions, or other cytokines such as TNF- ⁇ , CD40 binding proteins (e.g.
  • CD40-L 4-1BB antagonists (e.g., antibodies immunoreactive with 4-1BB and 4-IBB-L), interferon alpha, RANKL, CD30 ligand antagonists (e.g., CD30 ligand monoclonal antibodies and soluble CD30-Fc fusion polypeptides), and c-kit ligand.
  • 4-1BB antagonists e.g., antibodies immunoreactive with 4-1BB and 4-IBB-L
  • interferon alpha RANKL
  • CD30 ligand antagonists e.g., CD30 ligand monoclonal antibodies and soluble CD30-Fc fusion polypeptides
  • c-kit ligand e.g., CD40 ligand monoclonal antibodies and soluble CD30-Fc fusion polypeptides
  • the invention further includes a dendritic cell expansion medium comprising cell growth medium, autologous serum, and flt3-ligand alone or in combination with a cytokine from the group listed above.
  • the invention is directed to the use of flt3-ligand to generate large numbers of intermediate cell types from CD34+ hematopoietic progenitor cells and stem cells.
  • Such intermediate cell types include myeloid cells, monocytic cells, macrophages and dendritic cells.
  • the large numbers of these intermediate cell types are not naturally found in vivo and can be generated by administering flt3-ligand.
  • Such enhancement in overall cell number can augment the immune response to antigen in the host.
  • Another embodiment of the invention is the isolation and use of such intermediate cell types as antigen-presenting cells or the use as vaccine adjuvants.
  • the invention while particularly focused on the embodiment concerning dendritic cells, is also applicable to myeloid, monocytic and macrophage cell types.
  • flt3-ligand refers to a genus of polypeptides that are described in U.S. Pat. No. 5,554,512, EP 0627487 A2 and in WO 94/28391, both incorporated herein by reference.
  • a human flt3-ligand cDNA was deposited with the American Type Culture Collection, Rockville, Md., USA (ATCC) on Aug. 6, 1993 and assigned accession number ATCC 69382. The deposit was made under the terms of the Budapest Treaty.
  • Flt3-L can be made according to the methods described in the documents cited above.
  • flt3-L encompasses proteins having the amino acid sequence 1 to 235 of SEQ ID NO:2, as well as those proteins having a high degree of similarity or a high degree of identity with the amino acid sequence 1 to 235 of SEQ ID NO:2, and which proteins are biologically active and bind the flt3 receptor.
  • the term refers to biologically active gene products of the DNA of SEQ ID NO:1.
  • flt3-L the membrane-bound proteins (which include an intracellular region, a membrane region, and an extracellular region), and soluble or truncated proteins which comprise primarily the extracellular portion of the protein, retain biological activity and are capable of being secreted.
  • soluble proteins are those comprising the sequence of amino acids 28-160 of SEQ ID NO:2.
  • a “flt3-L variant” as referred to herein, means a polypeptide substantially homologous to native flt3-L, but which has an amino acid sequence different from that of native flt3-L (human, murine or other mammalian species) because of one or more deletions, insertions or substitutions.
  • the variant amino acid sequence preferably is at least 80% identical to a native flt3-L amino acid sequence, most preferably at least 90% identical. The percent identity may be determined, for example, by comparing sequence information using the GAP computer program, version 6.0 described by Devereux et al. ( Nucl. Acids Res. 12:387, 1984) and available from the University of Wisconsin Genetics Computer Group (UWGCG).
  • the GAP program utilizes the alignment method of Needleman and Wunsch ( J. Mol. Biol. 48:443, 1970), as revised by Smith and Waterman ( Adv. Appl. Math 2:482, 1981).
  • the preferred default parameters for the GAP program include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non-identities) for nucleotides, and the weighted comparison matrix of Gribskov and Burgess, Nucl. Acids Res. 14:6745, 1986, as described by Schwartz and Dayhoff, eds., Atlas of Protein Sequence and Structure , National Biomedical Research Foundation, pp.
  • Variants may comprise conservatively substituted sequences, meaning that a given amino acid residue is replaced by a residue having similar physiochemical characteristics. Examples of conservative substitutions include substitution of one aliphatic residue for another, such as Ile, Val, Leu, or Ala for one another, or substitutions of one polar residue for another, such as between Lys and Arg; Glu and Asp; or Gln and Asn. Other such conservative substitutions, for example, substitutions of entire regions having similar hydrophobicity characteristics, are well known. Naturally occurring flt3-L variants are also encompassed by the invention.
  • variants are proteins that result from alternate mRNA splicing events or from proteolytic cleavage of the flt3-L protein, wherein the flt3-L binding property is retained.
  • Alternate splicing of mRNA may yield a truncated but biologically active flt3-L protein, such as a naturally occurring soluble form of the protein, for example.
  • Variations attributable to proteolysis include, for example, differences in the N- or C-termini upon expression in different types of host cells, due to proteolytic removal of one or more terminal amino acids from the flt3-L protein (generally from 1-5 terminal amino acids).
  • an aspect of the invention is soluble flt3-L polypeptides.
  • Soluble flt3-L polypeptides comprise all or part of the extracellular domain of a native flt3-L but lack the transmembrane region that would cause retention of the polypeptide on a cell membrane.
  • Soluble flt3-L polypeptides advantageously comprise the native (or a heterologous) signal peptide when initially synthesized to promote secretion, but the signal peptide is cleaved upon secretion of flt3-L from the cell.
  • Soluble flt3-L polypeptides encompassed by the invention retain the ability to bind the flt3 receptor.
  • soluble flt3-L may also include part of the transmembrane region or part of the cytoplasmic domain or other sequences, provided that the soluble flt3-L protein can be secreted.
  • Soluble flt3-L may be identified (and distinguished from its non-soluble membrane-bound counterparts) by separating intact cells which express the desired protein from the culture medium, e.g., by centrifugation, and assaying the medium (supernatant) for the presence of the desired protein. The presence of flt3-L in the medium indicates that the protein was secreted from the cells and thus is a soluble form of the desired protein.
  • Soluble forms of flt3-L possess many advantages over the native bound flt3-L protein. Purification of the proteins from recombinant host cells is feasible, since the soluble proteins are secreted from the cells. Further, soluble proteins are generally more suitable for intravenous administration.
  • soluble flt3-L polypeptides include those comprising a substantial portion of the extracellular domain of a native flt3-L protein.
  • Such soluble mammalian flt3-L proteins comprise amino acids 28 through 182 of SEQ ID NO:2.
  • truncated soluble flt3-L proteins comprising less than the entire extracellular domain are included in the invention.
  • Such truncated soluble proteins are represented by the sequence comprising amino acids 28-160 of SEQ ID NO:2.
  • soluble flt3-L may additionally comprise one of the heterologous signal peptides described below that is functional within the host cells employed.
  • the protein may comprise the native signal peptide, such that the mammalian flt3-L comprises 1 through 182 of SEQ ID NO:2. Isolated DNA sequences encoding soluble flt3-L proteins are encompassed by the invention.
  • IL-3 refers to a genus of interleukin-3 polypeptides as described in U.S. Pat. No. 5,108,910, incorporated herein by reference. Such polypeptides include analogs that have amino acid sequences that are substantially similar to the native human interleukin-3 amino acid sequences disclosed, for example, in EP publ. Nos. 275,598 and 282,185, each incorporated herein by reference.
  • IL-3 also includes analogs and alleles of IL-3 molecules that exhibit at least some of the biological activity in common with native human IL-3. Exemplary analogs of IL-3 are disclosed in EP Publ. No. 282,185.
  • IL-3 forms include human IL-3[Pro 8 Asp 15 Asp 70 ], human IL-3[Ser 8 Asp 15 Asp 70 ] and human IL-3[Ser 8 ].
  • a DNA sequence encoding human IL-3 protein suitable for use in the invention is publicly available from the American Type Culture Collection (ATCC) under accession number ATCC 67747. The nomenclature used herein with respect to amino acid sequences in brackets designates which amino acids differ from the native human form.
  • human IL-3[Ser 8 Asp 15 Asp 70 ] refers to a human IL-3 protein in which amino acid 8 has been changed to a serine residue, amino acid 15 has been changed to an aspartic acid residue and the amino acid 70 has been changed to an aspartic acid residue.
  • IL-4 refers to a polypeptide as described in Mosley et al., Cell 59:335 (1989), Idzerda et al., J. Exp. Med. 171:861 (1990) and Galizzi et al., Intl. Immunol. 2:669 (1990), each of which is incorporated herein by reference.
  • Such IL-4 polypeptide includes analogs that have an amino acid sequence that is substantially similar to the native human IL-4 amino acid sequences described in Mosley et al., Idzerda et al., and Galizzi et al.
  • IL-4 also includes analogs of native human IL-4 molecules sufficient to retain biological activity of native human IL-4.
  • GM-CSF refers to a genus of proteins as described in U.S. Pat. Nos. 5,108,910, and 5,229,496 each of which is incorporated herein by reference. Such proteins include analogs that have an amino acid sequence that is substantially similar to native human GM-CSF amino acid sequences (e.g., as publicly available ATCC 53157 or ATCC 39900), and which are biologically active in that they are capable of binding to a GM-CSF receptor, transducing a biological signal initiated by binding GM-CSF receptor, or cross-reacting with anti-GM-CSF antibodies. Amino acid sequences are disclosed, for example in Anderson, et al., Proc. Natl. Acad.
  • GM-CSF also includes analogs of the native human GM-CSF molecules described in U.S. Pat. Nos. 5,108,910, and 5,229,496 sufficient to retain biological activity of native human GM-CSF.
  • Exemplary analogs of GM-CSF include, for example, those described in EP Publ. No. 212914 and WO 89/03881, each of which is incorporated herein by reference.
  • Other analogs of GM-CSF also may be used to construct fusion proteins with IL-3.
  • a DNA sequence encoding a particularly preferred GM-CSF protein having potential glycosylation sites removed is publicly available from the ATCC under accession numbers ATCC 67231.
  • GM-CSF/IL-3 fusion protein means a C-terminal to N-terminal fusion of GM-CSF and IL-3.
  • the fusion proteins are known and are described in U.S. Pat. Nos. 5,199,942, 5,108,910 and 5,073,627, each of which is incorporated herein by reference.
  • a preferred fusion protein is PIXY321 as described in U.S. Pat. No. 5,199,942.
  • c-kit ligand also known as Mast Cell Growth Factor (MGF), Steel Factor or Stem Cell Factor (SCF), refers to a polypeptide described in EP 423,980, which is incorporated herein by reference, and that claims priority from U.S. patent application Ser. No. 589,701, filed Oct. 1, 1990.
  • Such c-kit ligand polypeptide includes analogs that have an amino acid sequence that is substantially similar to the native human c-kit ligand amino acid sequences described in EP 423,980 and which are biologically active in that they are capable of binding to a c-kit receptor, transducing a biological signal initiated by binding c-kit receptor, or cross-reacting with anti-c-kit ligand antibodies.
  • c-kit ligand also includes analogs of native human c-kit ligand molecules sufficient to retain biological activity of native human c-kit ligand.
  • CD40 binding protein refers to polypeptides that bind CD40, including but not limited to CD40-L and antibodies immunoreactive with CD40, as described in PCT publications WO 93/08207 and WO 96/40918 each of which is incorporated herein by reference.
  • Such CD40 binding proteins include analogs that have an amino acid sequence that is substantially similar to the native human CD40-L amino acid sequences described in the PCT publications, and which are biologically active in that they are capable of binding to CD40, transducing a biological signal initiated by binding CD40, or cross-reacting with anti-CD40 antibodies.
  • the term CD40 binding proteins also includes analogs of native human CD40-L and antibodies reactive with CD40 that are sufficiently homologous to native molecules so as to retain biological activity of native CD40 binding proteins.
  • 4-1BB-L and “antibody to 4-1BB” refer to molecules that are described in U.S. Pat. No.5,674,704 and Alderson et al. Eur. J. Immunol. 24:2219-2227, 1994, which are incorporated herein by reference.
  • 4-1BB-L includes analogs that have an amino acid sequence that is substantially similar to the native 4-1BB-L amino acid sequence described in the above mentioned publication and which are biologically active in that they are capable of binding to 4-1BB or transducing a biological signal initiated by binding 4-1BB, such as inducing a proliferative response in stimulated primary T cells.
  • 4-1BB-L and antibodies to 4-1BB also includes analogs of native 4-1BB-L and analogs of antibodies reactive with 4-1BB that are sufficiently homologous to the native compounds so as to retain biological activity of 4-1BB-L and the described antibodies.
  • interferon alpha refers to naturally occurring forms of interferon alpha (leukocyte interferon) as isolated from cells, cell lines, tissue, and other sources, as well as recombinant forms, consensus forms, analogs, and variant forms that exhibit interferon alpha biological activity.
  • Hybrid “interferon alpha A/D,” also called “Universal Type I Interferon,” has been described by Fisher et al., Biochem Biophys. Res. Commun. 119(1):108 (1984), incorporated herein by reference.
  • Interferon alpha polypeptides have been described in, inter alia, Pestka et al., Ann. Rev. Biochem.
  • CD30 ligand antagonist refers to any molecule that interferes with the binding between CD30 ligand and the CD30 receptor.
  • CD30 ligand is described, inter alia, in U.S. Pat. No. 5,480,981 which is incorporated herein by reference.
  • CD30 ligand antagonists include, but are not limited to, CD30 ligand antibodies (including CD30 ligand monoclonal antibodies) and soluble CD30 polypeptides and analogs thereof (including soluble CD30-Fc fusion polypeptides).
  • CD30 ligand antagonists may also be antisense nucleic acids, ribozymes, muteins, aptamers, and small molecules that interfere with the CD30-CD30 ligand interaction.
  • RANK ligand (“RANKL”) polypeptides are described in, inter alia, WO 98/28426; “TRAIL” polypeptides are described in, inter alia, U.S. Pat. No. 5,763,223; “Tek” (also called Tie2, ork) polypeptides are described in, inter alia, U.S. Pat. No. 5,447,860. These publications are incorporated herein by reference.
  • adjuvant refers to a substance that enhances, augments or potentiates the host's immune response to a vaccine antigen.
  • Antagonist includes various classes of molecules that are capable of interfering with a specified biological interaction and/or activity. Antagonists include, but are not limited to, antibodies, soluble forms of a target polypeptide, antisense nucleic acids, ribozymes, muteins, aptamers, and small molecules.
  • the procedure for “ex vivo expansion” of hematopoietic stem and progenitor cells is described in U.S. Pat. No. 5,199,942, incorporated herein by reference. Briefly, the term means a method comprising: (1) collecting CD34 + hematopoietic stem and progenitor cells from a patient from peripheral blood harvest or bone marrow explants; and (2) expanding such cells ex vivo .
  • other factors such as flt3-ligand, IL-1, IL-3, c-kit ligand, can be used.
  • immunogenicity means relative effectiveness of an immunogen or antigen to induce an immune response.
  • substantially similar means a variant amino acid sequence preferably that is at least 80% identical to a native amino acid sequence, most preferably at least 90% identical.
  • Percent identity may be determined by visual inspection. Percent identity may be determined using the alignment method of Needleman and Wunsch (J. Mol. Biol. 48:443, 1970) as revised by Smith and Waterman (Adv. Appl. Math 2:482, 1981). Preferably, percent identity is determined by using a computer program, for example, the GAP computer program version 10.x available from the Genetics Computer Group (GCG; Madison, Wis., see also Devereux et al., Nucl. Acids Res. 12:387, 1984).
  • the preferred default parameters for the GAP program include: (1) a comparison matrix containing a value of 1 for identities and 0 for non-identities for nucleotides, and the weighted comparison matrix of Gribskov and Burgess, Nucl. Acids Res. 14:6745, 1986, as described by Schwartz and Dayhoff, eds., Atlas of Protein Sequence and Structure , National Biomedical Research Foundation, pp. 353-358, 1979 for amino acids; (2) a penalty of 30 (amino acids) or 50 (nucleotides) for each gap and an additional 1 (amino acids) or 3 (nucleotides) penalty for each symbol in each gap; (3) no penalty for end gaps; and (4) no maximum penalty for long gaps.
  • Variants may comprise conservatively substituted sequences, meaning that a given amino acid residue is replaced by a residue having similar physiochemical characteristics.
  • conservative substitutions include substitution of one aliphatic residue for another, such as Ile, Val, Leu, or Ala for one another, or substitutions of one polar residue for another, such as between Lys and Arg; Glu and Asp; or Gln and Asn.
  • Other such conservative substitutions for example, substitutions of entire regions having similar hydrophobicity characteristics, are well known.
  • Naturally occurring variants are also encompassed by the invention. Examples of such variants are proteins that result from alternate mRNA splicing events or from proteolytic cleavage of the native protein, wherein the native biological property is retained.
  • vaccine means an organism or material that contains an antigen in an innocuous form.
  • the vaccine is designed to trigger an immunoprotective response.
  • the vaccine may be recombinant or non-recombinant. When inoculated into a non-immune host, the vaccine will provoke active immunity to the organism or material, but will not cause disease.
  • Vaccines may take the form, for example, of a toxoid, which is defined as a toxin that has been detoxified but that still retains its major immunogenic determinants; or a killed organism, such as typhoid, cholera and poliomyelitis; or attenuated organisms, that are the live, but non-virulent, forms of pathogens, or it may be antigen encoded by such organism, or it may be a live tumor cell or an antigen present on a tumor cell.
  • a toxoid which is defined as a toxin that has been detoxified but that still retains its major immunogenic determinants
  • a killed organism such as typhoid, cholera and poliomyelitis
  • attenuated organisms that are the live, but non-virulent, forms of pathogens, or it may be antigen encoded by such organism, or it may be a live tumor cell or an antigen present on a tumor cell.
  • a variety of cell selection techniques are known for identifying and separating CD34 + hematopoietic stem or progenitor cells from a population of cells. Methods and materials for identifying and selecting such cell types are known. For example, monoclonal antibodies can be used to bind to a marker protein or surface antigen protein found on stem or progenitor cells. Such markers or cell surface antigens for hematopoietic stem cells include CD34 and Thy-1. In one method, antibodies are fixed to a surface, for example, glass beads, and contacted with a mixture of cells suspected of containing stem cells. This permits the antibodies to bind and secure the stem cells to the glass beads.
  • the antibodies can be incubated with the cell mixture and the resulting combination contacted with a surface having an affinity for the antibody-cell complex.
  • Undesired cells and cell matter are removed providing a relatively pure population of stem cells.
  • Stem or progenitor cells having the CD34 marker constitute only about 1% to 3% of the mononuclear cells in the bone marrow.
  • the amount of CD34 + stem or progenitor cells in the peripheral blood is approximately 10- to 100-fold less than in bone marrow.
  • Hematopoietic stem cells are selectable by virtue of their physical characteristics, such as expressing the membrane-bound flt3 receptor, or having the following cellular markers: CD34 or Thy-1.
  • Monoclonal antibodies that recognize any of these antigens have been described in U.S. Pat. No. 4,714,680 (anti-My-10) incorporated herein by reference, anti-CD34 is commercially available from Becton Dickinson, Franklin Lakes, N.J.), and anti-Thy-1 monoclonal antibodies can be readily generated using the methods described by Dalchau et al., J. Exp. Med.
  • a flt3 receptor binding protein also may be used, such as anti-flt3 monoclonal antibodies or the flt3-ligand.
  • the cell binding protein is brought into contact with the collected cell mixture and the combination is allowed to incubate for a period of time sufficient to permit the binding of the desired cell to the cell binding protein.
  • An alternative means of selecting the quiescent stem cells is to induce cell death in the dividing, more lineage-committed, cell types using an antimetabolite such as 5-fluorouracil (5-FU) or an alkylating agent such as 4-hydroxycyclophosphamide (4-HC).
  • the non-quiescent cells are stimulated to proliferate and differentiate by the addition of growth factors that have little or no effect on the stem cells, causing the non-stem cells to proliferate and differentiate and making them more vulnerable to the cytotoxic effects of 5-FU or 4-HC. See Berardi et al., Science, 267:104 (1995), which is incorporated herein by reference.
  • Isolation of the hematopoietic stem or progenitor cells can be performed by using, for example, affinity chromatography, antibody-coated magnetic beads, or antibodies fixed to a solid matrix, such as glass beads, flasks, etc.
  • Antibodies that recognize a stem or progenitor cell surface marker can be fused or conjugated to other chemical moieties such as biotin—which can be removed with an avidin or a streptavidin moiety secured to a solid support; fluorochromes useful in fluorescence activated cell sorting (FACS), or the like.
  • FACS fluorescence activated cell sorting
  • isolation is accomplished by an immunoaffinity column. Immunoaffinity columns can take any form, but usually comprise a packed bed reactor.
  • the packed bed in these bioreactors is preferably made of a porous material having a substantially uniform coating of a substrate.
  • the porous material which provides a high surface area-to-volume ratio, allows for the cell mixture to flow over a large contact area while not impeding the flow of cells out of the bed.
  • Typical substrates include avidin and streptavidin, while other conventional substrates can be used.
  • the substrate should, either by its own properties, or by the addition of a chemical moiety, display high-affinity for a moiety found on the cell-binding protein such as a monoclonal antibody.
  • the monoclonal antibodies recognize a cell surface antigen on the cells to be separated, and are typically further modified to present a biotin moiety.
  • biotin has a high affinity for avidin, and the affinity of these substances thereby removably secures the monoclonal antibody to the surface of the packed bed.
  • Such columns are well known in the art, see Berenson, et al., J. Cell Biochem., 10D:239 (1986).
  • the column is washed with a PBS solution to remove unbound material.
  • Target cells can be released from the beads using conventional methods.
  • Immunoaffinity columns of the type described above that utilize biotinylated anti-CD34 monoclonal antibodies secured to an avidin-coated packed bed are described for example, in PCT Publ. No. WO 93/08268.
  • a variation of this method utilizes cell binding proteins, such as the monoclonal antibodies or flt3-ligand as described above, removably-secured to a fixed surface in the isolating means.
  • the bound cell binding protein then is contacted with the collected cell mixture and allowed to incubate for a period of time sufficient to permit isolation of the desired cells.
  • the monoclonal antibodies that recognize the cell surface antigens can be labeled with a fluorescent label, e.g., chromophore or fluorophore, and separated by cell sorting according to the presence of absence or the amount of labeled product.
  • a fluorescent label e.g., chromophore or fluorophore
  • CD34 + cells are then exposed to either flt3-ligand alone or flt3-ligand in concurrent or sequential combination with one or more of the following cytokines: GM-CSF, TNF- ⁇ , IL-3, IL-4, c-kit-ligand, CD40-L, 4-IBB-L or GM-CSF/IL-3 fusion proteins.
  • CD34 + cells then are allowed to differentiate and commit to cells of the dendritic lineage.
  • the dendritic cells are collected and can either be (a) administered to a patient in order to augment the immune system and T-cell mediated or B-cell mediated immune responses to antigen, (b) exposed to an antigen prior to administration of the dendritic cells into a patient, (c) transfected with a gene encoding an antigen-specific polypeptide or (d) exposed to an antigen and then allowed to process and present the antigen, ex vivo, to T-cells collected from the patient followed by administration of the antigen-specific T-cells to the patient.
  • the invention provides for the use of an effective amount of flt3-ligand to increase or mobilize dendritic cells in vivo, for example, in the patient's peripheral blood or spleen.
  • the antigen may be one that already exists within the patient, such as a tumor antigen, or a bacterial or viral antigen.
  • Flt3-L may be used, therefore, to boost the patient's lymphocyte-mediated (e.g., T cell and B cell mediated) or myeloid-mediated immune response to the already present antigens thus potentially enabling a more effective antigen-presentation to the patient's T cells.
  • flt3-L may be used in combination therapies with one or more additional agents to enhance an immune response against tumor, viral or bacterial antigens.
  • CD40 binding proteins which enhance the ability of dendritic cells to process and present antigens to effector T cells can be administered in combination with flt3-L to dramatically enhance an immune response.
  • immune responses can include responses against viral or bacterial antigens that are responsible for infectious diseases and immune responses to tumor antigens.
  • Represen-tative CD40 binding proteins useful in combination therapy with flt3-L include CD40-L and antibodies immunoreactive with CD40 which are described in PCT publications WO 93/08207 and WO 96/40918.
  • 4-1BB-L and antibodies reactive with 4-1BB can be administered in combination with flt3-L to dramatically enhance immune responses.
  • 4-1BB-L and antibodies reactive with 4-1BB can be used in combination therapies to enhance immune responses to viral antigens and bacterial antigens responsible for infectious diseases and to enhance immune responses to tumor antigens. More particularly, as described in Example 5, when used in a combination therapy there is a surprising synergy between flt3-L and a 4-1BB-L or antibodies to 4-1BB for anti-tumor immune responses.
  • 4-IBB-L and antibodies reactive with 4-1BB are described in U.S. Pat. No. 5,674,704. The surprising synergy in the above described combination therapies for their ability to dramatically enhance anti-tumor immune responses suggests that stimulating more than one mechanism or more than one cell population is a promising approach to cancer treatment.
  • interferon alpha, RANKL, or a CD30 ligand antagonist can be administered in combination with flt3-L to dramatically enhance immune responses.
  • flt3-L can be administered in combination with flt3-L to dramatically enhance immune responses.
  • flt3-L can be administered in combination with flt3-L to dramatically enhance immune responses.
  • flt3-L when used in a combination therapy there is a surprising synergy between flt3-L and interferon alpha for anti-tumor immune responses.
  • flt3-L Other molecules that may be used in combination with flt3-L according to the present invention include IL-2, IL-12, IL-15, TRAIL, Fas ligand, VEGF antagonists, Tek antagonists, molecules that enhance dendritic cell function, survival, or expansion, molecules that enhance T cell activation or differentiation, molecules that enhance dendritic cell migration including various chemokines, molecules that increase the availability of target cell antigens, such as apoptotic factors and molecules that enhance MHC Class I presentation including the various interferons, angiogenesis inhibitors, inhibitors of immunosuppressive molecules released by tumors including IL-10, VEGF, and TGF- ⁇ , and tumor-specific antibodies including toxin- or radio-labeled antibodies.
  • flt3-ligand may be administered prior to, concurrently with or subsequent to administration of an antigen to a patient for immunization purposes.
  • flt3-ligand can generate large quantities of dendritic cells in vivo to more effectively present the antigen.
  • the overall response is a stronger and improved immune response and more effective immunization to the antigen.
  • flt3-L may be administered as a vaccine adjuvant in combination with additional active compounds prior to, concurrently with or subsequent to administration of an antigen to a patient for immunization purposes to enhance an immune response against tumor, viral or bacterial antigens.
  • CD40 binding proteins such as CD40-L and antibodies to CD40 which enhance the ability of dendritic cells to present antigens to T cells can be administered in combination with flt3-L to dramatically enhance an immune response.
  • 4-1BB-L, antibodies reactive with 4-1BB, interferon alpha, RANKL, or CD30 ligand antagonists can be administered in combination with flt3-L to enhance an immune response and provide more effective immunization to the antigen.
  • flt3-ligand not only is effective as a vaccine adjuvant, but as discussed supra., is effective in augmenting an immune response against previously existing antigens.
  • the inventors have shown that flt3-ligand administration to tumor-bearing mice results in at least a significant decrease in the growth rate of the tumor, and can result in tumor regression in a large proportion of the mice.
  • the data are presented in more detail in Example 3.
  • Flt3-L therefore is an important cytokine in the generation of an effective immune response in vivo against antigen.
  • Data in Examples 4-6 demonstrate that when used in combination therapy with additional agents, flt3-L can provide a dramatically enhanced immune response in vivo against antigen.
  • flt3-ligand Because of its ability to generate dendritic cells, flt3-ligand also finds use in promoting the survival of transplanted tissue or organs.
  • allogeneic organs or other tissue When allogeneic organs or other tissue is transplanted into a host they can transfer stem cells, immature dendritic cells, and mature dendritic cells from the donor. These cells are called passenger cells and such cells can graft into the hematopoietic system of the host. Additionally, stem cells, immature dendritic cells, and mature dendritic cells from the host may graft to the donor organ or tissue.
  • T-cells that recognize the major histocompatability complex (MHC) molecules that the dendritic cells express.
  • MHC major histocompatability complex
  • the donor T cells are “screened” so that they fail to recognize and react against the host (i.e., no graft versus host disease) and the host T-cells are screened so that they fail to recognize and react against the graft (i.e., no graft rejection).
  • a mutual tolerance can be achieved, and the graft is accepted.
  • Administration of flt3-ligand to the host or donor prior to transplantation would generate increased numbers of dendritic cells in such host or donor and permit increased tolerance and survival of the graft.
  • Suitable growth media are solutions containing nutrients or metabolic additives, and include those that are serum-depleted or serum-based.
  • Representative examples of growth media are RPMI, TC 199, Iscoves modified Dulbecco's medium (Iscove, et al., F. J. Exp. Med., 147:923 (1978)), DMEM, Fischer's, alpha medium, NCTC, F-10, Leibovitz's L-15, MEM and McCoy's.
  • nutrients include, serum albumin, transferrin, lipids, cholesterol, a reducing agent such as 2-mercaptoethanol or monothioglycerol, pyruvate, butyrate, and a glucocorticoid such as hydrocortisone 2-hemisuccinate.
  • the standard media includes an energy source, vitamins or other cell-supporting organic compounds, a buffer such as HEPES, Tris, that act to stabilize the pH of the media, various inorganic salts.
  • a buffer such as HEPES, Tris
  • peripheral blood progenitor cells PBPC
  • peripheral blood stem cells PBSC
  • PBPC and PBSC are collected using conventional devices, for example, a Haemonetics® Model V50 apheresis device (Haemonetics, Braintree, Mass.).
  • MNC mononuclear cells
  • the cells are suspended in standard media and then centrifuged to remove red blood cells and neutrophils. Cells located at the interface between the two phases (also known in the art as the buffy coat) are withdrawn and resuspended in HBSS. The suspended cells are predominantly mononuclear and a substantial portion of the cell mixture are early stem cells.
  • the resulting stem cell suspension then can be contacted with biotinylated anti-CD34 monoclonal antibodies or other cell-binding means. The contacting period is maintained for a sufficient time to allow substantial interaction between the anti-CD34 monoclonal antibodies and the CD34 antigens on the stem cell surface. Typically, times of at least one hour are sufficient.
  • the cell suspension then is brought into contact with the isolating means provided in the kit.
  • the isolating means can comprise a column packed with avidin-coated beads. Such columns are well known in the art, see Berenson, et al., J. Cell Biochem., 10D:239 (1986). The column is washed with a PBS solution to remove unbound material. Target stem cells can be released from the beads and from anti-CD34 monoclonal antibody using conventional methods. The stem cells obtained in this manner can be frozen in a controlled rate freezer (e.g., Cryo-Med®, Mt. Clemens, Mich.), then stored in the vapor phase of liquid nitrogen. Ten percent dimethylsulfoxide can be used as a cryoprotectant. After all collections from the donor have been made, the stem cells are thawed and pooled.
  • a controlled rate freezer e.g., Cryo-Med®, Mt. Clemens, Mich.
  • human IL-1 ⁇ or IL-4 may be added to the cultures.
  • the most preferred combination of expansion factors comprises flt3-ligand plus either IL-3 or a GM-CSF/IL-3 fusion protein.
  • flt3-ligand can be formulated according to known methods used to prepare pharmaceutically useful compositions.
  • Flt3-L can be combined in admixture, either as the sole active material or with other known active materials (e.g.
  • CD40 binding proteins such as CD40-L or antibodies reactive with CD40, 4-1BB-L or antibodies reactive with 4-1BB, interferon alpha, RANKL, CD30 ligand antagonists
  • pharmaceutically suitable diluents e.g., Tris-HCl, acetate, phosphate
  • preservatives e.g., Thimerosal, benzyl alcohol, parabens
  • emulsifiers e.g., solubilizers, adjuvants and/or carriers.
  • Suitable carriers and their formulations are described in Remington's Pharmaceutical Sciences, 16th ed. 1980, Mack Publishing Co.
  • compositions can contain flt3-ligand complexed with polyethylene glycol (PEG), metal ions, or incorporated into polymeric compounds such as polyacetic acid, polyglycolic acid, hydrogels, etc., or incorporated into liposomes, microemulsions, micelles, unilamellar or multilamellar vesicles, erythrocyte ghosts or spheroblasts.
  • PEG polyethylene glycol
  • metal ions or incorporated into polymeric compounds such as polyacetic acid, polyglycolic acid, hydrogels, etc.
  • liposomes such as polyacetic acid, polyglycolic acid, hydrogels, etc.
  • Such compositions will influence the physical state, solubility, stability, rate of in vivo release, and rate of in vivo clearance of flt3-ligand.
  • Flt3-L can be administered topically, parenterally, or by inhalation.
  • parenteral includes subcutaneous injections, intravenous, intramuscular, intracisternal injection, or infusion techniques. These compositions will typically contain an effective amount of the flt3-ligand, alone or in combination with an effective amount of any other active material, e.g. those described above. Effective amounts, or dosages, and desired concentrations of flt3-L and active compounds (e.g.
  • CD40-L and/or 4-1BB-L, antibodies reactive with 4-1BB, interferon alpha, RANKL, CD30 ligand antagonists) contained in the compositions may vary depending upon many factors, including the intended use, patient's body weight and age, and route of administration. Preliminary doses can be determined according to animal tests, and the scaling of dosages for human administration can be performed according to art-accepted practices. Keeping the above description in mind, typical dosages of flt3-ligand may range from about 10 ⁇ g per square meter to about 1000 ⁇ g per square meter. A preferred dose range is on the order of about 100 ⁇ g per square meter to about 300 ⁇ g per square meter.
  • This Example describes a method for using flt3-ligand to generate large numbers of dendritic cells ex vivo.
  • Cells having the CD34 + phenotype are isolated as described above, for example, first by generating a buffy coat of cells using a procedure described supra. Cells from the buffy coat are then incubated with a CD34 specific monoclonal antibody.
  • the CD34 + cells which are selected then are cultured in McCoy's enhanced media with 20 ng/ml each of GM-CSF, IL-4, TNF- ⁇ , or 100 ng/ml flt3-ligand or c-kit ligand. The culture is continued for approximately two weeks at 37° C. in 10% CO 2 in humid air.
  • Cells then are sorted by flow cytometry for CD1a + and HLA-DR + expression.
  • the combination of GM-CSF, IL-4 and TNF- ⁇ resulted in a six to seven-fold increase in the number of cells obtained after two weeks of culture.
  • the combination of flt3-ligand and c-kit ligand resulted in an additive 12-13-fold increase in absolute cell numbers. This correlated with an 18-fold expansion with either flt3-ligand or c-kit ligand or to a 34-fold expansion with the combination of flt3-ligand and c-kit ligand.
  • Phenotypic analysis of the cells showed that between 60-70% of the cells were HLA-DR + , CD86 + , with 40-50% of the cells expressing CD1 a in all factor combinations examined.
  • the addition of flt3-ligand increased the absolute number of CD1a + cells by 5-fold.
  • c-Kit ligand increased those cells by 6.7-fold and the combination of flt3-ligand and c-kit ligand by 11-fold.
  • Functional analysis of the resultant cells in an MLR revealed that the presence of flt3-ligand or c-kit ligand did not affect the stimulatory capacity of the resultant dendritic cells while increasing the numbers attained.
  • This Example describes a method for using flt3-ligand for dendritic cell expansion.
  • flt3-ligand Prior to cell collection, it may be desirable to mobilize or increase the numbers of circulating PBPC and PBSC. Mobilization can improve PBPC and PBSC collection, and is achievable through the intravenous administration of flt3-ligand or sargramostim (Leukine®, Immunex Corporation, Seattle, Wash.) to the patients prior to collection of such cells.
  • growth factors such as CSF-1, GM-CSF, c-kit ligand, G-CSF, EPO, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, GM-CSF/IL-3 fusion proteins, LIF, FGF and combinations thereof, can be likewise administered in sequence, or in concurrent combination with flt3-ligand.
  • Mobilized or non-mobilized PBPC and PBSC are collected using apheresis procedures known in the art. See, for example, Bishop et al., Blood, vol. 83, No. 2, pp.
  • PBPC and PBSC are collected using conventional devices, for example, a Haemonetics® Model V50 apheresis device (Haemonetics, Braintree, Mass.). Four-hour collections are performed typically no more than five times weekly until approximately 6.5 ⁇ 10 8 mononuclear cells (MNC)/kg patient are collected. Aliquots of collected PBPC and PBSC are assayed for granulocyte-macrophage colony-forming unit (CFU-GM) content by diluting approximately 1:6 with Hank's balanced salt solution without calcium or magnesium (HBSS) and layering over lymphocyte separation medium (Organon Teknika, Durham, N.C.).
  • CFU-GM granulocyte-macrophage colony-forming unit
  • MNC at the interface are collected, washed and resuspended in HBSS.
  • One milliliter aliquots containing approximately 300,000 MNC, modified McCoy's 5A medium, 0.3% agar, 200 U/mL recombinant human GM-CSF, 200 u/mL recombinant human IL-3, and 200 u/mL recombinant human G-CSF are cultured at 37° C. in 5% CO 2 in fully humidified air for 14 days.
  • flt3-ligand or GM-CSF/IL-3 fusion molecules may be added to the cultures.
  • CFU-GM colonies are scored using a dissecting microscope (Ward et al., Exp. Hematol., 16:358 (1988).
  • CFU-GM colonies can be assayed using the CD34/CD33 flow cytometry method of Siena et al., Blood, Vol. 77, No. 2, pp 400-409 (1991), or any other method known in the art.
  • CFU-GM containing cultures are frozen in a controlled rate freezer (e.g., Cryo-Med®, Mt. Clemens, Mich.), then stored in the vapor phase of liquid nitrogen. Ten percent dimethylsulfoxide can be used as a cryoprotectant.
  • CFU-GM containing cultures are thawed and pooled. The thawed cell collection is contacted with flt3-ligand either alone, sequentially or in concurrent combination with other cytokines listed above. Such exposure to flt3-ligand will drive the CFU-GM to dendritic cell lineage. The dendritic cells are reinfused intravenously to the patient.
  • This Example describes a method for using flt3-L to augment anti-tumor immune responses in vivo.
  • Female C57BL/10J (B10) mice (The Jackson Laboratory, Bar Harbor, Me.) were injected with 5 ⁇ 10 5 viable B10.2 fibrosarcoma tumor cells by intradermal injection in a midline ventral position in a total volume of 50 ⁇ l.
  • the fibrosarcoma B10.2 line is of B10 origin and has been described previously, see Lynch et a., Euro. J. Immunol., 21:1403 (1991) incorporated herein by reference.
  • the fibrosarcoma B10.2 line was induced by subcutaneous implantation of a paraffin pellet containing 5 mg of methylcholanthrene.
  • the tumor cell line was maintained in vitro in ⁇ -modified MEM containing 5% FBS, 2 nM L-glutamine, 50 U/ml penicillin and 50 ⁇ g/ml streptomycin.
  • Recombinant human flt3-L (10 ⁇ g/injection) was administered on a daily basis over a 19-day period (unless otherwise noted) by subcutaneous injection in a total volume of 100 ⁇ l.
  • Control mice were similarly injected with a similar volume of buffer containing 100 ng MSA. Tumor growth rates were determined by plotting the tumor size versus time after tumor challenge.
  • Tumor size was calculated as the product of two perpendicular diameters, measured by calipers, and is expressed as the mean tumor size of only those mice bearing a tumor within a particular treatment group. The number of mice bearing tumors compared to the number challenged for each treatment group at the termination of an experiment are shown in the data below.
  • This Example demonstrates the use of flt3-L in combination with a CD40 binding protein to augment anti-tumor immune responses in vivo.
  • C57BL/10J (B10) mice (The Jackson Laboratory, Bar Harbor, Me.) were injected intradermally with 5 ⁇ 10 5 cells of the viable B10.2 fibrosarcoma tumor cell line described in Example 3 above, and the mice were subdivided into four sets each containing eight mice.
  • recombinant human flt3-L 10 ⁇ g/injection/day was administered to each mouse on a daily basis over a 20-day period by subcutaneous injection in a total volume of 100 ⁇ l.
  • each mouse was injected with the same volume and amount of CD40-L each day for 20 days.
  • each mouse was injected with a combination of 10 ⁇ g flt3-L and 10 ⁇ g of CD40-L per day for 20 days.
  • Control mice were similarly injected with a similar volume of buffer containing 100 ng MSA.
  • Tumor growth rates were determined measuring tumor size each week after tumor challenge over a 6 week period. Tumor size was calculated as the product of two perpendicular diameters, measured by calipers, and is expressed as the mean tumor size. Only mice bearing tumors within each group were considered in determining the mean size. The frequency of tumor rejections was also determined and expressed as the number of mice bearing no tumors compared to the number challenged for each treatment group at the termination of an experiment.
  • Table II provides data in the form of mean tumor size in tumor bearing animals, calculated once a week over a 6 week period post challenge.
  • Table III details the percent frequency of tumor rejection for each set of mice over a 6 week period post challenge.
  • the data demonstrate that for tumor bearing mice, the mean tumor size mice in mice treated with flt3-L and flt3-L in combination with CD40-L is comparable and less than the tumor size in tumor bearing control mice.
  • mice receiving the combination therapy experienced significantly higher frequency of tumor rejection than mice receiving flt3-L or CD40-L alone. More specifically, 6 weeks post challenge, 62.5% of the mice receiving the combination therapy experienced complete tumor rejection.
  • mice were injected intradermally with 5 ⁇ 10 5 cells of a very aggressive tumor, the 87 fibrosarcoma tumor cell line (generated by chronic exposure of C3H/HeN(MTV ⁇ ) mice to ultraviolet radiation).
  • the mice were then subdivided into four sets, each containing ten mice.
  • recombinant human flt3-L (10 ⁇ g/injection/day) was administered to each mouse on a daily basis over a 20-day period by subcutaneous injection.
  • each mouse was injected with the same volume and amount of CD40-L each day, beginning at day 7 and continuing to day 20.
  • each mouse received a combination therapy of CD40-L and flt3-L.
  • the combination therapy included 10 ⁇ g/day of flt3-L beginning the day after tumor injection and continuing until day 20 and 10 ⁇ g/day of CD40-L beginning at day 7 and continuing until day 20.
  • Mice in a control group were similarly injected with a similar volume of buffer containing 100 ng MSA. Tumor growth rates were determined by measuring tumor size each week post tumor challenge over a 6 week period. Tumor size was calculated as the product of two perpendicular diameters, measured by calipers, and is expressed as the mean tumor size. Only mice bearing tumors were considered in determining the mean size. The frequency of tumor rejections was also determine and expressed as the number of mice bearing no tumors compared to the number challenged for each treatment group at the termination of an experiment.
  • Table IV provides data in the form of mean tumor size in tumor bearing animals, calculated once a week over a 6 week period post challenge.
  • Table V details the percent frequency of tumor rejection for each set of mice over a 6 week period post challenge.
  • the data demonstrate that for tumor bearing mice, the mean tumor size mice in mice treated with flt3-L in combination with CD40-L is significantly less than the tumor size in tumor bearing control mice and mice bearing tumors in the groups receiving only flt3-L and only CD40-L.
  • mice receiving the combination therapy experienced significantly higher frequency of tumor rejection than mice receiving flt3-L or CD40-L alone.
  • 6 weeks post challenge 50% of the mice receiving the combination therapy experienced complete tumor rejection.
  • 10% of the mice receiving flt3L alone experienced complete tumor rejection and none of the mice receiving CD40-L alone or MSA experienced complete tumor rejection.
  • This Example demonstrates the use of flt3-L in combination with an antibody reactive with 4-1BB to augment anti-tumor immune responses in vivo.
  • C57BL/10J (B10) mice (The Jackson Laboratory, Bar Harbor, Me.) were injected intradermally with 5 ⁇ 10 5 cells of the viable B10.2 fibrosarcoma tumor cell line described in Example 3 above.
  • recombinant human flt3-L (10 ⁇ g/injection/day) was administered to each mouse on a daily basis over a 14-day period by subcutaneous injection in a total volume of 100 ⁇ l.
  • mice In another set of mice each mouse was injected IP with 100 ⁇ g of rat anti mu 4-1BB (clone m6) on days 3 and 6 post tumor challenge. In a third set, each mouse was injected with 100 ⁇ g rat anti mu 4-1BB clone m6 on days 13 and 16. A fourth set of mice were injected with a combination of 10 ⁇ g flt3-L on days 1-14 and 100 ⁇ g of rat anti mu 4-1BB clone m6 on days 13 and 16 post tumor challenge. Control mice were injected with buffer containing 100 ng MSA. Tumor growth rates were determined measuring tumor size each week after tumor challenge over a 5 week period.
  • Tumor size was calculated as the product of two perpendicular diameters, measured by calipers, and is expressed as the mean tumor size in mm 2 . Only mice bearing tumors within each group were considered in determining the mean tumor size. The percent incidence of tumors was also determine and expressed as the number of mice bearing tumors compared to the number challenged for each treatment group at the termination of an experiment.
  • Table VI provides data in the form of mean tumor size in tumor bearing animals, calculated once a week over an 8 week period post challenge.
  • Table VII details the percent incidence of tumors for each set of mice over an 8 week period post challenge.
  • the data demonstrate that for tumor bearing mice, the mean tumor size in mice treated with flt3-L alone and the mean tumor size in mice treated with the anti 4-1BB regimen are similar. However, when flt3-L in combination with an antibody reactive with 4-1BB is administered to mice, mean tumor size in tumor bearing mice is remarkably decreased. Specifically, at 5 weeks post tumor challenge, mice receiving the combination therapy had a mean tumor size of 0, indicating 100% tumor rejection.
  • mice receiving the combination therapy experienced significantly lower incidence of tumors than mice receiving flt3-L or 4-1BB antibody alone. More specifically, at 5 weeks post challenge, all of the mice receiving the combination therapy experienced complete tumor rejection (0% tumor incidence). By contrast, at 5 weeks post challenge, 70% of the mice receiving flt3-L alone had tumors and 50% and 70% of the mice receiving 4-1BB antibody alone had tumors. This data provides evidence that anti-4-1BB synergizes with flt3-L in enhancing immune response.
  • This Example demonstrates the use of flt3-L in combination with interferon alpha to augment anti-tumor immune responses in vivo.
  • the B10.2 fibrosarcoma tumor cell line (described in Example 3 above) was implanted in C57BL/10J (B10) mice on day 0.
  • Control mice were injected with buffer containing 100 ng MSA.
  • Tumor growth rates were determined by measuring tumor size over a 7 week period. Tumor size was calculated as the product of two perpendicular diameters, measured by calipers, and is expressed as the mean tumor size in mm 2 . Only mice bearing tumors within each group were considered in determining the mean tumor size. The percent incidence of tumors was also determined (i.e., the number of mice bearing tumors compared to the number challenged) for each treatment group.
  • Table VIII shows the mean tumor size in tumor bearing animals, and Table IX shows the percent incidence of tumors. This data demonstrates that interferon alpha synergizes with flt3-L in enhancing immune response in the B10.2 tumor model. Most significantly, the tumor rejection rate was 40% for flt3-L alone and 80% for the combination of flt3-L and interferon alpha.

Abstract

Flt3-ligand can be used to generate large numbers of dendritic cells from hematopoietic progenitor and stem cells. Flt3-ligand can be used to augment immune responses in vivo, and expand dendritic cells ex vivo. Such dendritic cells can then be used to present tumor, viral or other antigens to naive T cells, can be useful as vaccine adjuvants. When flt3-L is used and/or administered in combination with other reactive agents, e.g. CD40 binding proteins, 4-1BBL or antibodies reactive with 4-1BB, CD30 ligand antagonists, RANKL, and/or interferon alpha the combination further enhances immune responses and the effectiveness of vaccine adjuvants.

Description

    RELATED APPLICATIONS
  • This application is a Continuation application of pending prior application Ser. No. 10/397,687 (Conf. No. 7724) filed Mar. 25, 2003; which is a Divisional of U.S. application Ser. No. 10/241,927, filed Sep. 11, 2002; which is a Continuation of U.S. application Ser. No. 09/444,027, filed Nov. 19, 1999 (now abandoned); which is a Continuation-In-Part of U.S. application Ser. No. 09/154,903, filed Sep. 17, 1998 (now abandoned); which is a Continuation-In-Part of U.S. application Ser. No. 08/725,540, filed Oct. 3, 1996 (now abandoned); which is a Continuation-In-Part of U.S. application Ser. No. 08/539,142, filed Oct. 4, 1995 (now abandoned); the disclosure of each of which is incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to a dendritic cell stimulatory factor, to methods of enhancing an immune response in vivo, methods of expanding dendritic cells ex vivo, and to preparations of purified dendritic cells, and to dendritic cell populations useful in the manipulation of T cell-mediated and B-cell mediated immune responses.
  • BACKGROUND OF THE INVENTION
  • The objective of vaccination is to provide effective immunity by establishing adequate levels of antibody and a primed population of cells that can rapidly expand on renewed contact with antigen. The first contact with antigen during vaccination must not be injurious to the recipient and thus usually consists of pathogenically-deficient antigen.
  • A frequent difficulty with active immunization protocols is that the vaccine antigen does not possess sufficient immunogenicity to promote a strong immune response, and therefore a sufficient level of protection against subsequent challenge by the same antigen. In addition, certain antigens may elicit only weak cell-mediated or antibody responses. For many antigens, both a strong humoral response and a strong cell-mediated response is desirable.
  • For decades, researchers have experimented with diverse compounds to increase the immunogenicity of vaccines. Immunopotentiators, also known as adjuvants, of vaccines are compositions of matter that facilitate a strong immune response to a vaccine. In addition, the relatively weak immunogenicity of certain novel recombinant antigens has required adjuvants to be more potent. Vaccine adjuvants have different modes of action, affecting the immune response both quantitatively and qualitatively. Such modes of action can be by mobilizing T cells, acting as depots and altering lymphocyte circulation so that these cells remain localized in draining lymph nodes. They may also serve to focus antigen at the site of immunization, thereby allowing antigen specific T cells and B cells to interact more efficiently with antigen-presenting cells. They may also stimulate proliferation and differentiation of T cells and have effects on B cells, such as enhancing the production of different Ig isotypes. Further, adjuvants may stimulate and affect the behavior of antigen-presenting cells, particularly dendritic cells and macrophages, rendering them more effective for presenting antigen to T cells and B cells.
  • Dendritic cells are a rare and heterogeneous cell population with distinctive morphology and a widespread tissue distribution. A discussion of the dendritic cell system and its role in immunogenicity is provided by Steinman, R. M., Annu. Rev. Immunol., 9:271-296 (1991), incorporated herein by reference. Dendritic cells display an unusual cell surface phenotype and can be characterized by the presence of the cell surface markers CD1, CD4, CD86, CD11c, DEC-205, CD40 or HLA-DR, and the absence of CD14 and other lineage markers. Dendritic cells have a high capacity for sensitizing MHC-restricted T cells and provide an effective pathway for presenting antigens to T cells in situ, both self-antigens during T cell development and foreign antigens during immunity. Thus, there is growing interest in using dendritic cells ex vivo as tumor or infectious disease vaccine adjuvants. See, for example, Romani, et al., J. Exp. Med, 180:83 (1994). The use of dendritic cells as immunostimulatory agents has been limited due to the low frequency of dendritic cells in peripheral blood, the limited accessibility to lymphoid organs and the dendritic cells' terminal state of differentiation. Dendritic cells originate from CD34+ bone marrow progenitors, and the proliferation and maturation of dendritic cells can be enhanced by the cytokines GM-CSF (sargramostim, Leukine®, Immunex Corporation, Seattle, Washington), TNF-α, c-kit ligand (also known as stem cell factor (SCF), steel factor (SF), or mast cell growth factor (MGF)) and interleukin-4. Therefore, an agent that stimulated the generation of large numbers of functionally mature dendritic cells in vivo or in vitro would be of wide importance.
  • SUMMARY OF THE INVENTION
  • Flt3-ligand (“flt3-ligand,” “flt3-L,” “Flt3-L”) is known to affect hematopoietic stem and progenitor cells. It was surprisingly found that flt3-ligand can also potently stimulate the generation of downstream or intermediate, cells such as myeloid precursor cells, monocytic cells, macrophages, B cells, and dendritic cells from CD34+ bone marrow progenitors and stem cells. The present invention pertains to a method of mobilizing dendritic cells in vivo, expanding dendritic cells ex vivo and to preparations of dendritic cells. The preparation of purified dendritic cells according to the invention would potentially find use as vaccine adjuvants. Also included within the embodiments of the invention is a method of preparing antigen-specific T cells using the dendritic cells mobilized with flt3-ligand.
  • The invention provides for the use of an effective amount of flt3-ligand to increase or mobilize the numbers of intermediate cells in vivo, for example, in the patient's peripheral blood or spleen. While the invention relates to the generation of large numbers of such downstream and intermediate cells (e.g., myeloid cells, monocytic cells and macrophages) from CD34+ cells using flt3-ligand, the focus is particularly on dendritic cells. By increasing the quantity of the patient's dendritic cells, such cells may themselves be used to present antigen to T cells. For example, the antigen may be one that already exists within the patient, such as a tumor antigen, or a bacterial or viral antigen. Flt3-L may be used, therefore, to increase the numbers of dendritic cells in vivo to boost a patient's immune response against existing antigens. The invention further provides for using combination therapy to enhance a patient's immune response. Such combination therapy includes administering flt3-ligand and one or more therapeutic reagents in amounts sufficiently effective to upregulate the patient's immune response. Alternatively, flt3-ligand may be administered prior to, concurrently with or subsequent to administration of an antigen to a patient for immunization purposes. Thus, as a vaccine adjuvant, flt3-ligand can generate large quantities of dendritic cells in vivo to more effectively present the antigen. The overall response is a stronger and improved immune response and more effective immunization to the antigen.
  • The invention also provides a method of generating large quantities of dendritic cells ex vivo. Following collection of the patient's CD34+ hematopoietic progenitors and stem cells, flt3-ligand can be used to expand such cells in vitro (also known as ex vivo expansion) and to drive such CD34+ cells to differentiate into dendritic cells of the lymphoid or myeloid lineage. The resulting collection of dendritic cells can be administered to a patient to provide a stronger and improved immune response to an antigen. Alternatively, the resulting dendritic cells find use as a vaccine adjuvant and can be administered prior to, concurrently with or subsequent to antigen administration.
  • The invention also provides a method of generating large quantities of antigen-presenting dendritic cells ex vivo. Following collection of the patient's CD34+ hematopoietic progenitors and stem cells, flt3-ligand can be used to expand such cells in vitro and to drive such CD34+ cells to differentiate into dendritic cells. The resulting collection of dendritic cells is exposed to an antigen and allowed to process and present the antigen in vitro (this procedure is sometimes referred to in the art as “antigen-pulsing”). An alternate method for preparing dendritic cells that present antigen is to transfect the dendritic cells with a gene encoding an antigen-specific polypeptide. Once the dendritic cells express the antigen, the antigen-presenting dendritic cells can be administered to a patient.
  • The invention also provides for the ex vivo preparation of antigen-specific T cells. Following the procedures described above for preparing large numbers of antigen-presenting dendritic cells ex vivo, the collected antigen-presenting dendritic cells are used to generate antigen-specific T cells from naive T cells that have been collected from a patient. After the antigen has been adequately presented to the T cells generated, the antigen-specific T cells can be administered to the patient.
  • The invention also provides a method of augmenting an immune response in a patient that has an infectious disease wherein the method comprises the step of administering an amount of flt3-ligand sufficient to increase the patient's number of dendritic cells. Embodiments of methods for augmenting an immune response include administering flt3-ligand in combination therapies with additional active compounds, including but not limited to CD40 binding proteins, 4-1BB-L, antibodies to 4-1BB, interferon alpha, RANKL, a CD30 ligand antagonist, and combinations thereof.
  • The invention also provides a method of augmenting an immune response in a patient that has a cancerous or neoplastic disease wherein the method comprises the step of administering an amount of flt3-ligand sufficient to increase the patient's number of dendritic cells. Embodiments of methods for augmenting an immune response include administering flt3-ligand in combination therapies with additional active compounds, including but not limited to CD40 binding proteins, 4-1BB-L, antibodies to 4-1BB, interferon alpha, RANKL, a CD30 ligand antagonist, and combinations thereof. Such method provides a means to enhance the patient's tumor-specific immune response.
  • A method for enhancing a patient's autoimmune tolerance wherein the method comprises the step of administering an amount of flt3-ligand sufficient to increase the patient's number of dendritic cells. Further included are methods for promoting survival of grafts and transplanted tissues and organs.
  • The methods of the invention can further comprise the use of an effective amount of a cytokine in sequential or concurrent combination with flt3-ligand. Such cytokines include, but are not limited to, interleukins (“ILs”) IL-3 and IL-4, a colony stimulating factor (“CSF”) selected from the group consisting of granulocyte macrophage colony stimulating factor (“GM-CSF”) or GM-CSF/IL-3 fusions, or other cytokines such as TNF-α, CD40 binding proteins (e.g. CD40-L), 4-1BB antagonists (e.g., antibodies immunoreactive with 4-1BB and 4-IBB-L), interferon alpha, RANKL, CD30 ligand antagonists (e.g., CD30 ligand monoclonal antibodies and soluble CD30-Fc fusion polypeptides), and c-kit ligand.
  • The invention further includes a dendritic cell expansion medium comprising cell growth medium, autologous serum, and flt3-ligand alone or in combination with a cytokine from the group listed above.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The invention is directed to the use of flt3-ligand to generate large numbers of intermediate cell types from CD34+ hematopoietic progenitor cells and stem cells. Such intermediate cell types include myeloid cells, monocytic cells, macrophages and dendritic cells. The large numbers of these intermediate cell types are not naturally found in vivo and can be generated by administering flt3-ligand. Such enhancement in overall cell number can augment the immune response to antigen in the host. Another embodiment of the invention is the isolation and use of such intermediate cell types as antigen-presenting cells or the use as vaccine adjuvants. The invention, while particularly focused on the embodiment concerning dendritic cells, is also applicable to myeloid, monocytic and macrophage cell types.
  • As used herein, the term “flt3-ligand” refers to a genus of polypeptides that are described in U.S. Pat. No. 5,554,512, EP 0627487 A2 and in WO 94/28391, both incorporated herein by reference. A human flt3-ligand cDNA was deposited with the American Type Culture Collection, Rockville, Md., USA (ATCC) on Aug. 6, 1993 and assigned accession number ATCC 69382. The deposit was made under the terms of the Budapest Treaty. Flt3-L can be made according to the methods described in the documents cited above.
  • As described in U.S. Pat. No. 5,554,512 at column 5, line 15, the term “flt3-L” encompasses proteins having the amino acid sequence 1 to 235 of SEQ ID NO:2, as well as those proteins having a high degree of similarity or a high degree of identity with the amino acid sequence 1 to 235 of SEQ ID NO:2, and which proteins are biologically active and bind the flt3 receptor. In addition, the term refers to biologically active gene products of the DNA of SEQ ID NO:1. Further encompassed by the term “flt3-L” are the membrane-bound proteins (which include an intracellular region, a membrane region, and an extracellular region), and soluble or truncated proteins which comprise primarily the extracellular portion of the protein, retain biological activity and are capable of being secreted. Specific examples of such soluble proteins are those comprising the sequence of amino acids 28-160 of SEQ ID NO:2.
  • A “flt3-L variant” as referred to herein, means a polypeptide substantially homologous to native flt3-L, but which has an amino acid sequence different from that of native flt3-L (human, murine or other mammalian species) because of one or more deletions, insertions or substitutions. The variant amino acid sequence preferably is at least 80% identical to a native flt3-L amino acid sequence, most preferably at least 90% identical. The percent identity may be determined, for example, by comparing sequence information using the GAP computer program, version 6.0 described by Devereux et al. (Nucl. Acids Res. 12:387, 1984) and available from the University of Wisconsin Genetics Computer Group (UWGCG). The GAP program utilizes the alignment method of Needleman and Wunsch (J. Mol. Biol. 48:443, 1970), as revised by Smith and Waterman (Adv. Appl. Math 2:482, 1981). The preferred default parameters for the GAP program include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non-identities) for nucleotides, and the weighted comparison matrix of Gribskov and Burgess, Nucl. Acids Res. 14:6745, 1986, as described by Schwartz and Dayhoff, eds., Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, pp. 353-358, 1979; (2) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap; and (3) no penalty for end gaps. Variants may comprise conservatively substituted sequences, meaning that a given amino acid residue is replaced by a residue having similar physiochemical characteristics. Examples of conservative substitutions include substitution of one aliphatic residue for another, such as Ile, Val, Leu, or Ala for one another, or substitutions of one polar residue for another, such as between Lys and Arg; Glu and Asp; or Gln and Asn. Other such conservative substitutions, for example, substitutions of entire regions having similar hydrophobicity characteristics, are well known. Naturally occurring flt3-L variants are also encompassed by the invention. Examples of such variants are proteins that result from alternate mRNA splicing events or from proteolytic cleavage of the flt3-L protein, wherein the flt3-L binding property is retained. Alternate splicing of mRNA may yield a truncated but biologically active flt3-L protein, such as a naturally occurring soluble form of the protein, for example. Variations attributable to proteolysis include, for example, differences in the N- or C-termini upon expression in different types of host cells, due to proteolytic removal of one or more terminal amino acids from the flt3-L protein (generally from 1-5 terminal amino acids).
  • As described in U.S. Pat. No. 5,554,512 at column 8, line 7, an aspect of the invention is soluble flt3-L polypeptides. Soluble flt3-L polypeptides comprise all or part of the extracellular domain of a native flt3-L but lack the transmembrane region that would cause retention of the polypeptide on a cell membrane. Soluble flt3-L polypeptides advantageously comprise the native (or a heterologous) signal peptide when initially synthesized to promote secretion, but the signal peptide is cleaved upon secretion of flt3-L from the cell. Soluble flt3-L polypeptides encompassed by the invention retain the ability to bind the flt3 receptor. Indeed, soluble flt3-L may also include part of the transmembrane region or part of the cytoplasmic domain or other sequences, provided that the soluble flt3-L protein can be secreted.
  • Soluble flt3-L may be identified (and distinguished from its non-soluble membrane-bound counterparts) by separating intact cells which express the desired protein from the culture medium, e.g., by centrifugation, and assaying the medium (supernatant) for the presence of the desired protein. The presence of flt3-L in the medium indicates that the protein was secreted from the cells and thus is a soluble form of the desired protein.
  • Soluble forms of flt3-L possess many advantages over the native bound flt3-L protein. Purification of the proteins from recombinant host cells is feasible, since the soluble proteins are secreted from the cells. Further, soluble proteins are generally more suitable for intravenous administration.
  • Examples of soluble flt3-L polypeptides include those comprising a substantial portion of the extracellular domain of a native flt3-L protein. Such soluble mammalian flt3-L proteins comprise amino acids 28 through 182 of SEQ ID NO:2. In addition, truncated soluble flt3-L proteins comprising less than the entire extracellular domain are included in the invention. Such truncated soluble proteins are represented by the sequence comprising amino acids 28-160 of SEQ ID NO:2. When initially expressed within a host cell, soluble flt3-L may additionally comprise one of the heterologous signal peptides described below that is functional within the host cells employed. Alternatively, the protein may comprise the native signal peptide, such that the mammalian flt3-L comprises 1 through 182 of SEQ ID NO:2. Isolated DNA sequences encoding soluble flt3-L proteins are encompassed by the invention.
  • The term “IL-3” refers to a genus of interleukin-3 polypeptides as described in U.S. Pat. No. 5,108,910, incorporated herein by reference. Such polypeptides include analogs that have amino acid sequences that are substantially similar to the native human interleukin-3 amino acid sequences disclosed, for example, in EP publ. Nos. 275,598 and 282,185, each incorporated herein by reference. The term “IL-3” also includes analogs and alleles of IL-3 molecules that exhibit at least some of the biological activity in common with native human IL-3. Exemplary analogs of IL-3 are disclosed in EP Publ. No. 282,185. Other forms of IL-3 include human IL-3[Pro8Asp15Asp70], human IL-3[Ser8Asp15Asp70] and human IL-3[Ser8]. A DNA sequence encoding human IL-3 protein suitable for use in the invention is publicly available from the American Type Culture Collection (ATCC) under accession number ATCC 67747. The nomenclature used herein with respect to amino acid sequences in brackets designates which amino acids differ from the native human form. For example, human IL-3[Ser8Asp15Asp70] refers to a human IL-3 protein in which amino acid 8 has been changed to a serine residue, amino acid 15 has been changed to an aspartic acid residue and the amino acid 70 has been changed to an aspartic acid residue.
  • The term “IL-4” refers to a polypeptide as described in Mosley et al., Cell 59:335 (1989), Idzerda et al., J. Exp. Med. 171:861 (1990) and Galizzi et al., Intl. Immunol. 2:669 (1990), each of which is incorporated herein by reference. Such IL-4 polypeptide includes analogs that have an amino acid sequence that is substantially similar to the native human IL-4 amino acid sequences described in Mosley et al., Idzerda et al., and Galizzi et al. and which are biologically active in that they are capable of binding to a IL-4 receptor, transducing a biological signal initiated by binding IL-4 receptor, or cross-reacting with anti-IL-4 antibodies. The term “IL-4” also includes analogs of native human IL-4 molecules sufficient to retain biological activity of native human IL-4.
  • As used herein, “GM-CSF” refers to a genus of proteins as described in U.S. Pat. Nos. 5,108,910, and 5,229,496 each of which is incorporated herein by reference. Such proteins include analogs that have an amino acid sequence that is substantially similar to native human GM-CSF amino acid sequences (e.g., as publicly available ATCC 53157 or ATCC 39900), and which are biologically active in that they are capable of binding to a GM-CSF receptor, transducing a biological signal initiated by binding GM-CSF receptor, or cross-reacting with anti-GM-CSF antibodies. Amino acid sequences are disclosed, for example in Anderson, et al., Proc. Natl. Acad. Sci., USA 82:6250 (1985). Commercially available GM-CSF (sargramostim, Leukine®) is obtainable from Immunex Corp., Seattle, Wash.). The term “GM-CSF” also includes analogs of the native human GM-CSF molecules described in U.S. Pat. Nos. 5,108,910, and 5,229,496 sufficient to retain biological activity of native human GM-CSF. Exemplary analogs of GM-CSF include, for example, those described in EP Publ. No. 212914 and WO 89/03881, each of which is incorporated herein by reference. Other analogs of GM-CSF also may be used to construct fusion proteins with IL-3. A DNA sequence encoding a particularly preferred GM-CSF protein having potential glycosylation sites removed is publicly available from the ATCC under accession numbers ATCC 67231.
  • The term “GM-CSF/IL-3 fusion protein” means a C-terminal to N-terminal fusion of GM-CSF and IL-3. The fusion proteins are known and are described in U.S. Pat. Nos. 5,199,942, 5,108,910 and 5,073,627, each of which is incorporated herein by reference. A preferred fusion protein is PIXY321 as described in U.S. Pat. No. 5,199,942.
  • The term “c-kit ligand” also known as Mast Cell Growth Factor (MGF), Steel Factor or Stem Cell Factor (SCF), refers to a polypeptide described in EP 423,980, which is incorporated herein by reference, and that claims priority from U.S. patent application Ser. No. 589,701, filed Oct. 1, 1990. Such c-kit ligand polypeptide includes analogs that have an amino acid sequence that is substantially similar to the native human c-kit ligand amino acid sequences described in EP 423,980 and which are biologically active in that they are capable of binding to a c-kit receptor, transducing a biological signal initiated by binding c-kit receptor, or cross-reacting with anti-c-kit ligand antibodies. The term “c-kit ligand” also includes analogs of native human c-kit ligand molecules sufficient to retain biological activity of native human c-kit ligand.
  • The term “CD40 binding protein” refers to polypeptides that bind CD40, including but not limited to CD40-L and antibodies immunoreactive with CD40, as described in PCT publications WO 93/08207 and WO 96/40918 each of which is incorporated herein by reference. Such CD40 binding proteins include analogs that have an amino acid sequence that is substantially similar to the native human CD40-L amino acid sequences described in the PCT publications, and which are biologically active in that they are capable of binding to CD40, transducing a biological signal initiated by binding CD40, or cross-reacting with anti-CD40 antibodies. The term CD40 binding proteins also includes analogs of native human CD40-L and antibodies reactive with CD40 that are sufficiently homologous to native molecules so as to retain biological activity of native CD40 binding proteins.
  • The terms “4-1BB-L” and “antibody to 4-1BB” refer to molecules that are described in U.S. Pat. No.5,674,704 and Alderson et al. Eur. J. Immunol. 24:2219-2227, 1994, which are incorporated herein by reference. 4-1BB-L includes analogs that have an amino acid sequence that is substantially similar to the native 4-1BB-L amino acid sequence described in the above mentioned publication and which are biologically active in that they are capable of binding to 4-1BB or transducing a biological signal initiated by binding 4-1BB, such as inducing a proliferative response in stimulated primary T cells. The terms 4-1BB-L and antibodies to 4-1BB also includes analogs of native 4-1BB-L and analogs of antibodies reactive with 4-1BB that are sufficiently homologous to the native compounds so as to retain biological activity of 4-1BB-L and the described antibodies.
  • The term “interferon alpha” refers to naturally occurring forms of interferon alpha (leukocyte interferon) as isolated from cells, cell lines, tissue, and other sources, as well as recombinant forms, consensus forms, analogs, and variant forms that exhibit interferon alpha biological activity. Hybrid “interferon alpha A/D,” also called “Universal Type I Interferon,” has been described by Fisher et al., Biochem Biophys. Res. Commun. 119(1):108 (1984), incorporated herein by reference. Interferon alpha polypeptides have been described in, inter alia, Pestka et al., Ann. Rev. Biochem. 56:727 (1987), Goeddel et al., Nature 290:20 (1980), Petska et al., in Human Cylokines, Blackwell Scientific Publications 1-16 (1992), Petska, Semin. Oncol. 24(3):S9-4 (197), U.S. Pat. No. 4,414,150, and WO 96/11953, all of which are incorporated herein by reference.
  • The term “CD30 ligand antagonist” refers to any molecule that interferes with the binding between CD30 ligand and the CD30 receptor. CD30 ligand is described, inter alia, in U.S. Pat. No. 5,480,981 which is incorporated herein by reference. CD30 ligand antagonists include, but are not limited to, CD30 ligand antibodies (including CD30 ligand monoclonal antibodies) and soluble CD30 polypeptides and analogs thereof (including soluble CD30-Fc fusion polypeptides). CD30 ligand antagonists may also be antisense nucleic acids, ribozymes, muteins, aptamers, and small molecules that interfere with the CD30-CD30 ligand interaction.
  • “RANK ligand” (“RANKL”) polypeptides are described in, inter alia, WO 98/28426; “TRAIL” polypeptides are described in, inter alia, U.S. Pat. No. 5,763,223; “Tek” (also called Tie2, ork) polypeptides are described in, inter alia, U.S. Pat. No. 5,447,860. These publications are incorporated herein by reference.
  • The term “adjuvant” refers to a substance that enhances, augments or potentiates the host's immune response to a vaccine antigen.
  • The term “antagonist” includes various classes of molecules that are capable of interfering with a specified biological interaction and/or activity. Antagonists include, but are not limited to, antibodies, soluble forms of a target polypeptide, antisense nucleic acids, ribozymes, muteins, aptamers, and small molecules.
  • The procedure for “ex vivo expansion” of hematopoietic stem and progenitor cells is described in U.S. Pat. No. 5,199,942, incorporated herein by reference. Briefly, the term means a method comprising: (1) collecting CD34+ hematopoietic stem and progenitor cells from a patient from peripheral blood harvest or bone marrow explants; and (2) expanding such cells ex vivo . In addition to the cellular growth factors described in U.S. Pat. No. 5,199,942, other factors such as flt3-ligand, IL-1, IL-3, c-kit ligand, can be used.
  • The term “immunogenicity” means relative effectiveness of an immunogen or antigen to induce an immune response.
  • The term “substantially similar” means a variant amino acid sequence preferably that is at least 80% identical to a native amino acid sequence, most preferably at least 90% identical. Percent identity may be determined by visual inspection. Percent identity may be determined using the alignment method of Needleman and Wunsch (J. Mol. Biol. 48:443, 1970) as revised by Smith and Waterman (Adv. Appl. Math 2:482, 1981). Preferably, percent identity is determined by using a computer program, for example, the GAP computer program version 10.x available from the Genetics Computer Group (GCG; Madison, Wis., see also Devereux et al., Nucl. Acids Res. 12:387, 1984). The preferred default parameters for the GAP program include: (1) a comparison matrix containing a value of 1 for identities and 0 for non-identities for nucleotides, and the weighted comparison matrix of Gribskov and Burgess, Nucl. Acids Res. 14:6745, 1986, as described by Schwartz and Dayhoff, eds., Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, pp. 353-358, 1979 for amino acids; (2) a penalty of 30 (amino acids) or 50 (nucleotides) for each gap and an additional 1 (amino acids) or 3 (nucleotides) penalty for each symbol in each gap; (3) no penalty for end gaps; and (4) no maximum penalty for long gaps.
  • Variants may comprise conservatively substituted sequences, meaning that a given amino acid residue is replaced by a residue having similar physiochemical characteristics. Examples of conservative substitutions include substitution of one aliphatic residue for another, such as Ile, Val, Leu, or Ala for one another, or substitutions of one polar residue for another, such as between Lys and Arg; Glu and Asp; or Gln and Asn. Other such conservative substitutions, for example, substitutions of entire regions having similar hydrophobicity characteristics, are well known. Naturally occurring variants are also encompassed by the invention. Examples of such variants are proteins that result from alternate mRNA splicing events or from proteolytic cleavage of the native protein, wherein the native biological property is retained.
  • As used herein, “vaccine” means an organism or material that contains an antigen in an innocuous form. The vaccine is designed to trigger an immunoprotective response. The vaccine may be recombinant or non-recombinant. When inoculated into a non-immune host, the vaccine will provoke active immunity to the organism or material, but will not cause disease. Vaccines may take the form, for example, of a toxoid, which is defined as a toxin that has been detoxified but that still retains its major immunogenic determinants; or a killed organism, such as typhoid, cholera and poliomyelitis; or attenuated organisms, that are the live, but non-virulent, forms of pathogens, or it may be antigen encoded by such organism, or it may be a live tumor cell or an antigen present on a tumor cell.
  • A variety of cell selection techniques are known for identifying and separating CD34+ hematopoietic stem or progenitor cells from a population of cells. Methods and materials for identifying and selecting such cell types are known. For example, monoclonal antibodies can be used to bind to a marker protein or surface antigen protein found on stem or progenitor cells. Such markers or cell surface antigens for hematopoietic stem cells include CD34 and Thy-1. In one method, antibodies are fixed to a surface, for example, glass beads, and contacted with a mixture of cells suspected of containing stem cells. This permits the antibodies to bind and secure the stem cells to the glass beads. Alternatively, the antibodies can be incubated with the cell mixture and the resulting combination contacted with a surface having an affinity for the antibody-cell complex. Undesired cells and cell matter are removed providing a relatively pure population of stem cells. Stem or progenitor cells having the CD34 marker constitute only about 1% to 3% of the mononuclear cells in the bone marrow. The amount of CD34+ stem or progenitor cells in the peripheral blood is approximately 10- to 100-fold less than in bone marrow.
  • With regard to the particular aspects of the invention, choosing suitable stem or progenitor cell selection means will depend on the desired phenotype of the cell to be isolated. Hematopoietic stem cells are selectable by virtue of their physical characteristics, such as expressing the membrane-bound flt3 receptor, or having the following cellular markers: CD34 or Thy-1. Monoclonal antibodies that recognize any of these antigens have been described in U.S. Pat. No. 4,714,680 (anti-My-10) incorporated herein by reference, anti-CD34 is commercially available from Becton Dickinson, Franklin Lakes, N.J.), and anti-Thy-1 monoclonal antibodies can be readily generated using the methods described by Dalchau et al., J. Exp. Med. 149:576 (1979), incorporated herein by reference. A flt3 receptor binding protein also may be used, such as anti-flt3 monoclonal antibodies or the flt3-ligand. The cell binding protein is brought into contact with the collected cell mixture and the combination is allowed to incubate for a period of time sufficient to permit the binding of the desired cell to the cell binding protein.
  • An alternative means of selecting the quiescent stem cells is to induce cell death in the dividing, more lineage-committed, cell types using an antimetabolite such as 5-fluorouracil (5-FU) or an alkylating agent such as 4-hydroxycyclophosphamide (4-HC). The non-quiescent cells are stimulated to proliferate and differentiate by the addition of growth factors that have little or no effect on the stem cells, causing the non-stem cells to proliferate and differentiate and making them more vulnerable to the cytotoxic effects of 5-FU or 4-HC. See Berardi et al., Science, 267:104 (1995), which is incorporated herein by reference.
  • Isolation of the hematopoietic stem or progenitor cells can be performed by using, for example, affinity chromatography, antibody-coated magnetic beads, or antibodies fixed to a solid matrix, such as glass beads, flasks, etc. Antibodies that recognize a stem or progenitor cell surface marker can be fused or conjugated to other chemical moieties such as biotin—which can be removed with an avidin or a streptavidin moiety secured to a solid support; fluorochromes useful in fluorescence activated cell sorting (FACS), or the like. Preferably, isolation is accomplished by an immunoaffinity column. Immunoaffinity columns can take any form, but usually comprise a packed bed reactor. The packed bed in these bioreactors is preferably made of a porous material having a substantially uniform coating of a substrate. The porous material, which provides a high surface area-to-volume ratio, allows for the cell mixture to flow over a large contact area while not impeding the flow of cells out of the bed. Typical substrates include avidin and streptavidin, while other conventional substrates can be used. The substrate should, either by its own properties, or by the addition of a chemical moiety, display high-affinity for a moiety found on the cell-binding protein such as a monoclonal antibody. The monoclonal antibodies recognize a cell surface antigen on the cells to be separated, and are typically further modified to present a biotin moiety. It is well-known that biotin has a high affinity for avidin, and the affinity of these substances thereby removably secures the monoclonal antibody to the surface of the packed bed. Such columns are well known in the art, see Berenson, et al., J. Cell Biochem., 10D:239 (1986). The column is washed with a PBS solution to remove unbound material. Target cells can be released from the beads using conventional methods. Immunoaffinity columns of the type described above that utilize biotinylated anti-CD34 monoclonal antibodies secured to an avidin-coated packed bed are described for example, in PCT Publ. No. WO 93/08268. A variation of this method utilizes cell binding proteins, such as the monoclonal antibodies or flt3-ligand as described above, removably-secured to a fixed surface in the isolating means. The bound cell binding protein then is contacted with the collected cell mixture and allowed to incubate for a period of time sufficient to permit isolation of the desired cells.
  • Alternatively, the monoclonal antibodies that recognize the cell surface antigens can be labeled with a fluorescent label, e.g., chromophore or fluorophore, and separated by cell sorting according to the presence of absence or the amount of labeled product.
  • The collected CD34+ cells are then exposed to either flt3-ligand alone or flt3-ligand in concurrent or sequential combination with one or more of the following cytokines: GM-CSF, TNF-α, IL-3, IL-4, c-kit-ligand, CD40-L, 4-IBB-L or GM-CSF/IL-3 fusion proteins. CD34+ cells then are allowed to differentiate and commit to cells of the dendritic lineage. The dendritic cells are collected and can either be (a) administered to a patient in order to augment the immune system and T-cell mediated or B-cell mediated immune responses to antigen, (b) exposed to an antigen prior to administration of the dendritic cells into a patient, (c) transfected with a gene encoding an antigen-specific polypeptide or (d) exposed to an antigen and then allowed to process and present the antigen, ex vivo, to T-cells collected from the patient followed by administration of the antigen-specific T-cells to the patient.
  • More specifically, the invention provides for the use of an effective amount of flt3-ligand to increase or mobilize dendritic cells in vivo, for example, in the patient's peripheral blood or spleen. By increasing the quantity of the patient's dendritic cells, such cells may themselves be used to present antigen to T cells. For example, the antigen may be one that already exists within the patient, such as a tumor antigen, or a bacterial or viral antigen. Flt3-L may be used, therefore, to boost the patient's lymphocyte-mediated (e.g., T cell and B cell mediated) or myeloid-mediated immune response to the already present antigens thus potentially enabling a more effective antigen-presentation to the patient's T cells.
  • Further, flt3-L may be used in combination therapies with one or more additional agents to enhance an immune response against tumor, viral or bacterial antigens. For example, CD40 binding proteins, which enhance the ability of dendritic cells to process and present antigens to effector T cells can be administered in combination with flt3-L to dramatically enhance an immune response. Such immune responses can include responses against viral or bacterial antigens that are responsible for infectious diseases and immune responses to tumor antigens. As described in Example 4, a surprising synergy between a CD40 binding protein and flt3-L has been discovered for their combined ability to enhance anti-tumor responses. Represen-tative CD40 binding proteins useful in combination therapy with flt3-L include CD40-L and antibodies immunoreactive with CD40 which are described in PCT publications WO 93/08207 and WO 96/40918.
  • Additionally, 4-1BB-L and antibodies reactive with 4-1BB, both of which are T-cell co-activation factors, can be administered in combination with flt3-L to dramatically enhance immune responses. 4-1BB-L and antibodies reactive with 4-1BB can be used in combination therapies to enhance immune responses to viral antigens and bacterial antigens responsible for infectious diseases and to enhance immune responses to tumor antigens. More particularly, as described in Example 5, when used in a combination therapy there is a surprising synergy between flt3-L and a 4-1BB-L or antibodies to 4-1BB for anti-tumor immune responses. 4-IBB-L and antibodies reactive with 4-1BB are described in U.S. Pat. No. 5,674,704. The surprising synergy in the above described combination therapies for their ability to dramatically enhance anti-tumor immune responses suggests that stimulating more than one mechanism or more than one cell population is a promising approach to cancer treatment.
  • Additionally, interferon alpha, RANKL, or a CD30 ligand antagonist can be administered in combination with flt3-L to dramatically enhance immune responses. As described in Example 6, when used in a combination therapy there is a surprising synergy between flt3-L and interferon alpha for anti-tumor immune responses.
  • Other molecules that may be used in combination with flt3-L according to the present invention include IL-2, IL-12, IL-15, TRAIL, Fas ligand, VEGF antagonists, Tek antagonists, molecules that enhance dendritic cell function, survival, or expansion, molecules that enhance T cell activation or differentiation, molecules that enhance dendritic cell migration including various chemokines, molecules that increase the availability of target cell antigens, such as apoptotic factors and molecules that enhance MHC Class I presentation including the various interferons, angiogenesis inhibitors, inhibitors of immunosuppressive molecules released by tumors including IL-10, VEGF, and TGF-β, and tumor-specific antibodies including toxin- or radio-labeled antibodies.
  • In addition to stimulating an immune response to an antigen that already exists within the patient, flt3-ligand may be administered prior to, concurrently with or subsequent to administration of an antigen to a patient for immunization purposes. Thus, as a vaccine adjuvant, flt3-ligand can generate large quantities of dendritic cells in vivo to more effectively present the antigen. The overall response is a stronger and improved immune response and more effective immunization to the antigen. Further, flt3-L may be administered as a vaccine adjuvant in combination with additional active compounds prior to, concurrently with or subsequent to administration of an antigen to a patient for immunization purposes to enhance an immune response against tumor, viral or bacterial antigens. For example, CD40 binding proteins, such as CD40-L and antibodies to CD40 which enhance the ability of dendritic cells to present antigens to T cells can be administered in combination with flt3-L to dramatically enhance an immune response. Similarly, 4-1BB-L, antibodies reactive with 4-1BB, interferon alpha, RANKL, or CD30 ligand antagonists can be administered in combination with flt3-L to enhance an immune response and provide more effective immunization to the antigen.
  • The systemic administration of flt3-ligand not only is effective as a vaccine adjuvant, but as discussed supra., is effective in augmenting an immune response against previously existing antigens. For example, the inventors have shown that flt3-ligand administration to tumor-bearing mice results in at least a significant decrease in the growth rate of the tumor, and can result in tumor regression in a large proportion of the mice. The data are presented in more detail in Example 3. Flt3-L therefore is an important cytokine in the generation of an effective immune response in vivo against antigen. Data in Examples 4-6 demonstrate that when used in combination therapy with additional agents, flt3-L can provide a dramatically enhanced immune response in vivo against antigen.
  • Because of its ability to generate dendritic cells, flt3-ligand also finds use in promoting the survival of transplanted tissue or organs. When allogeneic organs or other tissue is transplanted into a host they can transfer stem cells, immature dendritic cells, and mature dendritic cells from the donor. These cells are called passenger cells and such cells can graft into the hematopoietic system of the host. Additionally, stem cells, immature dendritic cells, and mature dendritic cells from the host may graft to the donor organ or tissue.
  • It is possible then to establish a tolerance between the graft and the host since the immature dendritic cells from the host and donor tissue interact with T-cells from the “other side.” Such interaction may include the deletion of T-cells that recognize the major histocompatability complex (MHC) molecules that the dendritic cells express. In this way, the donor T cells are “screened” so that they fail to recognize and react against the host (i.e., no graft versus host disease) and the host T-cells are screened so that they fail to recognize and react against the graft (i.e., no graft rejection). Thus, a mutual tolerance can be achieved, and the graft is accepted. Administration of flt3-ligand to the host or donor prior to transplantation would generate increased numbers of dendritic cells in such host or donor and permit increased tolerance and survival of the graft.
  • For the growth and culture of dendritic cells, a variety of growth and culture media can be used, and the composition of such media can be readily determined by a person having ordinary skill in the art. Suitable growth media are solutions containing nutrients or metabolic additives, and include those that are serum-depleted or serum-based. Representative examples of growth media are RPMI, TC 199, Iscoves modified Dulbecco's medium (Iscove, et al., F. J. Exp. Med., 147:923 (1978)), DMEM, Fischer's, alpha medium, NCTC, F-10, Leibovitz's L-15, MEM and McCoy's. Particular examples of nutrients that will be readily apparent to the skilled artisan include, serum albumin, transferrin, lipids, cholesterol, a reducing agent such as 2-mercaptoethanol or monothioglycerol, pyruvate, butyrate, and a glucocorticoid such as hydrocortisone 2-hemisuccinate. More particularly, the standard media includes an energy source, vitamins or other cell-supporting organic compounds, a buffer such as HEPES, Tris, that act to stabilize the pH of the media, various inorganic salts. Particular reference is made to PCT Publ. No. WO 95/00632, wherein a variety of serum-free cellular growth media is described, such disclosure is incorporated herein by reference.
  • For any of the ex vivo methods of the invention, peripheral blood progenitor cells (PBPC) and peripheral blood stem cells (PBSC) are collected using apheresis procedures known in the art. See, for example, Bishop et al., Blood, vol. 83, No. 2, pp. 610-616 (1994). Briefly, PBPC and PBSC are collected using conventional devices, for example, a Haemonetics® Model V50 apheresis device (Haemonetics, Braintree, Mass.). Four-hour collections are performed typically no more than five times weekly until, for example, approximately 6.5×108 mononuclear cells (MNC)/kg patient are collected. The cells are suspended in standard media and then centrifuged to remove red blood cells and neutrophils. Cells located at the interface between the two phases (also known in the art as the buffy coat) are withdrawn and resuspended in HBSS. The suspended cells are predominantly mononuclear and a substantial portion of the cell mixture are early stem cells. The resulting stem cell suspension then can be contacted with biotinylated anti-CD34 monoclonal antibodies or other cell-binding means. The contacting period is maintained for a sufficient time to allow substantial interaction between the anti-CD34 monoclonal antibodies and the CD34 antigens on the stem cell surface. Typically, times of at least one hour are sufficient. The cell suspension then is brought into contact with the isolating means provided in the kit. The isolating means can comprise a column packed with avidin-coated beads. Such columns are well known in the art, see Berenson, et al., J. Cell Biochem., 10D:239 (1986). The column is washed with a PBS solution to remove unbound material. Target stem cells can be released from the beads and from anti-CD34 monoclonal antibody using conventional methods. The stem cells obtained in this manner can be frozen in a controlled rate freezer (e.g., Cryo-Med®, Mt. Clemens, Mich.), then stored in the vapor phase of liquid nitrogen. Ten percent dimethylsulfoxide can be used as a cryoprotectant. After all collections from the donor have been made, the stem cells are thawed and pooled. Aliquots containing stem cells, growth medium, such as McCoy's 5A medium, 0.3% agar, and at least one of the expansion factors: recombinant human GM-CSF, IL-3, recombinant human flt3-ligand, and recombinant human GM-CSF/IL-3 fusion molecules (PIXY321) at concentrations of approximately 200 U/mL, are cultured and expanded at 37° C. in 5% CO2 in fully humidified air for 14 days. Optionally, human IL-1α or IL-4 may be added to the cultures. The most preferred combination of expansion factors comprises flt3-ligand plus either IL-3 or a GM-CSF/IL-3 fusion protein.
  • For in vivo administration to humans, flt3-ligand can be formulated according to known methods used to prepare pharmaceutically useful compositions. Flt3-L can be combined in admixture, either as the sole active material or with other known active materials (e.g. CD40 binding proteins, such as CD40-L or antibodies reactive with CD40, 4-1BB-L or antibodies reactive with 4-1BB, interferon alpha, RANKL, CD30 ligand antagonists), with pharmaceutically suitable diluents (e.g., Tris-HCl, acetate, phosphate), preservatives (e.g., Thimerosal, benzyl alcohol, parabens), emulsifiers, solubilizers, adjuvants and/or carriers. Suitable carriers and their formulations are described in Remington's Pharmaceutical Sciences, 16th ed. 1980, Mack Publishing Co. In addition, such compositions can contain flt3-ligand complexed with polyethylene glycol (PEG), metal ions, or incorporated into polymeric compounds such as polyacetic acid, polyglycolic acid, hydrogels, etc., or incorporated into liposomes, microemulsions, micelles, unilamellar or multilamellar vesicles, erythrocyte ghosts or spheroblasts. Such compositions will influence the physical state, solubility, stability, rate of in vivo release, and rate of in vivo clearance of flt3-ligand.
  • Flt3-L can be administered topically, parenterally, or by inhalation. The term “parenteral” includes subcutaneous injections, intravenous, intramuscular, intracisternal injection, or infusion techniques. These compositions will typically contain an effective amount of the flt3-ligand, alone or in combination with an effective amount of any other active material, e.g. those described above. Effective amounts, or dosages, and desired concentrations of flt3-L and active compounds (e.g. CD40-L and/or 4-1BB-L, antibodies reactive with 4-1BB, interferon alpha, RANKL, CD30 ligand antagonists) contained in the compositions may vary depending upon many factors, including the intended use, patient's body weight and age, and route of administration. Preliminary doses can be determined according to animal tests, and the scaling of dosages for human administration can be performed according to art-accepted practices. Keeping the above description in mind, typical dosages of flt3-ligand may range from about 10 μg per square meter to about 1000 μg per square meter. A preferred dose range is on the order of about 100 μg per square meter to about 300 μg per square meter.
  • In addition to the above, the following examples are provided to illustrate particular embodiments and not to limit the scope of the invention.
  • EXAMPLE 1 Generation of Dendritic Cells
  • This Example describes a method for using flt3-ligand to generate large numbers of dendritic cells ex vivo. Cells having the CD34+ phenotype are isolated as described above, for example, first by generating a buffy coat of cells using a procedure described supra. Cells from the buffy coat are then incubated with a CD34 specific monoclonal antibody. The CD34+ cells which are selected then are cultured in McCoy's enhanced media with 20 ng/ml each of GM-CSF, IL-4, TNF-α, or 100 ng/ml flt3-ligand or c-kit ligand. The culture is continued for approximately two weeks at 37° C. in 10% CO2 in humid air. Cells then are sorted by flow cytometry for CD1a+ and HLA-DR+ expression. The combination of GM-CSF, IL-4 and TNF-α, resulted in a six to seven-fold increase in the number of cells obtained after two weeks of culture. The combination of flt3-ligand and c-kit ligand resulted in an additive 12-13-fold increase in absolute cell numbers. This correlated with an 18-fold expansion with either flt3-ligand or c-kit ligand or to a 34-fold expansion with the combination of flt3-ligand and c-kit ligand. Phenotypic analysis of the cells showed that between 60-70% of the cells were HLA-DR+, CD86+, with 40-50% of the cells expressing CD1 a in all factor combinations examined. The addition of flt3-ligand increased the absolute number of CD1a+ cells by 5-fold. c-Kit ligand increased those cells by 6.7-fold and the combination of flt3-ligand and c-kit ligand by 11-fold. Functional analysis of the resultant cells in an MLR revealed that the presence of flt3-ligand or c-kit ligand did not affect the stimulatory capacity of the resultant dendritic cells while increasing the numbers attained.
  • EXAMPLE 2 Use of Flt3-L in Dendritic Cell Expansion
  • This Example describes a method for using flt3-ligand for dendritic cell expansion. Prior to cell collection, it may be desirable to mobilize or increase the numbers of circulating PBPC and PBSC. Mobilization can improve PBPC and PBSC collection, and is achievable through the intravenous administration of flt3-ligand or sargramostim (Leukine®, Immunex Corporation, Seattle, Wash.) to the patients prior to collection of such cells. Other growth factors such as CSF-1, GM-CSF, c-kit ligand, G-CSF, EPO, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, GM-CSF/IL-3 fusion proteins, LIF, FGF and combinations thereof, can be likewise administered in sequence, or in concurrent combination with flt3-ligand. Mobilized or non-mobilized PBPC and PBSC are collected using apheresis procedures known in the art. See, for example, Bishop et al., Blood, vol. 83, No. 2, pp. 610-616 (1994). Briefly, PBPC and PBSC are collected using conventional devices, for example, a Haemonetics® Model V50 apheresis device (Haemonetics, Braintree, Mass.). Four-hour collections are performed typically no more than five times weekly until approximately 6.5×108 mononuclear cells (MNC)/kg patient are collected. Aliquots of collected PBPC and PBSC are assayed for granulocyte-macrophage colony-forming unit (CFU-GM) content by diluting approximately 1:6 with Hank's balanced salt solution without calcium or magnesium (HBSS) and layering over lymphocyte separation medium (Organon Teknika, Durham, N.C.). Following centrifugation, MNC at the interface are collected, washed and resuspended in HBSS. One milliliter aliquots containing approximately 300,000 MNC, modified McCoy's 5A medium, 0.3% agar, 200 U/mL recombinant human GM-CSF, 200 u/mL recombinant human IL-3, and 200 u/mL recombinant human G-CSF are cultured at 37° C. in 5% CO2 in fully humidified air for 14 days. Optionally, flt3-ligand or GM-CSF/IL-3 fusion molecules (PIXY 321) may be added to the cultures. These cultures are stained with Wright's stain, and CFU-GM colonies are scored using a dissecting microscope (Ward et al., Exp. Hematol., 16:358 (1988). Alternatively, CFU-GM colonies can be assayed using the CD34/CD33 flow cytometry method of Siena et al., Blood, Vol. 77, No. 2, pp 400-409 (1991), or any other method known in the art.
  • CFU-GM containing cultures are frozen in a controlled rate freezer (e.g., Cryo-Med®, Mt. Clemens, Mich.), then stored in the vapor phase of liquid nitrogen. Ten percent dimethylsulfoxide can be used as a cryoprotectant. After all collections from the patient have been made, CFU-GM containing cultures are thawed and pooled. The thawed cell collection is contacted with flt3-ligand either alone, sequentially or in concurrent combination with other cytokines listed above. Such exposure to flt3-ligand will drive the CFU-GM to dendritic cell lineage. The dendritic cells are reinfused intravenously to the patient.
  • EXAMPLE 3 Use of Flt3-L in Augmenting Anti-Tumor Immune Responses
  • This Example describes a method for using flt3-L to augment anti-tumor immune responses in vivo. Female C57BL/10J (B10) mice (The Jackson Laboratory, Bar Harbor, Me.) were injected with 5×105 viable B10.2 fibrosarcoma tumor cells by intradermal injection in a midline ventral position in a total volume of 50 μl. The fibrosarcoma B10.2 line is of B10 origin and has been described previously, see Lynch et a., Euro. J. Immunol., 21:1403 (1991) incorporated herein by reference. The fibrosarcoma B10.2 line was induced by subcutaneous implantation of a paraffin pellet containing 5 mg of methylcholanthrene. The tumor cell line was maintained in vitro in α-modified MEM containing 5% FBS, 2 nM L-glutamine, 50 U/ml penicillin and 50 μg/ml streptomycin. Recombinant human flt3-L (10 μg/injection) was administered on a daily basis over a 19-day period (unless otherwise noted) by subcutaneous injection in a total volume of 100 μl. Control mice were similarly injected with a similar volume of buffer containing 100 ng MSA. Tumor growth rates were determined by plotting the tumor size versus time after tumor challenge. Tumor size was calculated as the product of two perpendicular diameters, measured by calipers, and is expressed as the mean tumor size of only those mice bearing a tumor within a particular treatment group. The number of mice bearing tumors compared to the number challenged for each treatment group at the termination of an experiment are shown in the data below.
  • From Table I, the data is a compilation of six different experiments wherein B10.2 tumor-bearing mice were either treated with flt3-ligand or MSA. Complete tumor regression was observed in 19 of 50 flt3-ligand treated mice compared to 1 of 30 in MSA-treated mice (p<0.0001 using Fishers Exact Test). The rate of tumor growth in flt3-ligand treated mice (mean tumor size in tumor-bearing mice at week 5 post-tumor challenge was 60+/−8 mm2) was significantly reduced (p.0001 by Analysis of Variance) compared to MSA-treated mice (mean tumor size at week 5 post-tumor challenge was 185+/−17 mm2).
    TABLE I
    B10.2 Fibrosarcoma +/− Flt3-L Composite of Six Experiments
    Tumor Size (mm2)
    Weeks
    Post MSA Control Standard Flt3-L Standard
    Tumor Challenge (100 ng/day) Error (10 μg/day) Error
    0 0 0 0 0
    1 25 2.6 24 2.2
    2 62 7.5 49 3.6
    3 98 10.6 49 3.9
    4 149 14.5 50 5
    5 185 16.8 60 8.4

    Tumor size was sharply retarded with flt3-L compared to the control. Therefore, the data show that flt3-L is an important cytokine in the augmentation of the immune response against tumor and foreign antigens, and in particular against cancer.
  • EXAMPLE 4 Use of Flt3-L in Combination Therapy to Activate an Immune Response
  • This Example demonstrates the use of flt3-L in combination with a CD40 binding protein to augment anti-tumor immune responses in vivo. In one study C57BL/10J (B10) mice (The Jackson Laboratory, Bar Harbor, Me.) were injected intradermally with 5×105 cells of the viable B10.2 fibrosarcoma tumor cell line described in Example 3 above, and the mice were subdivided into four sets each containing eight mice. In one set of mice, beginning on the same day as the tumor injections, recombinant human flt3-L (10 μg/injection/day) was administered to each mouse on a daily basis over a 20-day period by subcutaneous injection in a total volume of 100 μl. In another set of mice each mouse was injected with the same volume and amount of CD40-L each day for 20 days. In a third set, each mouse was injected with a combination of 10 μg flt3-L and 10 μg of CD40-L per day for 20 days. Control mice were similarly injected with a similar volume of buffer containing 100 ng MSA. Tumor growth rates were determined measuring tumor size each week after tumor challenge over a 6 week period. Tumor size was calculated as the product of two perpendicular diameters, measured by calipers, and is expressed as the mean tumor size. Only mice bearing tumors within each group were considered in determining the mean size. The frequency of tumor rejections was also determined and expressed as the number of mice bearing no tumors compared to the number challenged for each treatment group at the termination of an experiment.
  • Table II provides data in the form of mean tumor size in tumor bearing animals, calculated once a week over a 6 week period post challenge. Table III details the percent frequency of tumor rejection for each set of mice over a 6 week period post challenge. The data demonstrate that for tumor bearing mice, the mean tumor size mice in mice treated with flt3-L and flt3-L in combination with CD40-L is comparable and less than the tumor size in tumor bearing control mice. Significantly, however, mice receiving the combination therapy experienced significantly higher frequency of tumor rejection than mice receiving flt3-L or CD40-L alone. More specifically, 6 weeks post challenge, 62.5% of the mice receiving the combination therapy experienced complete tumor rejection. By contrast, at 6 weeks post challenge, 25% of the mice receiving flt3-L alone experienced complete tumor rejection and none of the mice receiving CD40-L alone or MSA experienced complete tumor rejection.
    TABLE II
    B10.2 Fibrosarcoma
    Tumor Size (mm2)
    Weeks Post CD40-L
    Tumor MSA Control Flt3-L (10 μg/ Flt3-L/CD40-L
    Challenge (100 ng/day) (10 μg/day) day) (each 10 μg/day)
    1 28 24 25 23
    2 68 51 62 46
    3 135 64 107 59
    4 239 81 212 80
    5 351 117 343 137
    6 482 159 493 213
  • TABLE III
    B10.2 Fibrosarcoma
    % Frequency of Tumor Rejection
    Weeks Post CD40-L
    Tumor MSA Control Flt3-L (10 μg/ Flt3-L/CD40-L
    Challenge (100 ng/day) (10 μg/day) day) (each 10 μg/day)
    1 0 0 0 0
    2 0 0 0 0
    3 0 0 0 0
    4 0 12.5 0 50
    5 0 25 0 62.5
    6 0 25 0 62.5
  • In another study C3H/HeN mice were injected intradermally with 5×105 cells of a very aggressive tumor, the 87 fibrosarcoma tumor cell line (generated by chronic exposure of C3H/HeN(MTV−) mice to ultraviolet radiation). The mice were then subdivided into four sets, each containing ten mice. In one set of mice, beginning the day after the tumor injections, recombinant human flt3-L (10 μg/injection/day) was administered to each mouse on a daily basis over a 20-day period by subcutaneous injection. In another set of mice, each mouse was injected with the same volume and amount of CD40-L each day, beginning at day 7 and continuing to day 20. In a third set, each mouse received a combination therapy of CD40-L and flt3-L. The combination therapy included 10 μg/day of flt3-L beginning the day after tumor injection and continuing until day 20 and 10 μg/day of CD40-L beginning at day 7 and continuing until day 20. Mice in a control group were similarly injected with a similar volume of buffer containing 100 ng MSA. Tumor growth rates were determined by measuring tumor size each week post tumor challenge over a 6 week period. Tumor size was calculated as the product of two perpendicular diameters, measured by calipers, and is expressed as the mean tumor size. Only mice bearing tumors were considered in determining the mean size. The frequency of tumor rejections was also determine and expressed as the number of mice bearing no tumors compared to the number challenged for each treatment group at the termination of an experiment.
  • Table IV provides data in the form of mean tumor size in tumor bearing animals, calculated once a week over a 6 week period post challenge. Table V details the percent frequency of tumor rejection for each set of mice over a 6 week period post challenge. The data demonstrate that for tumor bearing mice, the mean tumor size mice in mice treated with flt3-L in combination with CD40-L is significantly less than the tumor size in tumor bearing control mice and mice bearing tumors in the groups receiving only flt3-L and only CD40-L. Significantly, mice receiving the combination therapy experienced significantly higher frequency of tumor rejection than mice receiving flt3-L or CD40-L alone. More specifically, 6 weeks post challenge, 50% of the mice receiving the combination therapy experienced complete tumor rejection. By contrast, at 6 weeks post challenge, 10% of the mice receiving flt3L alone experienced complete tumor rejection and none of the mice receiving CD40-L alone or MSA experienced complete tumor rejection.
  • The observations described above demonstrate that a flt3-L and CD40-L combination therapy can dramatically up-regulate anti-tumor immune responses in vivo. The data indicate that a synergy exists between flt3-L and the CD40 binding protein, CD40-L, in that when used alone flt3-L and CD40-L show little or no tumor rejection. In combination the rejection is dramatic. In addition to synergy, studies indicated that the combination of CD40-L and flt3L indused expression of IL-12 mRNA in the tumors.
    TABLE IV
    87 Fibrosarcoma
    Tumor Size (mm2)
    Weeks
    Post MSA
    Tumor Control Flt3-L CD40-L Flt3-L/CD40-L
    Challenge (100 ng/day) (10 μg/day) (10 μg/day) (each 10 μg/day)
    1 23 23 30 29
    2 54 53 49 34
    3 108 94 87 44
    4 176 159 144 67
    5 286 256 247 115
    6 465 439 410 239
  • TABLE V
    87 Fibrosarcoma
    % Frequency of Tumor Rejection
    Weeks
    Post MSA
    Tumor Control Flt3-L CD40-L Flt3-L/CD40-L
    Challenge (100 ng/day) (10 μg/day) (10 μg/day) (each 10 μg/day)
    1 0 0 0 0
    2 0 0 0 0
    3 0 0 0 30
    4 0 10 0 40
    5 0 10 0 50
    6 0 10 0 50
  • EXAMPLE 5 Use of Flt3-L in Combination Therapy to Activate an Immune Response
  • This Example demonstrates the use of flt3-L in combination with an antibody reactive with 4-1BB to augment anti-tumor immune responses in vivo. In one study C57BL/10J (B10) mice (The Jackson Laboratory, Bar Harbor, Me.) were injected intradermally with 5×105 cells of the viable B10.2 fibrosarcoma tumor cell line described in Example 3 above. In one set of mice, beginning on the same day as the tumor injections, recombinant human flt3-L (10 μg/injection/day) was administered to each mouse on a daily basis over a 14-day period by subcutaneous injection in a total volume of 100 μl. In another set of mice each mouse was injected IP with 100 μg of rat anti mu 4-1BB (clone m6) on days 3 and 6 post tumor challenge. In a third set, each mouse was injected with 100 μg rat anti mu 4-1BB clone m6 on days 13 and 16. A fourth set of mice were injected with a combination of 10 μg flt3-L on days 1-14 and 100 μg of rat anti mu 4-1BB clone m6 on days 13 and 16 post tumor challenge. Control mice were injected with buffer containing 100 ng MSA. Tumor growth rates were determined measuring tumor size each week after tumor challenge over a 5 week period. Tumor size was calculated as the product of two perpendicular diameters, measured by calipers, and is expressed as the mean tumor size in mm2. Only mice bearing tumors within each group were considered in determining the mean tumor size. The percent incidence of tumors was also determine and expressed as the number of mice bearing tumors compared to the number challenged for each treatment group at the termination of an experiment.
  • Table VI provides data in the form of mean tumor size in tumor bearing animals, calculated once a week over an 8 week period post challenge. Table VII details the percent incidence of tumors for each set of mice over an 8 week period post challenge. The data demonstrate that for tumor bearing mice, the mean tumor size in mice treated with flt3-L alone and the mean tumor size in mice treated with the anti 4-1BB regimen are similar. However, when flt3-L in combination with an antibody reactive with 4-1BB is administered to mice, mean tumor size in tumor bearing mice is remarkably decreased. Specifically, at 5 weeks post tumor challenge, mice receiving the combination therapy had a mean tumor size of 0, indicating 100% tumor rejection. This data is supported by the numbers in Table VII which demonstrate that mice receiving the combination therapy experienced significantly lower incidence of tumors than mice receiving flt3-L or 4-1BB antibody alone. More specifically, at 5 weeks post challenge, all of the mice receiving the combination therapy experienced complete tumor rejection (0% tumor incidence). By contrast, at 5 weeks post challenge, 70% of the mice receiving flt3-L alone had tumors and 50% and 70% of the mice receiving 4-1BB antibody alone had tumors. This data provides evidence that anti-4-1BB synergizes with flt3-L in enhancing immune response.
    TABLE VI
    B10.2 Fibrosarcoma
    Tumor Size (mm2)
    anti 4-1BB
    Weeks Post MSA anti 4-1BB anti 4-1BB (days 13
    Tumor Control Flt3-L (100 μg, day (100 μg, days and 16)
    Challenge (100 ng/day) (10 μg/day) 3 and 6) 13 and 16) and flt3-L
    1 28 15 25 25 20
    2 60 35 55 60 35
    3 85 35 55 40 15
    4 125 45 60 50 8
    5 200 55 70 40 0
    6 280 95 80 45 0
    7 125 130 115 0
    8 160 135 0
  • TABLE VII
    B10.2 Fibrosarcoma
    % Tumor Incidence
    Flt3-L and
    Weeks Post MSA anti 4-1BB anti 4-1BB anti 4-1BB
    Tumor Control Flt3-L (days 3 (days 13 (days 13
    Challenge (100 ng/day) (10 μg/day) and 6) and 16) and 16)
    1 100 100 100 100 100
    2 100 100 100 100 90
    3 100 100 90 100 80
    4 100 90 80 50 30
    5 100 70 70 50 0
    6 100 60 60 40 0
    7 100 60 50 20 0
    8 100 60 50 20 0
  • EXAMPLE 6 Use of Flt3-L in Combination Therapy to Activate an Immune Response
  • This Example demonstrates the use of flt3-L in combination with interferon alpha to augment anti-tumor immune responses in vivo. In one study the B10.2 fibrosarcoma tumor cell line (described in Example 3 above) was implanted in C57BL/10J (B10) mice on day 0. One set of mice (n=10) was treated with recombinant human flt3-L (50 μg/day, by subcutaneous injection) on days 10 to 29 post tumor challenge. Another set of mice (n=5) was treated with human interferon alpha (interferon alpha A/D; 60,000 U/day, by subcutaneous injection) on days 21 to 25 post tumor challenge. A third set of mice (n=5) was treated with flt3-L on days 10 to 29 and also with interferon alpha on days 21 to 25. Control mice were injected with buffer containing 100 ng MSA. Tumor growth rates were determined by measuring tumor size over a 7 week period. Tumor size was calculated as the product of two perpendicular diameters, measured by calipers, and is expressed as the mean tumor size in mm2. Only mice bearing tumors within each group were considered in determining the mean tumor size. The percent incidence of tumors was also determined (i.e., the number of mice bearing tumors compared to the number challenged) for each treatment group.
  • Table VIII shows the mean tumor size in tumor bearing animals, and Table IX shows the percent incidence of tumors. This data demonstrates that interferon alpha synergizes with flt3-L in enhancing immune response in the B10.2 tumor model. Most significantly, the tumor rejection rate was 40% for flt3-L alone and 80% for the combination of flt3-L and interferon alpha.
    TABLE VIII
    B10.2 Fibrosarcoma
    Tumor size (mm2)
    Flt3L IFNα
    MSA (50 μg/day, (60K U/day, FL +
    Days Post (100 ng/day) days 1-20) days 11-15) IFNα
    Tumor Challenge (n = 10) (n = 10) (n = 5) (n = 5)
    7 17 16 16 16
    14 42 43 46 41
    21 71 76 77 69
    28 125 107 125 101
    35 183 126 198 90
    42 238 131 267 55
    49 322 219 364 186
  • TABLE IX
    B10.2 Fibrosarcoma
    % Tumor incidence
    Flt3L IFNα
    MSA (50 μg/day, (60K U/day, FL +
    Days Post (100 ng/day) days 1-20) days 11-15) IFNα
    Tumor Challenge (n = 10) (n = 10) (n = 5) (n = 5)
    7 100 100 100 100
    14 100 100 100 100
    21 100 100 100 80
    28 100 100 100 80
    35 100 100 100 80
    42 100 80 100 60
    49 100 60 100 20

Claims (7)

1. A vaccine composition comprising:
(a) as an adjuvant, Flt-3 ligand in an amount sufficient increase the number of dendritic cells in a patients in vivo; and
(b) an antigen,
wherein said Flt3-ligand comprises an amino acid sequence that is at least 90% identical to amino acids 28-Xaa of SEQ ID NO:2, wherein Xaa is an amino acid from 160-235 of SEQ ID NO:2, and wherein said Flt3-ligand binds Flt-3.
2. The vaccine composition of claim 1, wherein said Flt3-ligand comprises amino acids 28-Xaa of SEQ ID NO:2, wherein Xaa is an amino acid from 160-235 of SEQ ID NO:2.
3. The vaccine composition of claim 1, wherein said Flt3-ligand comprises amino acids 28-235 of SEQ ID NO:2.
4. The vaccine composition of claim 1, wherein said Flt3-ligand comprises amino acids 28-182 of SEQ ID NO:2.
5. The vaccine composition of claim 1, wherein said antigen is selected from the group consisting of a tumor antigen, a bacterial antigen and a viral antigen.
6. A method of immunizing a patient, comprising administering the vaccine composition of any of claim 1-5 to a patient in need thereof.
7. The method of claim 6, wherein said administering is parenteral, topical or inhalation.
US11/510,651 1995-10-04 2006-08-28 Vaccine composition comprising Flt3-ligand Abandoned US20060292166A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/510,651 US20060292166A1 (en) 1995-10-04 2006-08-28 Vaccine composition comprising Flt3-ligand

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US53914295A 1995-10-04 1995-10-04
US72554096A 1996-10-03 1996-10-03
US15490398A 1998-09-17 1998-09-17
US44402799A 1999-11-19 1999-11-19
US10/241,927 US7361330B2 (en) 1995-10-04 2002-09-11 Methods of using flt3-ligand in the treatment of fibrosarcoma
US10/397,687 US20030165531A1 (en) 1995-10-04 2003-03-25 Flt3-ligand as a vaccine adjuvant
US11/510,651 US20060292166A1 (en) 1995-10-04 2006-08-28 Vaccine composition comprising Flt3-ligand

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/397,687 Continuation US20030165531A1 (en) 1995-10-04 2003-03-25 Flt3-ligand as a vaccine adjuvant

Publications (1)

Publication Number Publication Date
US20060292166A1 true US20060292166A1 (en) 2006-12-28

Family

ID=27496166

Family Applications (4)

Application Number Title Priority Date Filing Date
US10/241,927 Expired - Fee Related US7361330B2 (en) 1995-10-04 2002-09-11 Methods of using flt3-ligand in the treatment of fibrosarcoma
US10/397,687 Abandoned US20030165531A1 (en) 1995-10-04 2003-03-25 Flt3-ligand as a vaccine adjuvant
US10/643,384 Abandoned US20040037845A1 (en) 1995-10-04 2003-08-19 Use of flt3-ligand in the treatment of infection
US11/510,651 Abandoned US20060292166A1 (en) 1995-10-04 2006-08-28 Vaccine composition comprising Flt3-ligand

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US10/241,927 Expired - Fee Related US7361330B2 (en) 1995-10-04 2002-09-11 Methods of using flt3-ligand in the treatment of fibrosarcoma
US10/397,687 Abandoned US20030165531A1 (en) 1995-10-04 2003-03-25 Flt3-ligand as a vaccine adjuvant
US10/643,384 Abandoned US20040037845A1 (en) 1995-10-04 2003-08-19 Use of flt3-ligand in the treatment of infection

Country Status (1)

Country Link
US (4) US7361330B2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017099474A1 (en) * 2015-12-08 2017-06-15 연세대학교 산학협력단 Antitumor composition comprising gm-csf gene, flt3l-trail fusion gene, shrna inhibiting tgf-β expression, and shrna inhibiting hsp expression

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7294331B2 (en) * 1994-03-07 2007-11-13 Immunex Corporation Methods of using flt3-ligand in hematopoietic cell transplantation
US7541184B2 (en) * 2000-02-24 2009-06-02 Invitrogen Corporation Activation and expansion of cells
ATE313334T1 (en) * 2000-04-25 2006-01-15 Immunex Corp TREATING TUMORS WITH PHOTODYNAMIC THERAPY
US20040175373A1 (en) * 2002-06-28 2004-09-09 Xcyte Therapies, Inc. Compositions and methods for eliminating undesired subpopulations of T cells in patients with immunological defects related to autoimmunity and organ or hematopoietic stem cell transplantation
US20050084967A1 (en) 2002-06-28 2005-04-21 Xcyte Therapies, Inc. Compositions and methods for eliminating undesired subpopulations of T cells in patients with immunological defects related to autoimmunity and organ or hematopoietic stem cell transplantation
US20050232931A1 (en) * 2003-06-13 2005-10-20 Oncomax Acquisition Corp. Preparation and application of anti-tumor bifunctional fusion proteins
US20040254108A1 (en) * 2003-06-13 2004-12-16 Jing Ma Preparation and application of anti-tumor bifunctional fusion proteins
EP1701736A4 (en) * 2003-12-24 2008-05-07 Inst Medical W & E Hall Therapeutic agents and uses therefor
US20080075734A1 (en) * 2006-08-30 2008-03-27 Neal Zane C Genetic Immunization
CN112218651A (en) 2018-01-08 2021-01-12 诺华公司 Immunopotentiating RNA for combination with chimeric antigen receptor therapy
US11246908B2 (en) 2018-01-10 2022-02-15 The Johns Hopkins University Compositions comprising albumin-FMS-like tyrosine kinase 3 ligand fusion proteins and uses thereof
US20210231683A1 (en) * 2018-04-20 2021-07-29 Unm Rainforest Innovations Rap1-GTP, Rac1-GTP and FMS-like Tyrosine Kinase 3 ligand (FLT3-L) As Biomarkers For Early Detection of Sepsis
WO2020263830A1 (en) * 2019-06-25 2020-12-30 Gilead Sciences, Inc. Flt3l-fc fusion proteins and methods of use

Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4399493A (en) * 1981-12-30 1983-08-16 Nihon Dennetsu Co., Ltd. Illuminated artificial flower ornament
US4745099A (en) * 1985-02-06 1988-05-17 Chugai Seiyaku Kabushiki Kaisha Pharmaceutical composition for the treatment of the anemia of malignant tumors
US5013824A (en) * 1985-11-19 1991-05-07 Schering Corporation Human interleukin-4 peptides and conjugates thereof
US5057420A (en) * 1987-06-05 1991-10-15 Granada Biosciences, Inc. Bovine nuclear transplantation
US5061620A (en) * 1990-03-30 1991-10-29 Systemix, Inc. Human hematopoietic stem cell
US5114710A (en) * 1988-02-10 1992-05-19 Green Cross Corporation M-csf as a therapeutic agent for thrombocytopenia
US5116964A (en) * 1989-02-23 1992-05-26 Genentech, Inc. Hybrid immunoglobulins
US5185438A (en) * 1991-04-02 1993-02-09 The Trustees Of Princeton University Nucleic acids encoding hencatoporetic stem cell receptor flk-2
US5192553A (en) * 1987-11-12 1993-03-09 Biocyte Corporation Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood and methods of therapeutic use
US5199942A (en) * 1991-06-07 1993-04-06 Immunex Corporation Method for improving autologous transplantation
US5270458A (en) * 1991-04-02 1993-12-14 The Trustees Of Princeton University Nucleic acids encoding fragments of hematopoietic stem cell receptor flk-2
US5326558A (en) * 1989-08-08 1994-07-05 Genetics Institute, Inc. Megakaryocytopoietic factor
US5367057A (en) * 1991-04-02 1994-11-22 The Trustees Of Princeton University Tyrosine kinase receptor flk-2 and fragments thereof
US5397706A (en) * 1991-11-06 1995-03-14 Correa; Paulo N. Serum-free basal and culture medium for hematopoietic and leukemia cells
US5437994A (en) * 1989-06-15 1995-08-01 Regents Of The University Of Michigan Method for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells
US5453357A (en) * 1992-10-08 1995-09-26 Vanderbilt University Pluripotential embryonic stem cells and methods of making same
US5459069A (en) * 1989-06-15 1995-10-17 The Regents Of The University Of Michigan Device for maintaining and growing human stem and/or hematopoietics cells
US5525708A (en) * 1994-03-28 1996-06-11 Cytomed, Inc. Covalent dimer of kit ligand
US5554512A (en) * 1993-05-24 1996-09-10 Immunex Corporation Ligands for flt3 receptors
US5627025A (en) * 1994-08-12 1997-05-06 The Rockefeller University Method for the identification of compounds capable of abrogating human immunodeficiency virus (HIV) infection of dendritic cells and T-lymphocytes
US5635388A (en) * 1994-04-04 1997-06-03 Genentech, Inc. Agonist antibodies against the flk2/flt3 receptor and uses thereof
US6316254B1 (en) * 1994-02-14 2001-11-13 University Of Washington Methods for stimulating erythropoiesis using hematopoietic proteins
US6497876B1 (en) * 1996-07-10 2002-12-24 Immunex Corp. Method of stimulating an immune response with activated dendritic cells
US6632424B1 (en) * 1993-05-24 2003-10-14 Immunex Corporation Human FLTS3 ligand

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5399493A (en) * 1989-06-15 1995-03-21 The Regents Of The University Of Michigan Methods and compositions for the optimization of human hematopoietic progenitor cell cultures
EP0584184A4 (en) 1991-04-09 1995-12-27 Indiana University Foundation System and process for supporting hematopoietic cells
CA2122141C (en) 1991-10-23 2007-07-17 Shelly Heimfeld Methods for selectively expanding stem cells
EP0563485A1 (en) 1992-03-30 1993-10-06 Schering-Plough In vitro generation of human dendritic cells and uses thereof
ATE196775T1 (en) 1993-05-19 2000-10-15 Schering Corp PURIFIED MAMMAL FLT3 LIGANDS, AGONISTS AND ANTAGONISTS THEREOF
CA2163105A1 (en) 1993-05-19 1994-11-24 Charles H. Hannum Purified mammalian flt3 ligands and agonists and antagonists thereof
AU7210394A (en) 1993-06-18 1995-01-17 Trustees Of Princeton University, The Totipotent hematopoietic stem cell receptors and their ligands
US5750119A (en) * 1994-01-13 1998-05-12 Mount Sinai School Of Medicine Of The City University Of New York Immunotherapeutic stress protein-peptide complexes against cancer
WO1996000779A1 (en) 1994-06-28 1996-01-11 Cornell Research Foundation, Inc. Process for ex vivo expansion of hematopoietic progenitor cells
US5626862A (en) * 1994-08-02 1997-05-06 Massachusetts Institute Of Technology Controlled local delivery of chemotherapeutic agents for treating solid tumors
BR0111948A (en) * 2000-06-26 2004-08-24 Rxkinetix Inc Composition for delivery of hematopoietic growth factor
PL377028A1 (en) * 2002-03-26 2006-01-23 Immunex Corporation Methods of using flt3-ligand in immunization protocols

Patent Citations (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4399493A (en) * 1981-12-30 1983-08-16 Nihon Dennetsu Co., Ltd. Illuminated artificial flower ornament
US4745099A (en) * 1985-02-06 1988-05-17 Chugai Seiyaku Kabushiki Kaisha Pharmaceutical composition for the treatment of the anemia of malignant tumors
US5013824A (en) * 1985-11-19 1991-05-07 Schering Corporation Human interleukin-4 peptides and conjugates thereof
US5057420A (en) * 1987-06-05 1991-10-15 Granada Biosciences, Inc. Bovine nuclear transplantation
US5192553A (en) * 1987-11-12 1993-03-09 Biocyte Corporation Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood and methods of therapeutic use
US5114710A (en) * 1988-02-10 1992-05-19 Green Cross Corporation M-csf as a therapeutic agent for thrombocytopenia
US5116964A (en) * 1989-02-23 1992-05-26 Genentech, Inc. Hybrid immunoglobulins
US5459069A (en) * 1989-06-15 1995-10-17 The Regents Of The University Of Michigan Device for maintaining and growing human stem and/or hematopoietics cells
US5437994A (en) * 1989-06-15 1995-08-01 Regents Of The University Of Michigan Method for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells
US5326558A (en) * 1989-08-08 1994-07-05 Genetics Institute, Inc. Megakaryocytopoietic factor
US5061620A (en) * 1990-03-30 1991-10-29 Systemix, Inc. Human hematopoietic stem cell
US5548065A (en) * 1991-04-02 1996-08-20 The Trustees Of Princeton University Tyrosine kinase receptor human flk-2-specific antibodies
US5367057A (en) * 1991-04-02 1994-11-22 The Trustees Of Princeton University Tyrosine kinase receptor flk-2 and fragments thereof
US5185438A (en) * 1991-04-02 1993-02-09 The Trustees Of Princeton University Nucleic acids encoding hencatoporetic stem cell receptor flk-2
US5283354A (en) * 1991-04-02 1994-02-01 The Trustees Of Princeton University Nucleic acids encoding hematopoietic stem cells receptors flk-1
US5270458A (en) * 1991-04-02 1993-12-14 The Trustees Of Princeton University Nucleic acids encoding fragments of hematopoietic stem cell receptor flk-2
US5199942A (en) * 1991-06-07 1993-04-06 Immunex Corporation Method for improving autologous transplantation
US5397706A (en) * 1991-11-06 1995-03-14 Correa; Paulo N. Serum-free basal and culture medium for hematopoietic and leukemia cells
US5453357A (en) * 1992-10-08 1995-09-26 Vanderbilt University Pluripotential embryonic stem cells and methods of making same
US5843423A (en) * 1993-05-24 1998-12-01 Immunex Corporation Methods of stimulating hematopoietic cells with flt3-ligand
US5554512A (en) * 1993-05-24 1996-09-10 Immunex Corporation Ligands for flt3 receptors
US6632424B1 (en) * 1993-05-24 2003-10-14 Immunex Corporation Human FLTS3 ligand
US6316254B1 (en) * 1994-02-14 2001-11-13 University Of Washington Methods for stimulating erythropoiesis using hematopoietic proteins
US5525708A (en) * 1994-03-28 1996-06-11 Cytomed, Inc. Covalent dimer of kit ligand
US5635388A (en) * 1994-04-04 1997-06-03 Genentech, Inc. Agonist antibodies against the flk2/flt3 receptor and uses thereof
US5627025A (en) * 1994-08-12 1997-05-06 The Rockefeller University Method for the identification of compounds capable of abrogating human immunodeficiency virus (HIV) infection of dendritic cells and T-lymphocytes
US6497876B1 (en) * 1996-07-10 2002-12-24 Immunex Corp. Method of stimulating an immune response with activated dendritic cells

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017099474A1 (en) * 2015-12-08 2017-06-15 연세대학교 산학협력단 Antitumor composition comprising gm-csf gene, flt3l-trail fusion gene, shrna inhibiting tgf-β expression, and shrna inhibiting hsp expression
EP3388522A4 (en) * 2015-12-08 2019-05-22 University-Industry Foundation, Yonsei University Antitumor composition comprising gm-csf gene, flt3l-trail fusion gene, shrna inhibiting tgf- expression, and shrna inhibiting hsp expression

Also Published As

Publication number Publication date
US7361330B2 (en) 2008-04-22
US20030113341A1 (en) 2003-06-19
US20030165531A1 (en) 2003-09-04
US20040037845A1 (en) 2004-02-26

Similar Documents

Publication Publication Date Title
AU697539B2 (en) Dendritic cell stimulatory factor
US20060292166A1 (en) Vaccine composition comprising Flt3-ligand
CA2259140C (en) Method of activating dendritic cells
US6017527A (en) Activated dendritic cells and methods for their activation
US6291661B1 (en) flt3-L mutants and method of use
US20090075886A1 (en) Dendritic cell stimulatory factor
US20030124091A1 (en) Endothelial cell derived hematopoietic growth factor
US20030077263A1 (en) Method of activating dendritic cells
US7150992B1 (en) Methods of preparing dendritic cells with flt3-ligand and antigen
MXPA98002464A (en) Dendrit cellular stimulating factor
US20050048645A1 (en) Method of treating or preventing disease characterized by cryptococcus neoformans infection

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION