WO1997029781A1 - Methods and compositions for modulating an immune response - Google Patents

Methods and compositions for modulating an immune response Download PDF

Info

Publication number
WO1997029781A1
WO1997029781A1 PCT/US1997/002350 US9702350W WO9729781A1 WO 1997029781 A1 WO1997029781 A1 WO 1997029781A1 US 9702350 W US9702350 W US 9702350W WO 9729781 A1 WO9729781 A1 WO 9729781A1
Authority
WO
WIPO (PCT)
Prior art keywords
dna
protein
peptide
seq
leu
Prior art date
Application number
PCT/US1997/002350
Other languages
French (fr)
Inventor
Richard J. Armitage
William C. Fanslow
Carlos Escobar
Jodee Zappone
Original Assignee
Immunex Corporation
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 Corporation filed Critical Immunex Corporation
Priority to AU22733/97A priority Critical patent/AU2273397A/en
Publication of WO1997029781A1 publication Critical patent/WO1997029781A1/en

Links

Classifications

    • 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
    • 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
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • 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
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • 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
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/204IL-6
    • 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
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2066IL-10
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70596Molecules with a "CD"-designation not provided for elsewhere
    • 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
    • A61K2039/53DNA (RNA) vaccination
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • the present invention relates generally to the field of rnarnmalian proteins having immunoregulatory activity, and more specifically to mammalian proteins involved in regulation of a humoral immune response.
  • CD83 is a member of the immunoglobulin superfamily that is expressed on the surface of certain dendri ⁇ c lineage cells and some lymphoblastoid cell types (Zhou et al., J. Immunol. 149:735. 19S2: Zhou et al., ./. Immunol. 154:3821 , 1995).
  • the presence of CD83 on dendritic cells lias led to the hypothesis that it is somehow involved in antigen presentation; however, prior to the present invention, no biological functions were known for CD83.
  • Vaccination is ar; efficient means of preventing death or disability from infectious diseases.
  • Vaccination is ar; efficient means of preventing death or disability from infectious diseases.
  • Parenteral routes of administration the numbers of different vaccinations required and the need for, and frequency of, booster immunizations all impe ⁇ e efforts to control or eliminate disease.
  • inability to modulate the type of response, and isotype of antibody made, during immunization has hampered vaccination programs.
  • numerous vaccine adjuvants are known, alum is the only adjuvant widely used in humans.
  • Tht te was furthermore a need to develop agents useful in stimulating secretion of antibody, . > develop effective methods of immunization, and to discover alternative tvpes of ants, suitable for use in humans.
  • the present inv.ntion provides a method of stimulating a humoral immune response, comprising administering a CD83 reagent and an antigen, in a pharmaceutically acceptable carrier, wherein the CD83 stimulates production of antigen-specific antibodies
  • Useful CD83 reagents include DNA's encoding CD83 and CD83 polypeptides, as well as derivatives and analogs of such reagents that have CD83 biological activity.
  • the present invention further provides vaccine compositions useful in stimulating a humoral immune response.
  • CD83 DNA's that are useful in the inventive methods and compositions include a DNA having a nucleotide sequence encoding an amino acid sequence of amino acids 1 through 124 of SEQ ID NO: 2 and DNA molecules capable of hybridization to such DNA under stringent conditions and which encode biologically active CD83.
  • Useful CD83 proteins include a CD83 peptide having an amino acid sequence of amino acids 1 through 124 of SEQ ID NO: 2, fragments of such a peptide according that have CD83 biological activity; and peptides encoded by DNA molecules capable of hybridization to a DNA encoding such peptide under stringent conditions, and which encode biologically active CD83.
  • CD83 reagents are DNA's that encode, and CD83 peptides that comprise, the extracellular domain of CD83.
  • cytokine that modulates an immune response in conjunction with a CD43 composition (either sequentially, simultaneously or separately), particularly cytokines selected from the group consisting of In.e ⁇ eukins 1 , 2, 4, 5, 6, 7, 10, 12 and 15; granulocyte-macrophage colony stimulating factor, granulocyte colony stimulating factor; a fusion protein comprising Interleukin and granulocyte-macrophage colony stimulating factor, Interferon- ⁇ , TNF; TGF-I ⁇ ; flt-3 ligand; soluble CD40 ligand; biologically active derivatives of these cytokines; and combinations thereof.
  • cytokines selected from the group consisting of In.e ⁇ eukins 1 , 2, 4, 5, 6, 7, 10, 12 and 15; granulocyte-macrophage colony stimulating factor, granulocyte colony stimulating factor; a fusion protein comprising Interleukin and granulocyte-macrophage colony stimulating factor, Interferon- ⁇ , TNF; TGF
  • the present invention thus also provides a method of inhibiting undesirable antigen specific responses in a mammal. Such methods of inhibiting undesirable antigen specific responses are useful in preventing or treating autoimmune disease as well as tissue or organ transplant rejection, and in treatment or prevention of allergy or asthma.
  • Figure 1 demonstrates that mice immunized with antigen (DNA encoding TNFr Fc) in the presence of CD8? have significantly higher serum titers of TNFr/Fc-specific IgG2b than mice immunized wuh antigen alone.
  • Figure 2 illusrrat-S the ability of CD83 to stimulate higher levels of antigen-specific IgG2b when the antigen used is a soluble protein antigen (TNFr/Fc).
  • CD83 was cloned from a Raji cell library by polymerase chain reaction, using primers based on the p iblished sequence (Zhou et al., J. Immunol. 149:735, 1992).
  • Several different soluble forms of CD83 were expressed, including a Type I Fc/CD83 fusion protein, a Flag ® /C1 83 fusion protein, and a soluble form of CD83 consisting of the extracellular domain.
  • CD83 (also referred to as HB15) is a 45KD glycoprotein predominantly expressed on the surface of dendritic lineage cells, such as skin Langerhans cells and interdigitating reticulum cells present in the T cell zones of lymphoid organs. It is also weakly expressed by some lymphoblastoid cell types, and can be upregulated under certain activation conditions. Structural analysis of the predicted amino acid sequence of this protein established it as a me .r of the immunoglobulin superfamily (Zhou et al., J. Immunol. 149:735, 1992). It has more recently been shown that human blood dendritic cells express CD83 (Zhou et al., ./. I.nmun ⁇ l. 154:3821, 1995).
  • CD83 both in protein fo ⁇ n and in DNA form
  • CD83 will be useful as a vaccine adjuvant.
  • CD83 can be administered in ivnjunction with other immunomodulatory molecules, as described herein.
  • DN ⁇ ending CD83 can be incorporated into attenuated live viral or bacterial vaccine strain ,, to enhance the immune response to the infectious agent.
  • antagonist s of CD83 will be useful in suppressing an undesirable, antigen- specific immune respon ,e.
  • the protective immune r. sponse An immune response to a pathogen can be classified broadly as either being cell- mediated (cellular immunity) or antibody mediated (humoral immunity). In cellular immunity, activated mac ophages and cytotoxic lymphocytes carry out elimination of the pathogen. Humoral immunity, in contrast, operates primarily through antibody production. It is currently believed that these two arms of the immune response are regulated by distinct su isets of helper T (TH) cells which secrete specific arrays of cytokines (reviewed in Immunological Reviews 123, 1991).
  • TH helper T
  • Type 1 TH cells mediate delayed type hypersensitivity (DTH), and secrete Interferon- ⁇ (IF,' - ⁇ ) and Interleukin-2 (IL-2), while Type 2 TH cells (TH2 cells) secrete primarily Interleukins 4, 5 and 10 (IL-4, IL-5 and IL-10, respectively) and provide B cell help.
  • DTH delayed type hypersensitivity
  • IF,' - ⁇ Interferon- ⁇
  • IL-2 Interleukin-2
  • Type 2 TH cells secrete primarily Interleukins 4, 5 and 10 (IL-4, IL-5 and IL-10, respectively) and provide B cell help.
  • the immune response may be manipulated toward either a THI or TH2 by the appropriate administration of cytokines, or cytokine antagonists.
  • administration of IFN- ⁇ or an antibody that neutralizes IL-4 would enhance a THI response
  • administration of IL-10 or a molecule that inhibited the action of IFN- ⁇ would stimulate a TH2 response.
  • This ability to manipulate the immune response provides a useful tool not only in infectious disease, but in inflammatory and allergic diseases as well (see, for example, ow ⁇ e and Cotfman, Immunol. Today 14:270, 1993).
  • IgM Early antibody ⁇ . -.ponses. both in the life cycle of an animal and in the ontogeny of individual B cell clones primarily consist of IgM. Under the control of helper T cells, the isotype of antibody pro uced by B cells is switched from IgM to IgG, IgE or IgA.
  • the latter isotypes are representative of a more mature immune response, and generally include antibodies of higher affinity and avidity as well as increased effector function. Stimulation of non-IgM isotypes is considered a desirable effect of any vaccination protocol, since it is the IgG, IgE and IgA aim bodies that are generally protective against infectious disease, and which are likely to play a role in tumor immunity.
  • the IgG subclasses are preferred for the generation of monoclonal antibodies, since these exhibit useful characteristics (i.e., easier purification, higher affinity, greater therapeutic effectiveness due to enhanced effector tun tions).
  • Immunization I a centuries old, and highly effective, means of inducing a protective immune res p. use against pathogens in order to prevent or ameliorate disease.
  • the vaccines that have been used for such induction are generally live, attenuated microorganisms, or preparations of killed organisms or fractions thereof. Live, attenuated vaccines are generally thought to more closely mimic the immune response that occurs with a natural infection than do those prepared from killed microbes or non-infective preparations derived ti >m pathogens (i.e., toxoids, recombinant protein vaccines). However, attenuated vaccines also present a risk of reversion to pathogemcity, and can cause illness, especially M immunocompromised individuals.
  • Numerous vaccines are licensed for administration to humans, including live virus vaccines for certain adenoviruses, measles, mumps and rubella viruses, and poliovirus, diphtheria and tetanus toxoid vaccines, and Haemophilus b and meirngococcal polysaccharide vaccines (Hinman et al., in Principles and Practice of Infectious Diseases. 3rd edition; G.L. Mandell, R.G. Douglas and J.E. Bennett, eds., Churchill Livingstone Inc., NY, NY; 2320-2333; Table 2).
  • Useful adjuvants include for example alum, fragments of bacterial membranes, l osomes, coupling a protein of interest to a larger immunogenic protein, RIBI, non-ion ⁇ ⁇ block co-polymer surfactants and TiterMax ® .
  • Other useful vaccine adjuvants and e xcipients are described by Vogel and Powell (A Compendium of Vaccine Adjuvants and Excipients.
  • cytokines to modulate an immune re:- >onse.
  • Some cytokines e.g., interleukin-4 (IL-4) and GM-CSF, attract and activate antigen-presenting cells for more efficient presentation of antigens to T cells.
  • IL-4 interleukin-4
  • GM-CSF GM-CSF
  • cytokines have been co-administered with antigen to increase antigenic activity
  • Other studies have shown that the host response to tumor challenge can be increased by inoculation of tumor cells genetically engineered to express particular cytokines, including ⁇ -H " N, TNF- ⁇ , IL-2, IL-4, IL-6, IL-7, or GM-CSF Recombinant antigens have been expressed as fusion proteins with cytokines, for example as described in USSN 08/271,875, fil-d July 7, 1994
  • the present invention provides isolated CD83 DNA s and proteins (referred to as CD83 agents) having .mmunoregulatory activity
  • CD83 agents referred to as CD83 agents
  • Such DNA's and proteins are substantially free of contaminating endogenous mate ⁇ als and, opnonally, without associated native-pattern glycosylation
  • De ⁇ vatives of the CD83 proteins within the scope of the invention also ⁇ n ⁇ ude various structural forms of the p ⁇ mary protein which retain biological activity Due to the presence of lonizable amino and carboxyl groups, for example, a CD83 proteir may be in the form of acidic or basic salts, or may be in neutral form Individual ammo « ⁇ c ⁇ d residues may also be modified by oxidation or reduction.
  • the primary ammo acid structure may be modified by forming covalent or aggregative conjugates with other chemical moieties, such as glycosyl groups, lipids, phosphate, acetyl groups and the like, or by creaung a ino acid sequence mutants.
  • Covalent de ⁇ vatives are prepared by linking pa ⁇ icular functional groups to amino acid side chains or at the N- or C-t.rmini
  • de ⁇ vatives of the CD83 protein within the scope of this invention include covalent or aggiegative -onjugates of the protein or its fragments with other proteins or polypeptides, such as , synthesis in recombinant culture as N-terminal or C-terminal fusions
  • the conjugated peptide may be a signal (or leader) polypepude sequence at the N-te ⁇ inal region of the protein which co-translationally or post- translationally directs uansfer of the protein from its site of synthesis to its site of funcuon inside or outside ot the c ⁇ 11 membrane or wall (e.g , the yeast ⁇ -factor leader).
  • Protein fusions can comprise peptides added to facilitate punficauon or identification of CD83
  • oteins e g , poly-His
  • the amino acid sequence of the CD83 proteins can also be linl .d to an identification peptide such as that desc ⁇ bed by Hopp et al., Bio/Technology 6: 1-04 ( 1988). Such a highly antigenic peptide provides an epitope reversibly bound by a .pecific monoclonal antibody, enabling rapid assay and facile purification of expressed recombinant protein..
  • the sequence of Hopp et al. is also specifically cleaved by bovine mucosal enterokinase, allowing removal of the peptide from the purified protein. Fusion proteins capped with such peptides may also be resistant to intracellular degradation n E. coll.
  • Fusion proteins fu ⁇ her comprise the amino acid sequence of a CD83 protein linked to an immunoglobulin Iv region.
  • An exemplary Fc region is a human IgGl having a nucleotide and amino acd sequence set fo ⁇ h in SEQ ID NO:3. Fragments of an Fc region may also be used.
  • a CD83 protein may be expressed as a dimer, through formation of interchain disulfide bonds. If CD83 fusion proteins are made with both heavy and light chains of an antibody, it is possible to form a protein oligomer with as many as four CD83 protein regions.
  • CD83 proteins fu ⁇ her comprise an oligomerizing zipper domain. Oligomerizing zipper domains are described in USSN 08/107,353, filed August 13, 1993, the relevant disclosure of which is incorporated by reference herein.
  • leucine zipper domains are those found in the yeast transcription factor GCN4 and a heat-stable DNA-bindinj; protein found in rat liver (C EBP; Landschulz et al., Science 243: 1681, 1989), the nut lear transforming proteins,/os and jun, which preferentially form a heterodimer (O'Shea et al., Science 245:646, 1989; Turner and Tjian, Science 243:1689, 1989), and the gene product of the murine proto-oncogene, c-myc (Landschulz et al., Science 240: 1759, 198! ⁇ ).
  • the fusogenic proteins of several different viruses including paramyxovirus, coronavirus, measles virus and many retroviruses, also possess leucine zipper domains (Buckland and Wild, Nature 338:547, 1989; Britton, Nature 353:394, 1991; Delwart and Mosialos, AIDS Research and Human Retroviruses 6:703, 1990).
  • fusion proteins include fusions of CD83 with an antigen against which it is desired to elicit an ir. mune response, for example as described in USSN 08/271,875, filed July 7, 1994. for GM-CSF.
  • fusion proteins consisting of CD83 and another cytokine or cyto* ines are also contemplated.
  • CD83 DNA's the DNA's encoding such fusion proteins will also have utility in the instant invention.
  • a very useful DNA may ii elude not only sequences encoding CD83 and another cytokine (for example, CD40L). but also sequences encoding the antigen(s).
  • CD83 protein -erivatives may also be used as immunogens, reagents in immunoassays, or as binding agents for affinity purification procedures, for example, in purifying CD83 antibodies.
  • CD83 protein derivatives may also be obtained by cross- linking agents, such as M-maleimidobenzoyl succinimide ester and N-hydroxysuccinimide, at cysteine and lysitie re sidues.
  • CD83 proteins may also be covalently bound through reactive side croups to various insoluble substrates, such as cyanogen bromide-activated, bisoxirane-activated, car-tonyldiimidazole-activated or tosyl-activated agarose structures, or by adsorbing to polyolel.n surfaces (with or without glutaraldehyde cross-linking).
  • substrates such as cyanogen bromide-activated, bisoxirane-activated, car-tonyldiimidazole-activated or tosyl-activated agarose structures, or by adsorbing to polyolel.n surfaces (with or without glutaraldehyde cross-linking).
  • proteins Once bound to a substrate, proteins may be used to selectively bind (for purposes of assay or purification) antibodies raised against the CD83 protein or against other proteins which are similar to the CD83 prot in.
  • the present invention also includes CD83 proteins with or without associated native-pattern glycosylation.
  • Proteins expressed in yeast or mammalian expression systems e.g., COS-7 ce lls, may be similar or slightly different in molecular weight and glycosylation pattern tha.i the native molecules, depending upon the expression system.
  • Expression of CD83 LNA's in bacteria such as E. coli provides non-glycosylated molecules.
  • Functional mutant analogs of CD83 protein having inactivated N-glycosylation sites can be produced by oligonucleotide synthesis and ligation or by site-specific mutagenesis techniques These analog proteins can be produced in a homogeneous, reduced-carbohydrate foi m in good yield using yeast expression systems.
  • N-glycosylation sites in eukaryotic proieii s are characterized by the amino acid triplet Asn-A . -Z, where Al is any amino acid excepi Pro, and Z is Ser or Thr.
  • asparagine provides a side chain amino group for covalent attachment of carbohydrate.
  • Such a site can be eliminated by substituting another amino acid for Asn or for residue Z, deleting Asn or Z, or inserting a non-Z amino acid between Al and Z, or an amino acid other than Asn between Asn and A i .
  • CD83 protein derivatives may also be obtained by mutations of the DNA encoding native CD83 protein or ii •. subunits.
  • An CD83 mutated protein as referred to herein, is a polypeptide homologous to a CD83 protein but which has an amino acid sequence different from the native CD83 protein because of one or a plurality of deletions, insertions or substitutions.
  • the effec. of any mutation made in a DNA encoding a CD83 peptide may be easily determined by analyzing the ability of the mutated CD83 peptide to bind antibodies to CD83, or by analyzing ;ne ability of the CD83 mutein to stimulate secretion of antibody classes characteristic of a secondary immune response as described herein
  • Bioequivalent analogs of CD83 proteins may be constructed by, for example, making various substitii.ions of residues or sequences or deleting terminal or internal residues or sequences n:-t needed for biological activity.
  • cysteine residues can be deleted or replied with other amino acids to prevent formation of incorrect intramolecular disulfide bridges upon renaturation.
  • Other approaches to mutagenesis involve modification of a Jjacent dibasic amino acid residues to enhance expression in yeast systems in which KEX2 protease activity is present.
  • substitutions should be made conservatively; i.e., the most preferred substitute amino acids ;uc those which do not affect the ability of the CD83 proteins to bind CD83 antibodies, or to stimulate secretion of antibodies from human cells.
  • conservative substitutions include substitution of amino acids outside of the binding doma ⁇ n(s), and substitution of amino acids that do not alter the secondary and or tertiary structure of CD83. Additional examples include substituting one aliphauc residue for another, such as lie, 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 Gin and Asn.
  • Other such conservanve substitutions for example, substitutions of entire regions having similar hydrophobicity charactei istics, are well known.
  • Subunits of CD83 proteins may be constucted by deleting terminal or internal residues or sequences. Additional guidance as to the types of mutations that can be made is provided by a compa ⁇ son of the sequence of CD83 to the sequences and structures of other lmmunoglobuhn supertamily members
  • Mutations in mi. leotide sequences constructed for expression of analog CD83 proteins must, of cours- preserve the reading frame phase of the coding sequences and preferably will not creat complementary regions that could hybridize to produce secondary mRNA structures such a- loops or hairpins which would adversely affect translation of the receptor mRNA Althoigh a mutation site may be predetermined, it is not necessary that the nature ot the mutatio.i per se be predetermined. For example, in order to select for optimum characteristics ⁇ f mutants at a given site, random mutagenesis may be conducted at the target codon and the expressed mutated CD83 proteins screened for the desired activity
  • DNA's that enccde any of the forgoing CD83 peptides will also be useful in stimulating a humoial immune response, as will other CD83 DNA's.
  • nucleoti ie sequence which encodes a CD83 protein will be expressed in the final product, tor example, nucleotide substitutions may be made to enhance expression, primarily u avoid secondary structure loops in the transcribed mRNA (see EPA 75,444A, incorporited herein by reference), or to provide codons that are more readily translated by the 'elected host, e.g., the well-known E. coli preference codons for E. coli expression.
  • Mutations can r introduced at pa ⁇ icular loci by synthesizing oligonucleotides containing a mutant se ⁇ .-nee, flanked by restneuon sites enabling ligauon to fragments of the native sequence. Fo lowing ligauon, the resulting reconstructed sequence encodes an analog having the desired amino acid inse ⁇ ion, substitution, or deletion.
  • oh;,onucleotide-d ⁇ rected site-specific mutagenesis procedures can be employed to provide an altered gene having particular codons altered according to the substitution, deletion, oi insertion required. Exemplary methods of making the alterauons set fo ⁇ h above are disclosed by Walder et al.
  • DNA's capable of hybridizing under moderately stringent conditions prewashing solution of 5 X SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0) and hybridization conditions of 50°C, 5 X SSC, overnight
  • DNA's hybridizing under stringent conditions represent a preferred embodiment.
  • D83 peptides are at least about 70% identical in amino acid sequence to the amino a ⁇ id sequence of CD83 as set forth in SEQ ID NO:l.
  • analogs of CD83 proteins are at least about 80% identical in amino acid sequence to the na ⁇ i ⁇ e form of the proteins.
  • Percent identity may be determined using a computer program, for example, the GAP computer prograir Jescribed by Devereux et al. (Nucl. Acids Res. 12:387, 1984) and available from the Univ ;rsity of Wisconsin Genetics Computer Group (UWGCG). For fragments derived from the CD83 protein, the identity is calculated based on that portion of the CD83 protein that is present in the fragment.
  • Purified CD83 p oteins or analogs are prepared by culturing suitable host/vector systems to express the recombinant translation products of the DNA's described herein, which are then purified from culture media or cell extracts.
  • supernatants from systems which secrete recombinant protein into culture media can be first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pell icon ltrafiltratio ⁇ unit.
  • a suitable affinity matrix can comprise a CD83 antibody molecule bound to a su. table support.
  • an anion exchange resin can be employed, for example. : ⁇ matrix or substrate having pendant diethylaminoethyl (DEAE) groups.
  • the matrices .an be acrylamide, agarose, dextran, cellulose or other types commonly employed in protein purification.
  • a cation exchange step can be employed.
  • Suitable caiion exchangers include various insoluble matrices comprising sulfopropyl or carboxyrr ethyl groups. Sulfopropyl groups are preferred.
  • Gel filtration chromatography also provides a means of purifying CD83 proteins.
  • Affinity chromatography is a pamcularly preferred method of purifying CD83 proteins.
  • a CD83 protein expressed as a fusion protein comprising an immunoglobulin Fc region can be purified using Protein A or Protein G affinity chromatography
  • a CD83 protein comprising an oligomerizing zipper domain may be punfied on a resm comprising an antibody specific to the oligomerizing zipper domain.
  • Monoclonal antibodies against a CD83 protein may also be useful in affinity chromatography purificaiion, by utilizing methods that are well-known in the art.
  • HPLC steps employing hydrophobic RP-HPLC media, e.g., silica gel having pendant methyl or other aliphatic groups, can be employed to further purify a viral protein composition
  • hydrophobic RP-HPLC media e.g., silica gel having pendant methyl or other aliphatic groups
  • Some or all of the foregoing purification steps, in va ⁇ ous combinations, can also be employed to provide a homogeneous recombinant protein.
  • Recombinant CDS3 protein produced in bacte ⁇ al culture is usually isolated by initial extraction from cell pellets, followed by one or more concentration, salting-out, aqueous ion exchange oi size exclusion chromatography steps. Finally, high performance liquid chromatography (HPLC can be employed for final purification steps.
  • Microbial cells employed in expression of recombinant CD83 protein can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents Femientation of yeast which express CD83 protein as a secreted protein greatly simplifies purification Secreted recombinant protein resulting from a large-scale fermentation can be purried by methods analogous to those disclosed by Urdal et al. (J.
  • CD83 protein synihesized in leco binant culture is characterized by the presence of non-CD83 cell components, including proteins, in amounts and of a character which depend upon the punlic ition steps taken to recover the viral protein from the culture.
  • These components ordinarily will be of yeast, prokaryotic or non-human higher eukaryotic o ⁇ gin and preferably are present in innocuous contaminant quantities, on the order of less than about 1 percent by . eight. Fu ⁇ her, recombinant cell culture enables the production of CD83 protein free of cttier proteins which may be normally associated with the CD83 protein as it is found in I iture in its species of origin.
  • Useful CD83 O A s may be punfied by any suitable method of pu ⁇ fying DNA's known in the an Seve.al useful methods are described in Sambrook et al. (Molecular
  • DNA is amplified in prokaryotic cells, and isolated by a tandard alkaline lysis procedure followed by resin pu ⁇ ficauon as desc ⁇ bed in standard ki (tor example, from Promega Biotec, Madison, WT, or Quiagen, Chatswo ⁇ h, CA) The isolated DNA is then resuspended in a suitable diluent or earner
  • the present invention provides methods of using therapeudc compos ons comprising an effective amount of a CD83 reagent and a suitable diluent and earner, and methods for regulating mi immune response
  • CD83 proteins in conjunction with soluble cytokine re, ⁇ ptors or cytokines, or other immunoregulatory molecules is also contemplated CD83 DNA's and/or proteins are administered for the purpose of stimulating a humoral immune response
  • CD83 protein c mpositions administered to stimulate a humoral immune response can be given by bolus n lection, continuous infusion, sustained release from implants, or other suitable technique
  • a therapeutic agent will be administered in the form of a composition comprising purified CD83 protein in conjunction with physiologically acceptable earners, excipients or diluents Such earners will be nontoxic to recipients at the dosages and concentratio is employed
  • the of such CD83 protein compositions entails combining the CD83 piotein w ith butters, antioxidants such as ascorbic acid, low molecular weight (less than about 10 residu es) polypeptides, proteins, amino acids, carbohydrates including glucose, sucrose or deu ⁇ ns, chelating agents such as EDTA, glutathi
  • DNA vaccines provide the potential for long-term expression of antigens, with commensurate dilution of an immune response Simple saline solutions appear to be suitable earners foi DN ⁇ and various routes of administration have been shown to be useful, including v iamuscular (Ulmer et al , Science 259 1745, 1993, Montgomery et al , DNA Cell Biol 12 777, 1993) oi inuadermal injection (Raz et al., Proc Natl Acad. Sci USA 91 951 l) .
  • cytokines for use is stimul mng a ce ⁇ ain type of immune response
  • administration of other cytokines along with either CD83 DNA or CD83 protein is also contemplated.
  • cytokines or peptide regulatory factors
  • Such factors include (alone or in combination) Interleukins 1, 2, 4, 5, 6, 7, 10, 12 and 1 , granulocyte-macrophage colony sumulaung factor, granulocyte colony stimulating factor, a fusion protein comprising Interleuk ⁇ n-3 and granulocyte- macrophage colony stimulating factor, Interferon-y, TNF, TGF- ⁇ , flt-3 ligand and biologically active derivatives thereof
  • a pa ⁇ icularly prefened cytokine is CD40 ligand (CD40L)
  • a soluble r,., ⁇ n of CD40L is desc ⁇ bed in USSN 08/484,624, filed June 7, 1995 DNA encoding such cytokines
  • Antagonists of CD83 will be useful in inhibiting a humoral immune response
  • Exemplary conditions in which it is advantageous to inhibit such undesirable responses include autoimmune syndiomes, including myasthenia gravis, multiple sclerosis and systemic lupus erythe ⁇ ⁇ iosis, and others as desc ⁇ bed in U S Patent 5,284,935
  • This example describes construction of an HB 15 (CD83) DNA construct to express a soluble CD83 protein Using conventional techniques of PCR amphficauon, enzyme cutting and hgation, sceral CD83-encod ⁇ ng constructs were prepared, including one encoding a CD83/Fc his, on protein, a Flag ® /CD83 protein, and a soluble form of CD83
  • CD83/Fc Type I A soluble form of CD83 from amino acid -19 to amino acid 124 of SEQ ID NO:l, and a Flag® form consisting of amino acid -19 to amino acid 124 of SEQ ID NO:l linked to the eight amino acid sequence described by Hopp et al. (BiolTechnology 6:1204,1988; SEQ ID NO:5) were prepared in an expression vector (pDC304, which is derived from pDC302, described by Mosley et al., Cell, 59:335 (1989), by deleting the adenovirus tripa ⁇ ite leader (TPL) in pDC302).
  • pDC304 which is derived from pDC302, described by Mosley et al., Cell, 59:335 (1989), by deleting the adenovirus tripa ⁇ ite leader (TPL) in pDC302).
  • the resultant expression vectors are transfected into the monkey kidney cell line CV-1/EBNA (ATCC CRL 10478). Large scale cultures of CV-1/EBNA cells transfected with the various constructs are grown to accumulate supernatant containing the different forms of CD83 protein
  • the CV-1/EBNA cell line permits expression of recombinant proteins ligated into veciors containing the EBV origin of replication.
  • the CD83 proteins in supernatant fluid are p.irified as described below.
  • the Flag ® /CD83 protein was uirified by affinity chromatography. Briefly, culture supernatant containing the Flag*/CDi;3 protein was purified by filtering mammalian cell supernatants (e.g., in a 0.45 ⁇ filter) an j applying filtrate to an affinity column comprising a monoclonal antibody specific for the Flag ® peptide, coupled to Affi-gel active ester agarose (Bio-Rad, Richmond, CA), at room temperature, at a flow rate of approximately 60 to 80 ml/hr for a 1.5 cm x 12.0 cm column.
  • mammalian cell supernatants e.g., in a 0.45 ⁇ filter
  • Affi-gel active ester agarose Bio-Rad, Richmond, CA
  • CD83/Fc protein 's purified by conventional methods using Protein A or Protein G chromatography.
  • Approximately one liter of culture supernatant containing CD83 protein is purified by filtering n ⁇ mmalian cell supernatants (e.g., in a 0.45m filter) and applying filtrate to a protein A/G antibody affinity column (Schleicher and Schuell, Keene, NH) at 4 * C at a flow rate of 80 I il/hr for a 1.5 cm x 12.0 cm column.
  • the column is washed with 0.5 M NaCl in PBS until free protein is not detected in the wash buffer.
  • the column is washed with PBS.
  • Bound fusion protein is eluted from the column with 25 mM citrate buffer, pH 2.8, and brought to pH 7 with 500 mM Hepes buffer, pH 9.1.
  • nets can be prepared and the expressed protein purified using methods that are known in the art.
  • a CD83 protein comprising a poly-His peptide may be detected and/or purified using a poly-His system, substantially as described in US Patent 5,284,933, issued February 8, 1994.
  • Biological activity is measured in any biological assay which quantifies an antigen-specific immun response, for example, as described in the Examples herein.
  • mice On day 0, 6 BALB/c mice were injected subcutaneously with 200 ⁇ l of a suspension containing 4 ⁇ g of ovalbumin (OVA). On day 14, they were injected with either 500 ⁇ g of CD83/Fc or 500 rat IgG as a control. Six hours later they were re-immunized with 1 ⁇ g ovalbumin. The mice were bled at day 21 and titers of ovalbumin-specific antibodies of various subclasses were determined by ELISA. The mice that were given CD83/Fc prior to the secondary immunization exhibited slightly higher levels of certain subclasses of ovalbumin-specific antibodies, particularly IgG2b-
  • the example illustrates the ability of DNA encoding CD83 to stimulate a primary humoral immune response with high levels of IgG2t > - BALB/c mice (six per group) were injected with 50 ⁇ g of DNA comprising DNA encoding a tumor necrosis factor/immunoglobulin I ' c fusion protein (described by Mohler et al., J Immunol 151 :1548.1993: and in EP 418014), along with either 50 ⁇ g CD83-encoding vector DNA or control vector DNA. Mice were bled at day 14 for determination of TNFr/Fc-specific antibody titers by ELIS ⁇ .
  • results are shown in table 1 below; titration curves for the TNFr/Fc-specific I G2h re shown in Figure 1.
  • T. ble End-point titers of TNFr/Fc-specific antibody in animals injected with TNFr/Fc protein and either control DNA or CD83 DNA*
  • Naive B cells are obtained by one of several methods known in the art.
  • surface IgD-positive tonsillar B cells, neonatal B cells obtained from cord blood and B cell obtained from individuals suffering from X-linked hyper-IgM syndrome represent populations of cells that are substantially devoid of isotype-committed B cells.
  • Mononuclear cells are isolated by a method such as centrifugation over Ficoll- Hypaque, and depleted of T cells by rosetting with 2-aminoethylisothiouronium bromide- treated SRBC (sheep reJ blood cells).
  • the resulting E" cells can be fu ⁇ her purified by negative selection using antibodies to cell surfaces markers found on non-B cells (i.e., CD2, CD3, CD 14), and/or by positive selection using antibodies to markers found on non- isotype committed B cells (i.e., slgD).
  • the cells are cultured under suitable conditions, and stimulated with cytokines to induce immunoglobulin secretion.
  • the cytokines to be used include a soluble vrimeric from of CD40 ligand (CD40L) as described in USSN 08/484,624, filed June 7. 1995: 1L-4, and IL- 10.
  • Other cytokines may also be included, for example, transforming growth factor ⁇ and IL-2.
  • CD83 stimulates secretion of IgG2 b from murine B cells under such conditions; it will likewise stimulate isotype switching and secretion of IgG2 from naive human B cells.
  • EXAMPLE 6 This example illustrates the preparation of monoclonal antibodies against CD83.
  • Preparations of purified r.combinant CD83/Fc, for example, or transfected cells expressing high levels of CD83 are employed to generate monoclonal antibodies against CD83 using conventional techniques, such as those disclosed in U.S. Patent 4,41 1,993.
  • Such antibodies are likely 10 t>. useful in interfering with CD83 binding or biological activity, as components of diagnostic or research assays for CD83, or in affinity purification of CD83.
  • CD83 immunogen is emulsified in an adjuvant (such as complete or incomplete Freund's adjuvant, alum, or another adjuvant, such as Ribi adjuvant R700 (Ribi, H. milton, MT), and injected in amounts ranging from 10- 100 ⁇ g subcutaneously into a se! ,-cted rodent, for example, BALB/c mice or Lewis rats.
  • an adjuvant such as complete or incomplete Freund's adjuvant, alum, or another adjuvant, such as Ribi adjuvant R700 (Ribi, H. milton, MT)
  • Ribi adjuvant R700 Ribi, H. milton, MT
  • Serum samples are periodically taken by retro-orbital bleeding or tail-tip excision for testing by dot-blot assay (antibody sandwich) or ELISA (enzyme-linked immunosorbent assay).
  • Other assay procedures are also suitable, for example, FACS analysis using cells expressing membrane-bound CD83.
  • positive animals are given an intravenous injection of antigen in saline Three to four d ⁇ ys later, the animals are sacnficed, splenocytes harvested, and fused to a mu ⁇ ne myeloma cell line (e.g., NS 1 or preferably Ag 8.653 [ATCC CRL 1580]).
  • a mu ⁇ ne myeloma cell line e.g., NS 1 or preferably Ag 8.653 [ATCC CRL 1580]
  • Hyb ⁇ doma cell lines generated by this procedure are plated in multiple microdter plates in a selective medium (for example, one containing hypoxanthine, a inopte ⁇ n, and thymidine, or HAT) to inhibit proliferation of non-fused cells, myeloma-myeloma hybrids, and splenocyte-splenocyie hybrids.
  • a selective medium for example, one containing hypoxanthine, a inopte ⁇ n, and thymidine, or HAT
  • Hyb ⁇ doma clon s thus generated can be screened by ELISA for reactivity with CD83, tor example, b adaptations ot the techniques disclosed by Engvall et al., Immunochem #-871 ( H71 ) and in U.S. Patent 4,703,004.
  • a preferred screening technique utilizes fluores ;ence activated cell somng to detect binding to cells that express CD83, for example, Ra cells Positive clones are then generated and injected into the pe ⁇ toneal cavities of sy lgeneic rodents to produce ascites containing high concentrations (>1 mg/ml) ot anti-CDS -> monoclonal antibody.
  • the resulting monoclonal antibody can be punfied by ammonium • ultate precipitation followed by gel exclusion chromatography.
  • affinity clnomatography based upon binding of antibody to protein A or protein G can also be us.d. as can affinity chromatography based upon binding to CD83 protein
  • EXAMPLE 7 This example de -. iibes two solid-phase binding assays, the first of which, (a), can be used to quantify sol ble CD83, and the second of which, (b), is used to detect the presence ot soluble CD8 ⁇
  • Antibody to CDK 1 is purified and used to coat 96-well plates (for example, Corning EasyWash ELISA plate 1 . Corning, NY, USA).
  • the plates arc coated for one hour at r om temperature with 50 ⁇ l of PBS containing 5 ⁇ g/ml of M43 (desc ⁇ bed m Example 6) and blocked with KK) ⁇ l/well of 5 % non-fat d ⁇ ed milk in PBS for 1 hour at room tempt ⁇ tuie.
  • Samples to be tested are diluted in 10% normal goat serum (NGS) in PBS.
  • Soluble CD83 ( 1 ⁇ i of c ⁇ ide supernatant or column fracnons) is adsorbed to dry BA85/21 nitrocellulose membranes (Schleicher and Schuell, Keene, NH) and allowed to dry.
  • the membianes ar, incubated in tissue culture dishes for one hour in Tris (0.05 M) buffered salme (0 15 V pH 7 5 containing 1 % w/v BSA to block nonspecific binding sites At the end ol this time, the membranes are washed three times in PBS, and rabbit ant ⁇ -CD83 antibod) is idded at an approximate concentration of 10 ⁇ g/ml in PBS containing ⁇ r /c BSA.
  • the membranes are incubated for one hour at room temperauue
  • the membianes ⁇ ie again washed as described, and a horseradish peroxidase (HRP)-l ⁇ beled antibody such as goat anti-rabbit lg, Southern Biotech, Birmingham, AL) at an approximate dilution ot 1 :1000 in PBS containing 1% BSA is added.
  • HRP horseradish peroxidase
  • chromogen i.e. 4-chloronaphthol re igent, Kukegard and Pe ⁇ y, Gaithersburg, MD
  • Peripheral blood B and T cells are obtained by any suitable method known in the an For example.
  • PBMCs are isolated from a healthy donor by centrifugation of heparu zed blood over Isolymph (Gallard-Schlesinger Industnes, Inc., Norway) and washed tl ree times (i.e., with culture medium consisting of RPMI 1640 supplemented with 10 , ⁇ FCS, 50 U/ml penicillin, 50 ⁇ g ml streptomycin, 2mM glutamine).
  • Isolated PBMCs are separated into T cell and non-T-cell fractions by rosetting with 2-aminoethylisothiouronium bromide (AET)-treated sheep red blood cells. Twice rosetted cells are suspended in RPMI 1640 culture media with 10 % FCS and incubated on plastic dishes for 1 hr at 37°C to remove any remaining adherent cells. The resulting cell preparations were always at least 90% T cells (98% CD2 + ; 90% CD3 + ) as determined by flow cytometric analysis B cells are fu ⁇ her purified from the E" preparation by positive selection using CD 19 monoclonal antibody (mAb) on a MACS column (Miltenyi Biotec, Sunnyvale, CA) according to the manufacturers instructions. CD19 + B cells purified in this way were routinely :-95% pure as determined by reactivity with CD20 mAb.
  • mAb monoclonal antibody
  • Peripheral blood T or B cells were cultured at lxlO 6 in 1ml for 48 or 24 hours with the following stimuli: immobilized CD3 mAb, 5 ⁇ g/ml; PHA, 1%; IL-2, lOng/ml; PMA, lOng/ml; Ionomycin, 500ng/ml: IL-4, lOng/ml; soluble trimeric CD40L, 2 ⁇ g/ml; SAC, 0.01 %. Levels of soluble CD83 in culture supernatants were measured by ELISA A protease inhibitor, TAP!
  • soluble CD83 was expressed as a result of a proteolytic cleavage of membrane-bound CD83.
  • B cells produced larger quantities of soluble CD83 than did T cells, and for both cell types, certain stimuli caused shedding of greater quantities of CDH than did other stimuli.
  • Isotype-switched B cells are obtained by one of several methods known in the ar..
  • surface IgD-negative B cells are fu ⁇ her purified from peripheral mononuclear cells (isolated by a method such as centrifugation over Ficoll- Hypaque, and depleted of T cells by rosetting with 2-aminoethylisothiouronium bromide- treated SRBC) and depleting the E- cells of IgD + B cells (i.e., by positive selection on a MACS column).
  • the resulting cells can also be fu ⁇ her purified by positive selection for a B cell marker such as CD 19.
  • IgD B cells may also be obtained from tonsils, using methods that are known in the art (for example, Liu et al., Eur. J. Immmunol. 21 :1107, 1991 ; or Lagresle et al.. .' «. Immunol. 5: 1250, 1993)
  • IgD peripheral ood B cells (5x1 Orwell) were cultured for twelve days in the presence of soluble, trimcrie CD40L (2 ⁇ g/ml) and IL-2 (lOng/ml); IL-6 (lOng/ml) or IL-10

Abstract

There is disclosed a method of stimulating an antigen-specific humoral immune response. Useful vaccine compositions are also disclosed.

Description

TITLE
METHODS AND COMPOSITIONS FOR MODULATING AN IMMUNE RESPONSE
TECHNICAL FIELD OF THE INVENTION
The present invention relates generally to the field of rnarnmalian proteins having immunoregulatory activity, and more specifically to mammalian proteins involved in regulation of a humoral immune response.
BACKGROUND OF THF INVENTION
CD83 is a member of the immunoglobulin superfamily that is expressed on the surface of certain dendriπc lineage cells and some lymphoblastoid cell types (Zhou et al., J. Immunol. 149:735. 19S2: Zhou et al., ./. Immunol. 154:3821 , 1995). The presence of CD83 on dendritic cells lias led to the hypothesis that it is somehow involved in antigen presentation; however, prior to the present invention, no biological functions were known for CD83.
Vaccination is ar; efficient means of preventing death or disability from infectious diseases. Despite the successes achieved with the use of vaccines, however, there are still many challenges in the t.eld of vaccine development. Parenteral routes of administration, the numbers of different vaccinations required and the need for, and frequency of, booster immunizations all impeαe efforts to control or eliminate disease. Moreover, inability to modulate the type of response, and isotype of antibody made, during immunization has hampered vaccination programs. Although numerous vaccine adjuvants are known, alum is the only adjuvant widely used in humans.
Thus, prior to t! :< present invention, there was a need in the art to determine the function of CD83. Tht te was furthermore a need to develop agents useful in stimulating secretion of antibody, . > develop effective methods of immunization, and to discover alternative tvpes of ants, suitable for use in humans.
SUMMARY OF THE INVENTION
The present inv.ntion provides a method of stimulating a humoral immune response, comprising administering a CD83 reagent and an antigen, in a pharmaceutically acceptable carrier, wherein the CD83 stimulates production of antigen-specific antibodies
Useful CD83 reagents include DNA's encoding CD83 and CD83 polypeptides, as well as derivatives and analogs of such reagents that have CD83 biological activity. The present invention further provides vaccine compositions useful in stimulating a humoral immune response.
CD83 DNA's that are useful in the inventive methods and compositions include a DNA having a nucleotide sequence encoding an amino acid sequence of amino acids 1 through 124 of SEQ ID NO: 2 and DNA molecules capable of hybridization to such DNA under stringent conditions and which encode biologically active CD83. Useful CD83 proteins include a CD83 peptide having an amino acid sequence of amino acids 1 through 124 of SEQ ID NO: 2, fragments of such a peptide according that have CD83 biological activity; and peptides encoded by DNA molecules capable of hybridization to a DNA encoding such peptide under stringent conditions, and which encode biologically active CD83. In a preferred embodiment, CD83 reagents are DNA's that encode, and CD83 peptides that comprise, the extracellular domain of CD83.
Another aspect of the inventive methods and compositions involves administering a cytokine that modulates an immune response in conjunction with a CD43 composition (either sequentially, simultaneously or separately), particularly cytokines selected from the group consisting of In.eπeukins 1 , 2, 4, 5, 6, 7, 10, 12 and 15; granulocyte-macrophage colony stimulating factor, granulocyte colony stimulating factor; a fusion protein comprising Interleukin and granulocyte-macrophage colony stimulating factor, Interferon-γ, TNF; TGF-IΪ; flt-3 ligand; soluble CD40 ligand; biologically active derivatives of these cytokines; and combinations thereof.
In studies performed using antibodies to CD83, the antibodies inhibited various antigen specific responses. The present invention thus also provides a method of inhibiting undesirable antigen specific responses in a mammal. Such methods of inhibiting undesirable antigen specific responses are useful in preventing or treating autoimmune disease as well as tissue or organ transplant rejection, and in treatment or prevention of allergy or asthma.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 demonstrates that mice immunized with antigen (DNA encoding TNFr Fc) in the presence of CD8? have significantly higher serum titers of TNFr/Fc-specific IgG2b than mice immunized wuh antigen alone.
Figure 2 illusrrat-S the ability of CD83 to stimulate higher levels of antigen-specific IgG2b when the antigen used is a soluble protein antigen (TNFr/Fc).
DETAIL ED DESCRIPTION OF THE INVENTION
CD83 was cloned from a Raji cell library by polymerase chain reaction, using primers based on the p iblished sequence (Zhou et al., J. Immunol. 149:735, 1992). Several different soluble forms of CD83 were expressed, including a Type I Fc/CD83 fusion protein, a Flag®/C1 83 fusion protein, and a soluble form of CD83 consisting of the extracellular domain. A detailed explanation of the experimental results and their relevance to the instant invention, along with ceπain technical background information, are given below.
CD83 HB15
CD83 (also referred to as HB15) is a 45KD glycoprotein predominantly expressed on the surface of dendritic lineage cells, such as skin Langerhans cells and interdigitating reticulum cells present in the T cell zones of lymphoid organs. It is also weakly expressed by some lymphoblastoid cell types, and can be upregulated under certain activation conditions. Structural analysis of the predicted amino acid sequence of this protein established it as a me .r of the immunoglobulin superfamily (Zhou et al., J. Immunol. 149:735, 1992). It has more recently been shown that human blood dendritic cells express CD83 (Zhou et al., ./. I.nmunυl. 154:3821, 1995). U.S. Patent 5,316,920, issued May 31, 1994, discloses and claims DNA's encoding CD83; WO93/21318 is a corresponding published internationai patent application. WO 95/29236 discloses related proteins and DNA's encoding them as well as antibodies reactive with these proteins.
Although the presence of CD83 on dendritic cells has led to the hypothesis that it is somehow involved in antigen presentation, prior to the present invention, no biological functions were known for this protein. The discovery that CD83 stimulates production of antibodies led to the inve itive uses and compositions described herein. Because of its role, CD83 (both in protein foιτn and in DNA form) will be useful as a vaccine adjuvant. CD83 can be administered in ivnjunction with other immunomodulatory molecules, as described herein. Moreover, DNΛ ending CD83 can be incorporated into attenuated live viral or bacterial vaccine strain ,, to enhance the immune response to the infectious agent. Additionally, antagonist s of CD83 will be useful in suppressing an undesirable, antigen- specific immune respon ,e.
The protective immune r. sponse An immune response to a pathogen can be classified broadly as either being cell- mediated (cellular immunity) or antibody mediated (humoral immunity). In cellular immunity, activated mac ophages and cytotoxic lymphocytes carry out elimination of the pathogen. Humoral immunity, in contrast, operates primarily through antibody production. It is currently believed that these two arms of the immune response are regulated by distinct su isets of helper T (TH) cells which secrete specific arrays of cytokines (reviewed in Immunological Reviews 123, 1991).
Type 1 TH cells (TH I cells) mediate delayed type hypersensitivity (DTH), and secrete Interferon-γ (IF,' -γ) and Interleukin-2 (IL-2), while Type 2 TH cells (TH2 cells) secrete primarily Interleukins 4, 5 and 10 (IL-4, IL-5 and IL-10, respectively) and provide B cell help. Development of the immune response along either TH1 or TH2 pathway is often apparent early in an infection, and appears to be governed by the type of organism causing the infection (Scott and Kaufmann, Immunol. Today 12:346, 1991), and by the genetic makeup of the infected host. Failure to resolve disease or development of immunopathology can result when the immune response proceeds inappropriately.
The immune response may be manipulated toward either a THI or TH2 by the appropriate administration of cytokines, or cytokine antagonists. For example, administration of IFN-γ or an antibody that neutralizes IL-4 would enhance a THI response, whereas administration of IL-10 or a molecule that inhibited the action of IFN-γ would stimulate a TH2 response. This ability to manipulate the immune response provides a useful tool not only in infectious disease, but in inflammatory and allergic diseases as well (see, for example, owπe and Cotfman, Immunol. Today 14:270, 1993).
Early antibody π. -.ponses. both in the life cycle of an animal and in the ontogeny of individual B cell clones primarily consist of IgM. Under the control of helper T cells, the isotype of antibody pro uced by B cells is switched from IgM to IgG, IgE or IgA. The latter isotypes are representative of a more mature immune response, and generally include antibodies of higher affinity and avidity as well as increased effector function. Stimulation of non-IgM isotypes is considered a desirable effect of any vaccination protocol, since it is the IgG, IgE and IgA aim bodies that are generally protective against infectious disease, and which are likely to play a role in tumor immunity. Moreover, the IgG subclasses are preferred for the generation of monoclonal antibodies, since these exhibit useful characteristics (i.e., easier purification, higher affinity, greater therapeutic effectiveness due to enhanced effector tun tions).
Vaccines and disease
Immunization I , a centuries old, and highly effective, means of inducing a protective immune res p. use against pathogens in order to prevent or ameliorate disease. The vaccines that have been used for such induction are generally live, attenuated microorganisms, or preparations of killed organisms or fractions thereof. Live, attenuated vaccines are generally thought to more closely mimic the immune response that occurs with a natural infection than do those prepared from killed microbes or non-infective preparations derived ti >m pathogens (i.e., toxoids, recombinant protein vaccines). However, attenuated vaccines also present a risk of reversion to pathogemcity, and can cause illness, especially M immunocompromised individuals.
Along with impn ved sanitation, immunization has been the most efficient means of preventing death or disability from numerous infectious diseases in humans and in other animals. Vaccination of susceptible populations has been responsible for eliminating small pox world wide, and for drastic decreases in the occurrence of such diseases as diphtheria, pertussis, and paralytic polio in the developed nations. Numerous vaccines are licensed for administration to humans, including live virus vaccines for certain adenoviruses, measles, mumps and rubella viruses, and poliovirus, diphtheria and tetanus toxoid vaccines, and Haemophilus b and meirngococcal polysaccharide vaccines (Hinman et al., in Principles and Practice of Infectious Diseases. 3rd edition; G.L. Mandell, R.G. Douglas and J.E. Bennett, eds., Churchill Livingstone Inc., NY, NY; 2320-2333; Table 2).
Despite the successes achieved with these vaccines, however, there are still numerous challenges in :he field (Science 265:1371; 1994). HTV infection is a public health problem in both developed and developing nations; there has been little progress in developing an effective \ iccine against this virus despite significant research efforts in this area. Malaria and tub.'rculosis represent significant public health challenges in the developing world, with high morbidity and mortality rates, and problematic treatment regimes. Respiratory s/ncytial virus (RSV) and pneumococcal disease pose similar difficulties in the developed world.
Even for disease; for which there are effective vaccines available, maintaining an sufficient rate of immunization in susceptible populations presents a public health challenge. Many children in the United States are not vaccinated for common childhood diseases such as diphthe ria and pertussis. Adults may not receive necessary boosting immunizations for tetar. is or other diseases. Parenteral routes of administration, the numbers of different vaccinations required and the need for, and frequency of, booster immunizations all imped- efforts to achieve patient compliance with vaccine programs. Developing countries al .o face additional challenges in trying to store and administer vaccines. Several aspects of vaccine preparation and administration have been investigated.
These include route of ac inistration and encapsulation of antigen preparations to provide sustained release of the antigen (see, for example, USSN 08/508,229, filed July 27, 1995), and the use of ad' ivants. Useful adjuvants include for example alum, fragments of bacterial membranes, l osomes, coupling a protein of interest to a larger immunogenic protein, RIBI, non-ion < block co-polymer surfactants and TiterMax®. Other useful vaccine adjuvants and e xcipients are described by Vogel and Powell (A Compendium of Vaccine Adjuvants and Excipients. in: Vaccine Design, Powell and Newman, eds.; Plenum Publishing Corporation. MY, NY: 1994). Of these, alum is the only adjuvant widely used in humans. Other areas of i iterest in the field of vaccination are the use of cytokines to modulate an immune re:- >onse. Some cytokines, e.g., interleukin-4 (IL-4) and GM-CSF, attract and activate antigen-presenting cells for more efficient presentation of antigens to T cells. These cytokines have been co-administered with antigen to increase antigenic activity Other studies have shown that the host response to tumor challenge can be increased by inoculation of tumor cells genetically engineered to express particular cytokines, including γ-H"N, TNF-α, IL-2, IL-4, IL-6, IL-7, or GM-CSF Recombinant antigens have been expressed as fusion proteins with cytokines, for example as described in USSN 08/271,875, fil-d July 7, 1994
The use of "naked DNA" represents one of the newest approaches to vaccination (Pardoll and Beckerlei' Immunity 3 165, 1995) The utility of DNA in vaccine preparations rests upon ι .e ability of purified DNA to be taken up and expressed by cells in vivo with much greater efficiency than is seen in vitro Large scale producnon of DNA is relatively simple, and thi resulting DNA can be readily purified to a very great degree, reducing the potential for dangerous contaminants. Moreover, purified DNA is much more stable than purified proteins and other biological mateπals, which can ameliorate storage and administration prob ms
DNA's. Proteins and An ogs
The present invention provides isolated CD83 DNA s and proteins (referred to as CD83 agents) having .mmunoregulatory activity Such DNA's and proteins are substantially free of contaminating endogenous mateπals and, opnonally, without associated native-pattern glycosylation Deπvatives of the CD83 proteins within the scope of the invention also ιn< ude various structural forms of the pπmary protein which retain biological activity Due to the presence of lonizable amino and carboxyl groups, for example, a CD83 proteir may be in the form of acidic or basic salts, or may be in neutral form Individual ammo «ιcιd residues may also be modified by oxidation or reduction.
The primary ammo acid structure may be modified by forming covalent or aggregative conjugates with other chemical moieties, such as glycosyl groups, lipids, phosphate, acetyl groups and the like, or by creaung a ino acid sequence mutants. Covalent deπvatives are prepared by linking paπicular functional groups to amino acid side chains or at the N- or C-t.rmini
Other deπvatives of the CD83 protein within the scope of this invention include covalent or aggiegative -onjugates of the protein or its fragments with other proteins or polypeptides, such as , synthesis in recombinant culture as N-terminal or C-terminal fusions For example, the conjugated peptide may be a signal (or leader) polypepude sequence at the N-teπ inal region of the protein which co-translationally or post- translationally directs uansfer of the protein from its site of synthesis to its site of funcuon inside or outside ot the c< 11 membrane or wall (e.g , the yeast α-factor leader).
Protein fusions can comprise peptides added to facilitate punficauon or identification of CD83 | oteins (e g , poly-His) The amino acid sequence of the CD83 proteins can also be linl .d to an identification peptide such as that descπbed by Hopp et al., Bio/Technology 6: 1-04 ( 1988). Such a highly antigenic peptide provides an epitope reversibly bound by a .pecific monoclonal antibody, enabling rapid assay and facile purification of expressed recombinant protein.. The sequence of Hopp et al. is also specifically cleaved by bovine mucosal enterokinase, allowing removal of the peptide from the purified protein. Fusion proteins capped with such peptides may also be resistant to intracellular degradation n E. coll.
Fusion proteins fuπher comprise the amino acid sequence of a CD83 protein linked to an immunoglobulin Iv region. An exemplary Fc region is a human IgGl having a nucleotide and amino acd sequence set foπh in SEQ ID NO:3. Fragments of an Fc region may also be used. Depending on the portion of the Fc region used, a CD83 protein may be expressed as a dimer, through formation of interchain disulfide bonds. If CD83 fusion proteins are made with both heavy and light chains of an antibody, it is possible to form a protein oligomer with as many as four CD83 protein regions.
In another emboc iment, CD83 proteins fuπher comprise an oligomerizing zipper domain. Oligomerizing zipper domains are described in USSN 08/107,353, filed August 13, 1993, the relevant disclosure of which is incorporated by reference herein. Examples of leucine zipper domains are those found in the yeast transcription factor GCN4 and a heat-stable DNA-bindinj; protein found in rat liver (C EBP; Landschulz et al., Science 243: 1681, 1989), the nut lear transforming proteins,/os and jun, which preferentially form a heterodimer (O'Shea et al., Science 245:646, 1989; Turner and Tjian, Science 243:1689, 1989), and the gene product of the murine proto-oncogene, c-myc (Landschulz et al., Science 240: 1759, 198!< ). The fusogenic proteins of several different viruses, including paramyxovirus, coronavirus, measles virus and many retroviruses, also possess leucine zipper domains (Buckland and Wild, Nature 338:547, 1989; Britton, Nature 353:394, 1991; Delwart and Mosialos, AIDS Research and Human Retroviruses 6:703, 1990).
Other useful fusion proteins include fusions of CD83 with an antigen against which it is desired to elicit an ir. mune response, for example as described in USSN 08/271,875, filed July 7, 1994. for GM-CSF. Similarly, fusion proteins consisting of CD83 and another cytokine or cyto* ines are also contemplated. As shown herein for CD83 DNA's, the DNA's encoding such fusion proteins will also have utility in the instant invention. A very useful DNA may ii elude not only sequences encoding CD83 and another cytokine (for example, CD40L). but also sequences encoding the antigen(s).
CD83 protein -erivatives may also be used as immunogens, reagents in immunoassays, or as binding agents for affinity purification procedures, for example, in purifying CD83 antibodies. CD83 protein derivatives may also be obtained by cross- linking agents, such as M-maleimidobenzoyl succinimide ester and N-hydroxysuccinimide, at cysteine and lysitie re sidues. CD83 proteins may also be covalently bound through reactive side croups to various insoluble substrates, such as cyanogen bromide-activated, bisoxirane-activated, car-tonyldiimidazole-activated or tosyl-activated agarose structures, or by adsorbing to polyolel.n surfaces (with or without glutaraldehyde cross-linking). Once bound to a substrate, proteins may be used to selectively bind (for purposes of assay or purification) antibodies raised against the CD83 protein or against other proteins which are similar to the CD83 prot in.
The present invention also includes CD83 proteins with or without associated native-pattern glycosylation. Proteins expressed in yeast or mammalian expression systems, e.g., COS-7 ce lls, may be similar or slightly different in molecular weight and glycosylation pattern tha.i the native molecules, depending upon the expression system. Expression of CD83 LNA's in bacteria such as E. coli provides non-glycosylated molecules. Functional mutant analogs of CD83 protein having inactivated N-glycosylation sites can be produced by oligonucleotide synthesis and ligation or by site-specific mutagenesis techniques These analog proteins can be produced in a homogeneous, reduced-carbohydrate foi m in good yield using yeast expression systems. N-glycosylation sites in eukaryotic proieii s are characterized by the amino acid triplet Asn-A .-Z, where Al is any amino acid excepi Pro, and Z is Ser or Thr. In this sequence, asparagine provides a side chain amino group for covalent attachment of carbohydrate. Such a site can be eliminated by substituting another amino acid for Asn or for residue Z, deleting Asn or Z, or inserting a non-Z amino acid between Al and Z, or an amino acid other than Asn between Asn and A i .
CD83 protein derivatives may also be obtained by mutations of the DNA encoding native CD83 protein or ii •. subunits. An CD83 mutated protein, as referred to herein, is a polypeptide homologous to a CD83 protein but which has an amino acid sequence different from the native CD83 protein because of one or a plurality of deletions, insertions or substitutions. The effec. of any mutation made in a DNA encoding a CD83 peptide may be easily determined by analyzing the ability of the mutated CD83 peptide to bind antibodies to CD83, or by analyzing ;ne ability of the CD83 mutein to stimulate secretion of antibody classes characteristic of a secondary immune response as described herein
Bioequivalent analogs of CD83 proteins may be constructed by, for example, making various substitii.ions of residues or sequences or deleting terminal or internal residues or sequences n:-t needed for biological activity. For example, cysteine residues can be deleted or replied with other amino acids to prevent formation of incorrect intramolecular disulfide bridges upon renaturation. Other approaches to mutagenesis involve modification of a Jjacent dibasic amino acid residues to enhance expression in yeast systems in which KEX2 protease activity is present.
Generally, substitutions should be made conservatively; i.e., the most preferred substitute amino acids ;uc those which do not affect the ability of the CD83 proteins to bind CD83 antibodies, or to stimulate secretion of antibodies from human cells. Examples of conservative substitutions include substitution of amino acids outside of the binding domaιn(s), and substitution of amino acids that do not alter the secondary and or tertiary structure of CD83. Additional examples include substituting one aliphauc residue for another, such as lie, 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 Gin and Asn. Other such conservanve substitutions, for example, substitutions of entire regions having similar hydrophobicity charactei istics, are well known.
Similarly, when a deletion or mseπion strategy is adopted, the potential effect of the deletion or insertion on biological activity should be considered. Subunits of CD83 proteins may be constucted by deleting terminal or internal residues or sequences. Additional guidance as to the types of mutations that can be made is provided by a compaπson of the sequence of CD83 to the sequences and structures of other lmmunoglobuhn supertamily members
Mutations in mi. leotide sequences constructed for expression of analog CD83 proteins must, of cours- preserve the reading frame phase of the coding sequences and preferably will not creat complementary regions that could hybridize to produce secondary mRNA structures such a- loops or hairpins which would adversely affect translation of the receptor mRNA Althoigh a mutation site may be predetermined, it is not necessary that the nature ot the mutatio.i per se be predetermined. For example, in order to select for optimum characteristics < f mutants at a given site, random mutagenesis may be conducted at the target codon and the expressed mutated CD83 proteins screened for the desired activity
DNA's that enccde any of the forgoing CD83 peptides will also be useful in stimulating a humoial immune response, as will other CD83 DNA's. For example, not all mutations in the nucleoti ie sequence which encodes a CD83 protein will be expressed in the final product, tor example, nucleotide substitutions may be made to enhance expression, primarily u avoid secondary structure loops in the transcribed mRNA (see EPA 75,444A, incorporited herein by reference), or to provide codons that are more readily translated by the 'elected host, e.g., the well-known E. coli preference codons for E. coli expression.
Mutations can r introduced at paπicular loci by synthesizing oligonucleotides containing a mutant se μ .-nee, flanked by restneuon sites enabling ligauon to fragments of the native sequence. Fo lowing ligauon, the resulting reconstructed sequence encodes an analog having the desired amino acid inseπion, substitution, or deletion. Alternatively, oh;,onucleotide-dιrected site-specific mutagenesis procedures can be employed to provide an altered gene having particular codons altered according to the substitution, deletion, oi insertion required. Exemplary methods of making the alterauons set foπh above are disclosed by Walder et al. (Gene 42: 133, 1986); Bauer et al. (Gene J7:73, 1985): Craik ( ioTechniques, January 1985, 12-19); Smith et al. (Genetic Engineering: Principles and Methods, Plenum Press, 1981); and U.S. Patent Nos. 4,518,584 and 4,737,462 disclose suitable techniques, and are incorporated by reference herein. Due to code degeneracy, there can be considerable variation in nucleotide sequences encoding the same amine acid sequence. Such degenerate CD83 DNA's will also be useful in the instant invention. Other embodiments include DNA's capable of hybridizing under moderately stringent conditions (prewashing solution of 5 X SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0) and hybridization conditions of 50°C, 5 X SSC, overnight) to the DNA sequences encoding CD protein. Conditions of higher stringency are known in the art; DNA's hybridizing under stringent conditions represent a preferred embodiment. In a preferred embodiment, D83 peptides are at least about 70% identical in amino acid sequence to the amino a< id sequence of CD83 as set forth in SEQ ID NO:l. In a most prefeπed embodiment, analogs of CD83 proteins are at least about 80% identical in amino acid sequence to the naιi\ e form of the proteins.
Percent identity may be determined using a computer program, for example, the GAP computer prograir Jescribed by Devereux et al. (Nucl. Acids Res. 12:387, 1984) and available from the Univ ;rsity of Wisconsin Genetics Computer Group (UWGCG). For fragments derived from the CD83 protein, the identity is calculated based on that portion of the CD83 protein that is present in the fragment.
Purification of CD83 pro.eins or DNA's
Purified CD83 p oteins or analogs are prepared by culturing suitable host/vector systems to express the recombinant translation products of the DNA's described herein, which are then purified from culture media or cell extracts. For example, supernatants from systems which secrete recombinant protein into culture media can be first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pell icon ltrafiltratioπ unit.
Following the ι oncentration step, the concentrate can be applied to a suitable purification matrix. For ; xample, a suitable affinity matrix can comprise a CD83 antibody molecule bound to a su. table support. Alternatively, an anion exchange resin can be employed, for example. :ι matrix or substrate having pendant diethylaminoethyl (DEAE) groups. The matrices .an be acrylamide, agarose, dextran, cellulose or other types commonly employed in protein purification. Alternatively, a cation exchange step can be employed. Suitable caiion exchangers include various insoluble matrices comprising sulfopropyl or carboxyrr ethyl groups. Sulfopropyl groups are preferred. Gel filtration chromatography also provides a means of purifying CD83 proteins. Affinity chromatography is a pamcularly preferred method of purifying CD83 proteins. For example, a CD83 protein expressed as a fusion protein comprising an immunoglobulin Fc region can be purified using Protein A or Protein G affinity chromatography Moreover, a CD83 protein comprising an oligomerizing zipper domain may be punfied on a resm comprising an antibody specific to the oligomerizing zipper domain. Monoclonal antibodies against a CD83 protein may also be useful in affinity chromatography purificaiion, by utilizing methods that are well-known in the art.
Finally, one or more reversed-phase high performance liquid chromatography (RP-
HPLC) steps employing hydrophobic RP-HPLC media, e.g., silica gel having pendant methyl or other aliphatic groups, can be employed to further purify a viral protein composition Some or all of the foregoing purification steps, in vaπous combinations, can also be employed to provide a homogeneous recombinant protein.
Recombinant CDS3 protein produced in bacteπal culture is usually isolated by initial extraction from cell pellets, followed by one or more concentration, salting-out, aqueous ion exchange oi size exclusion chromatography steps. Finally, high performance liquid chromatography (HPLC can be employed for final purification steps. Microbial cells employed in expression of recombinant CD83 protein can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents Femientation of yeast which express CD83 protein as a secreted protein greatly simplifies purification Secreted recombinant protein resulting from a large-scale fermentation can be purried by methods analogous to those disclosed by Urdal et al. (J.
Chromatog. 296: 171 , 1 J84). This reference describes two sequential, reversed-phase
HPLC steps tor purification of recombinant human GM-CSF on a preparanve HPLC column.
CD83 protein synihesized in leco binant culture is characterized by the presence of non-CD83 cell components, including proteins, in amounts and of a character which depend upon the punlic ition steps taken to recover the viral protein from the culture. These components ordinarily will be of yeast, prokaryotic or non-human higher eukaryotic oπgin and preferably are present in innocuous contaminant quantities, on the order of less than about 1 percent by . eight. Fuπher, recombinant cell culture enables the production of CD83 protein free of cttier proteins which may be normally associated with the CD83 protein as it is found in I iture in its species of origin.
Useful CD83 O A s may be punfied by any suitable method of puπfying DNA's known in the an Seve.al useful methods are described in Sambrook et al. (Molecular
Cloning: A Laboratory y anual. Cold Spring Harbor Press, Cold Spring Harbor, New
York, second edition: 1989), paπicularly in Chapter 1, in the section relating to extraction and purification of plasmid DNA ( 1.21 ) For example, DNA is amplified in prokaryotic cells, and isolated by a tandard alkaline lysis procedure followed by resin puπficauon as descπbed in standard ki (tor example, from Promega Biotec, Madison, WT, or Quiagen, Chatswoπh, CA) The isolated DNA is then resuspended in a suitable diluent or earner
Administration of CD83 Protein and DNA Compositions
The present invention provides methods of using therapeudc compos ons comprising an effective amount of a CD83 reagent and a suitable diluent and earner, and methods for regulating mi immune response The use of CD83 proteins in conjunction with soluble cytokine re, ^ptors or cytokines, or other immunoregulatory molecules is also contemplated CD83 DNA's and/or proteins are administered for the purpose of stimulating a humoral immune response
For therapeutic , se, α purified CD83 reagent is administered to an individual, preferably α human, toi ueαtment in α manner appropπate to the indication Thus, for example, CD83 protein c mpositions administered to stimulate a humoral immune response can be given by bolus n lection, continuous infusion, sustained release from implants, or other suitable technique Typically, a therapeutic agent will be administered in the form of a composition comprising purified CD83 protein in conjunction with physiologically acceptable earners, excipients or diluents Such earners will be nontoxic to recipients at the dosages and concentratio is employed Ordinal lly, the of such CD83 protein compositions entails combining the CD83 piotein w ith butters, antioxidants such as ascorbic acid, low molecular weight (less than about 10 residu es) polypeptides, proteins, amino acids, carbohydrates including glucose, sucrose or deuπns, chelating agents such as EDTA, glutathione and other stabilizers and excipients Neutral buffered saline or saline mixed with conspecific serum albumin are exemplar* ippropπate diluents Preferably, product is formulated as a lyophi zate using approi πate excipient solutions (e.g , sucrose) as diluents Appropπate dosages can be determi' e l in trials The amount and frequency of administranon will depend, of course, on su. h factors as the nature and severity of the indication being treated, the desired response, the .ondition of the individual, the nature of an antigen for which the CD83 is being used as ar adjuvant, and so foπh
Since DNA can b_ integrated into the genome, or be maintained in episomal form, DNA vaccines provide the potential for long-term expression of antigens, with commensurate dilution of an immune response Simple saline solutions appear to be suitable earners foi DNΛ and various routes of administration have been shown to be useful, including v iamuscular (Ulmer et al , Science 259 1745, 1993, Montgomery et al , DNA Cell Biol 12 777, 1993) oi inuadermal injection (Raz et al., Proc Natl Acad. Sci USA 91 951l). 1994), as well as the use of a "gene gun" (Tang et al , Nature 365: 152, 1992, Fynan el al , Proc Natl Acad Sci USA 90.1148, 1993; Eisenbraun et al, DNA Cell Biol 12 791 , 1993)
For use is stimul mng a ceπain type of immune response, administration of other cytokines along with either CD83 DNA or CD83 protein, is also contemplated. Several useful cytokines (or peptide regulatory factors) are discussed in Schrader, J.W (Mol Immunol 28 295, 1991 ) Such factors include (alone or in combination) Interleukins 1, 2, 4, 5, 6, 7, 10, 12 and 1 , granulocyte-macrophage colony sumulaung factor, granulocyte colony stimulating factor, a fusion protein comprising Interleukιn-3 and granulocyte- macrophage colony stimulating factor, Interferon-y, TNF, TGF-β, flt-3 ligand and biologically active derivatives thereof A paπicularly prefened cytokine is CD40 ligand (CD40L) A soluble r,.,τn of CD40L is descπbed in USSN 08/484,624, filed June 7, 1995 DNA encoding such cytokines will also be useful in the invennve methods Administration of these immunomodulatory molecules includes simultaneous, separate or sequential administiatioi with suitable CD83 compositions (proteins or DNA's) and antigens
Antagonists of CD83 will be useful in inhibiting a humoral immune response Exemplary conditions in which it is advantageous to inhibit such undesirable responses include autoimmune syndiomes, including myasthenia gravis, multiple sclerosis and systemic lupus erytheπ αiosis, and others as descπbed in U S Patent 5,284,935 Moreover, CD83 antαgoi ists can also be useful to prevent or treat rejection of tissue and/or organ tiansplants Othci conditions tor which CD83 antagonists can be useful include those which involve und. suable immune responses to foreign antigens, for example those which occur in allergy or asthma
The following ι * amples are offered by way of illustration, and not by way of limitation Those skilled in the tu t will recognize that variations of the invention embodied in the examples can be made, especially in light of the teachings of the vanous references cited herein, the disclosi es of which aie incorporated by reference
EXAMPLE 1
This example describes construction of an HB 15 (CD83) DNA construct to express a soluble CD83 protein Using conventional techniques of PCR amphficauon, enzyme cutting and hgation, sceral CD83-encodιng constructs were prepared, including one encoding a CD83/Fc his, on protein, a Flag®/CD83 protein, and a soluble form of CD83 An expression vector (pDC4()9, which differs from pDC406 (McMahan et al., EMBO J 10:2821, 1991 ) in that a 'igl II restriction site outside of the mulαple cloning site has been deleted, making the B II site within the multiple cloning sue unique) compπsmg appropriate ιegulator> ements, and sequences encoding the signal pepnde and extracellular domain of OD83 from amino acid -19 to amino acid 124 of SEQ ID NO:l, along with a suitable Fc region of an immunoglobulin (SEQ ID NO:3; three amino acids in the hinge region have bc:n changed to reduce affinity for Fc receptor) was prepared and expressed. The resulting fusion protein was refened to as CD83/Fc Type I. A soluble form of CD83 from amino acid -19 to amino acid 124 of SEQ ID NO:l, and a Flag® form consisting of amino acid -19 to amino acid 124 of SEQ ID NO:l linked to the eight amino acid sequence described by Hopp et al. (BiolTechnology 6:1204,1988; SEQ ID NO:5) were prepared in an expression vector (pDC304, which is derived from pDC302, described by Mosley et al., Cell, 59:335 (1989), by deleting the adenovirus tripaπite leader (TPL) in pDC302).
The resultant expression vectors are transfected into the monkey kidney cell line CV-1/EBNA (ATCC CRL 10478). Large scale cultures of CV-1/EBNA cells transfected with the various constructs are grown to accumulate supernatant containing the different forms of CD83 protein The CV-1/EBNA cell line permits expression of recombinant proteins ligated into veciors containing the EBV origin of replication. The CD83 proteins in supernatant fluid are p.irified as described below.
EXAMPLE 2
This example ill istrates purification of various forms of CD83 proteins. The Flag®/CD83 protein was uirified by affinity chromatography. Briefly, culture supernatant containing the Flag*/CDi;3 protein was purified by filtering mammalian cell supernatants (e.g., in a 0.45μ filter) an j applying filtrate to an affinity column comprising a monoclonal antibody specific for the Flag® peptide, coupled to Affi-gel active ester agarose (Bio-Rad, Richmond, CA), at room temperature, at a flow rate of approximately 60 to 80 ml/hr for a 1.5 cm x 12.0 cm column. The column was washed with approximately 20 column volumes of PBS (phosphate buffered saline), until free protein could not be detected in wash buffer. Bound CC !i3 protein was eluted from the column by competition with excess Flag® peptide ( 100 μg/i. I in PBS), and stabilized in 10% glycerol.
CD83/Fc protein 's purified by conventional methods using Protein A or Protein G chromatography. Approximately one liter of culture supernatant containing CD83 protein is purified by filtering nπmmalian cell supernatants (e.g., in a 0.45m filter) and applying filtrate to a protein A/G antibody affinity column (Schleicher and Schuell, Keene, NH) at 4*C at a flow rate of 80 I il/hr for a 1.5 cm x 12.0 cm column. The column is washed with 0.5 M NaCl in PBS until free protein is not detected in the wash buffer. Finally, the column is washed with PBS. Bound fusion protein is eluted from the column with 25 mM citrate buffer, pH 2.8, and brought to pH 7 with 500 mM Hepes buffer, pH 9.1.
Additional const; nets can be prepared and the expressed protein purified using methods that are known in the art. For example, a CD83 protein comprising a poly-His peptide may be detected and/or purified using a poly-His system, substantially as described in US Patent 5,284,933, issued February 8, 1994.
Ability to bind antibodies to CD83 is used as an assay for detection of CD83 activity. Biological activity is measured in any biological assay which quantifies an antigen-specific immun response, for example, as described in the Examples herein.
EXAMPLE 3
This example illustrates the effect of CD83/Fc on a secondary immune response. On day 0, 6 BALB/c mice were injected subcutaneously with 200μl of a suspension containing 4 μg of ovalbumin (OVA). On day 14, they were injected with either 500 μg of CD83/Fc or 500 rat IgG as a control. Six hours later they were re-immunized with 1 μg ovalbumin. The mice were bled at day 21 and titers of ovalbumin-specific antibodies of various subclasses were determined by ELISA. The mice that were given CD83/Fc prior to the secondary immunization exhibited slightly higher levels of certain subclasses of ovalbumin-specific antibodies, particularly IgG2b-
EXAMPLE
The example illustrates the ability of DNA encoding CD83 to stimulate a primary humoral immune response with high levels of IgG2t>- BALB/c mice (six per group) were injected with 50 μg of DNA comprising DNA encoding a tumor necrosis factor/immunoglobulin I 'c fusion protein (described by Mohler et al., J Immunol 151 :1548.1993: and in EP 418014), along with either 50 μg CD83-encoding vector DNA or control vector DNA. Mice were bled at day 14 for determination of TNFr/Fc-specific antibody titers by ELISΛ. Results are shown in table 1 below; titration curves for the TNFr/Fc-specific I G2h re shown in Figure 1. The results demonstrated that CD83 cDNA significantly enhanced antigen-specific antibody titers of all isotypes, especially IgG2a an IgG2b-
Table 1 : End-point titers of TNFr/Fc-specific antibody in animals injected with TNFr Fc DNA and either control DNA or CD83 DNA*
Figure imgf000018_0002
Lowest reciprocal dilution of sera at which all animals in each group have detectable liters
In a second expcnment, mice
Figure imgf000018_0001
injected with either 50 μg CD83-encodιng vector DNA (6 mice) ot . ontrol vector DNA (5 mice) at day 0, given lntradermally near the base ot the tail At day 3 they were given 5 μg of TNFr/Fc protein subcutaneously at the back of the neck The i.nce were bled at day 14 for determmauon of TNFr/Fc-specific antibody titers by ELISΛ Results are shown in table 2 below; ntrauon curves for the TNFr- pecific IgG2b ■" . shown in Figure 2. The results confirmed that CD83 cDNA significantly enhanced a itigen-specific antibody titers of all isotypes, especially IgG2a an IgG2b
T. ble 2. End-point titers of TNFr/Fc-specific antibody in animals injected with TNFr/Fc protein and either control DNA or CD83 DNA*
Lowest reciprocal dilution ot sera ai which all animals in each group have detectable liters
These results demonstrated that use of CD83 as a vaccine adjuvant results in a pnmary humoral immi. l response that qualitatively and quantitatively resembles a secondary immune iespt use EXAMPLE 5
This example illuurates the ability of CD83 proteins to induce isotype switching in naive human B cells. Naive B cells are obtained by one of several methods known in the art. For Example, surface IgD-positive tonsillar B cells, neonatal B cells obtained from cord blood and B cell obtained from individuals suffering from X-linked hyper-IgM syndrome represent populations of cells that are substantially devoid of isotype-committed B cells. Mononuclear cells are isolated by a method such as centrifugation over Ficoll- Hypaque, and depleted of T cells by rosetting with 2-aminoethylisothiouronium bromide- treated SRBC (sheep reJ blood cells). The resulting E" cells can be fuπher purified by negative selection using antibodies to cell surfaces markers found on non-B cells (i.e., CD2, CD3, CD 14), and/or by positive selection using antibodies to markers found on non- isotype committed B cells (i.e., slgD). The cells are cultured under suitable conditions, and stimulated with cytokines to induce immunoglobulin secretion. The cytokines to be used include a soluble vrimeric from of CD40 ligand (CD40L) as described in USSN 08/484,624, filed June 7. 1995: 1L-4, and IL- 10. Other cytokines may also be included, for example, transforming growth factor β and IL-2. After stimulation, supernatants are harvested and tested for the presence of various classes of immunoglobulins by ELISA. CD83 stimulates secretion of IgG2b from murine B cells under such conditions; it will likewise stimulate isotype switching and secretion of IgG2 from naive human B cells.
EXAMPLE 6 This example illustrates the preparation of monoclonal antibodies against CD83. Preparations of purified r.combinant CD83/Fc, for example, or transfected cells expressing high levels of CD83, are employed to generate monoclonal antibodies against CD83 using conventional techniques, such as those disclosed in U.S. Patent 4,41 1,993. Such antibodies are likely 10 t>. useful in interfering with CD83 binding or biological activity, as components of diagnostic or research assays for CD83, or in affinity purification of CD83.
To immunize rodents, CD83 immunogen is emulsified in an adjuvant (such as complete or incomplete Freund's adjuvant, alum, or another adjuvant, such as Ribi adjuvant R700 (Ribi, H. milton, MT), and injected in amounts ranging from 10- 100 μg subcutaneously into a se! ,-cted rodent, for example, BALB/c mice or Lewis rats. Ten days to three weeks clays late: . the immunized animals are boosted with additional immunogen and periodically boost ;d thereafter on a weekly, biweekly or every third week immunization schedule. Serum samples are periodically taken by retro-orbital bleeding or tail-tip excision for testing by dot-blot assay (antibody sandwich) or ELISA (enzyme-linked immunosorbent assay). Other assay procedures are also suitable, for example, FACS analysis using cells expressing membrane-bound CD83. Following detection of an appropriate antibody t e:, positive animals are given an intravenous injection of antigen in saline Three to four d ιys later, the animals are sacnficed, splenocytes harvested, and fused to a muπne myeloma cell line (e.g., NS 1 or preferably Ag 8.653 [ATCC CRL 1580]). Hybπdoma cell lines generated by this procedure are plated in multiple microdter plates in a selective medium (for example, one containing hypoxanthine, a inopteπn, and thymidine, or HAT) to inhibit proliferation of non-fused cells, myeloma-myeloma hybrids, and splenocyte-splenocyie hybrids.
Hybπdoma clon s thus generated can be screened by ELISA for reactivity with CD83, tor example, b adaptations ot the techniques disclosed by Engvall et al., Immunochem #-871 ( H71 ) and in U.S. Patent 4,703,004. A preferred screening technique utilizes fluores ;ence activated cell somng to detect binding to cells that express CD83, for example, Ra cells Positive clones are then generated and injected into the peπtoneal cavities of sy lgeneic rodents to produce ascites containing high concentrations (>1 mg/ml) ot anti-CDS -> monoclonal antibody. The resulting monoclonal antibody can be punfied by ammonium ultate precipitation followed by gel exclusion chromatography. Alternatively, affinity clnomatography based upon binding of antibody to protein A or protein G can also be us.d. as can affinity chromatography based upon binding to CD83 protein
Using these meth >ds, three hybπdoma clones secreting antibodies that bound CD83 were generated, the antibodies were refeπed to as M43, M240, and M245 The antibodies were able to partially compete with each other for CD83 binding, as determined by ELISA and FACS. initial result . indicated that they bound to slightly different epitopes. The antibodies were also able to inhibit antigen-specific proliferation of peπpheral blood T cells.
EXAMPLE 7 This example de -. iibes two solid-phase binding assays, the first of which, (a), can be used to quantify sol ble CD83, and the second of which, (b), is used to detect the presence ot soluble CD8 <
(a) Quantitative < Y>&? ELISA
Antibody to CDK 1 is purified and used to coat 96-well plates (for example, Corning EasyWash ELISA plate1 . Corning, NY, USA). In a preferred method, the plates arc coated for one hour at r om temperature with 50 μl of PBS containing 5 μg/ml of M43 (descπbed m Example 6) and blocked with KK) μl/well of 5 % non-fat dπed milk in PBS for 1 hour at room tempt αtuie. Samples to be tested are diluted in 10% normal goat serum (NGS) in PBS. and 50 μl is added per well A titration of unknown samples is run in duplicate, and a titration ol reference standard of CD83 is run to generate a standard curve. The plates ore incubated v\ ith the samples and controls for one hour at room temperature, then washed four times / ith PBS Second step reagent, for example, rabbit anti-CD83 (50 μl/well, diluted 1 -500 n PBS/10 % NGS), is added and the plates are incubated at room temperature tor one ho The plates are again washed as previously descnbed, and donkey anti-rabbit IgG .oiiμigated to horseradish peroxidase (Jackson Labs Westgrove, PA; diluted 1.2(KX) in PBS/10 % NGS) is added. Plates are incubated for one hour at room temperauue, washed as described, and the presence of CD83 is detected by the addition of chromogen/sub trate. tetiamethyl benzidene/peroxidase (TMB, 50 μl/well; Kirkegard and Perry, Gaithersberg, MD) at room temperature until development of color. The chromogemc reaction is ^topped by the addition of 50 μl/well 2N H2SO4, and the OD450 of the wells determined The quantity of soluble CD83 can be determined by companng the OD values obtained wuh the unknown samples to the values generated for the standard curve Values are expressed as the number of picograms per ml. (b) Qualitative Dot Blot
Soluble CD83 ( 1 μi of cπide supernatant or column fracnons) is adsorbed to dry BA85/21 nitrocellulose membranes (Schleicher and Schuell, Keene, NH) and allowed to dry. The membianes ar, incubated in tissue culture dishes for one hour in Tris (0.05 M) buffered salme (0 15 V pH 7 5 containing 1 % w/v BSA to block nonspecific binding sites At the end ol this time, the membranes are washed three times in PBS, and rabbit antι-CD83 antibod) is idded at an approximate concentration of 10 μg/ml in PBS containing \ r/c BSA. following which the membranes are incubated for one hour at room temperauue The membianes αie again washed as described, and a horseradish peroxidase (HRP)-lαbeled antibody such as goat anti-rabbit lg, Southern Biotech, Birmingham, AL) at an approximate dilution ot 1 :1000 in PBS containing 1% BSA is added. After incubating for one houi it room temperature, the membranes are washed and chromogen (i.e. 4-chloronaphthol re igent, Kukegard and Peπy, Gaithersburg, MD) is added. Color is allowed to develop loi ten minutes at room temperature, and the reaction is stopped by πnsing the membranes mth watei The membranes are washed, and the presence of soluble CDS 3 is deteniv ed by analyzing tor the presence of a blue-black color This assay is used to determine th piesence or absence of soluble CD83 in cell culture supernatant fluids and 111 purification column tractions The assay fuπher provides a semi-quantrtauve method of determining iclative amounts of soluble CD83 by companng the intensity of the color in unknown sα p s to the intensity of known quantities of controls.
EXAMPLE 8
This example denionstiαtes that soluble CD83 is shed from the surface of activated peripheral blood B and T cells. Peripheral blood B and T cells are obtained by any suitable method known in the an For example. PBMCs are isolated from a healthy donor by centrifugation of heparu zed blood over Isolymph (Gallard-Schlesinger Industnes, Inc., Norway) and washed tl ree times (i.e., with culture medium consisting of RPMI 1640 supplemented with 10 ,< FCS, 50 U/ml penicillin, 50 μg ml streptomycin, 2mM glutamine). Isolated PBMCs are separated into T cell and non-T-cell fractions by rosetting with 2-aminoethylisothiouronium bromide (AET)-treated sheep red blood cells. Twice rosetted cells are suspended in RPMI 1640 culture media with 10 % FCS and incubated on plastic dishes for 1 hr at 37°C to remove any remaining adherent cells. The resulting cell preparations were always at least 90% T cells (98% CD2+; 90% CD3+) as determined by flow cytometric analysis B cells are fuπher purified from the E" preparation by positive selection using CD 19 monoclonal antibody (mAb) on a MACS column (Miltenyi Biotec, Sunnyvale, CA) according to the manufacturers instructions. CD19+ B cells purified in this way were routinely :-95% pure as determined by reactivity with CD20 mAb.
Peripheral blood T or B cells were cultured at lxlO6 in 1ml for 48 or 24 hours with the following stimuli: immobilized CD3 mAb, 5μg/ml; PHA, 1%; IL-2, lOng/ml; PMA, lOng/ml; Ionomycin, 500ng/ml: IL-4, lOng/ml; soluble trimeric CD40L, 2μg/ml; SAC, 0.01 %. Levels of soluble CD83 in culture supernatants were measured by ELISA A protease inhibitor, TAP! (described in USSN 08/292.547, filed August 18, 1994, now allowed; lOOμM final c ncentration), was included in the culture medium to ascertain whether a proteolytic re: rtion was required for expression of soluble CD83. After culture for either 48 hours (T cells) or 24 hours (B cells), 50 μl of supernatant fluid was removed and tested for the presence of soluble CD83 by quantitative EIA as described in Example 7 above. Results (expressed in pg/ml of CD83) are shown in Table 3 below.
Figure imgf000022_0001
These results indicated that soluble CD83 was expressed as a result of a proteolytic cleavage of membrane-bound CD83. In general, B cells produced larger quantities of soluble CD83 than did T cells, and for both cell types, certain stimuli caused shedding of greater quantities of CDH than did other stimuli.
EXAMPLE ? This example illustrates the ability of CD83 proteins to enhance immunoglobulin secretion from human B cells. Isotype-switched B cells are obtained by one of several methods known in the ar.. For example, surface IgD-negative B cells are fuπher purified from peripheral mononuclear cells (isolated by a method such as centrifugation over Ficoll- Hypaque, and depleted of T cells by rosetting with 2-aminoethylisothiouronium bromide- treated SRBC) and depleting the E- cells of IgD+ B cells (i.e., by positive selection on a MACS column). The resulting cells can also be fuπher purified by positive selection for a B cell marker such as CD 19. IgD" B cells may also be obtained from tonsils, using methods that are known in the art (for example, Liu et al., Eur. J. Immmunol. 21 :1107, 1991 ; or Lagresle et al.. .'«. Immunol. 5: 1250, 1993)
IgD" peripheral ood B cells (5x1 Orwell) were cultured for twelve days in the presence of soluble, trimcrie CD40L (2μg/ml) and IL-2 (lOng/ml); IL-6 (lOng/ml) or IL-10
(20ng/ml) were also included in some cultures. Recombinant soluble CD83 was added to cultures at a final conc itrαtion of 10 or lOOng/ml. IgGl and IgG2 levels in culture supernatants were deterr ined at day twelve by ELISA. The results are shown in Table 4.
Table 4: Effect of Soluble CD83 of Immunoglobulin Secretion b Human B Cells
Figure imgf000023_0001
These results demonstrated that soluble CD83 enhanced secretion of both IgGi and
IgG2 by IgD" human peripheral blood B cells. SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: IMMUNEX CORPORATION
(ii) TITLE OF I NVENTION: METHODS AND COMPOSITIONS FOR MODULATING AN
IMMUNE RESPONSE
(iii) NUMBER OF SEQUENCES: 5
(iv) CORRESPOND NCE ADDRESS: (A) ADDRESSEE: IMMUNEX CORPORATION
(B) STREET: 51 UNIVERSITY STREET
(C) CITY: SEATTLE
(D) STATE: WASHINGTON
(E) COUNTHY: USA (F) ZIP: '.8101
(v) COMPUTER Rl ADABLE FORM:
(A) MEDIU TYPE: Floppy disk
(B) COMPUTER. Apple Macintosh (C) OPERA'IING SYSTEM: Apple Operating System 7.5.2
(D) SOFTWARE: Microsoft Word for Power Macintosh, Version 6.0.1
(vi) CURRENT AP-LICATION DATA: (A) APPLICATION NUMBER:
(B) FILINC. DATE: 13 FEBRUARY 1997
(C) CLASSIFICATION:
(vii) PRIOR APPLICATION DATA: (A) APPLI' ATION NUMBER: USSN 08/720,284
(B) FILINC DATE: 26 SEPTEMBER 1996
(C) CLASSIFICATION:
(vii) PRIOR APPLICATION DATA: (A) APPLICATION NUMBER: USSN 08/673,753
<B) FILINC DATE: 27 JUNE 1996 (C) CLASSIFICATION:
(vii) PRIOR APPLICATION DATA: (A) APPLICATION NUMBER: USSN 08/601,954
(B) FILIN' DATE: 15 FEBRUARY 1996
(C) CLASSI ICATION:
(viii) ATTORNEY/P ENT INFORMATION: (A) NAME: Perkins, Patricia Anne
(B) REGISTRATION NUMBER: 34,693
(C) REFER.-NCE/DOCKET NUMBER: 2622-WO
(ix) TELECOMMUNICATION INFORMATION: (A) TELEPHONE: (206)587-0430
(B) TELEFAX: (206)233-0644
(2) INFORMATION FOR SEQ ID NO: 1 :
(i) SEQUENCE r ΛPACTERISTICS:
7"> (A) LENGTH: 618 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(iii) HYPOTHETICAL: NO (IV) ANTI-SENSE NO
(vu) IMMEDIATE JDURCE: (B) CLONE- HB15
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..618 (ix) FEATURE:
(A) NAME/KEY: mat_peptιde
(B) LOCAT . :ri: 58..615
(xi) SEQUENCE D.' SCRIPTION: SEQ ID NO: 1 :
ATG TCG CGC GGC CTC CAG CTT CTG CTC CTG AGC TGC GCC TAC AGC CTG 48 Met Ser Arg Gly Leu Gin Leu Leu Leu Leu Ser Cys Ala Tyr Ser Leu -19 -15 -10 -5
GCT CCC GCG ACG CCG SAG GTG AAG GTG GCT TGC TCC GAA GAT GTG GAC 96 Ala Pro Ala Thr Pro Glu Val Lys Val Ala Cys Ser Glu Asp Val Asp 1 5. 10 TTG CCC TGC ACC GCC CCC TGG GAT CCG CAG GTT CCC TAC ACG GTC TCC 144 Leu Pro Cys Thr Ala ?rc Trp Asp Pro Gin Val Pro Tyr Thr Val Ser 15 20 25
TGG GTC AAG TTA TTG GAG GGT GGT GAA GAG AGG ATG GAG ACA CCC CAG 192 Trp Val Lys Leu Leu Glu Gly Gly Glu Glu Arg Met Glu Thr Pro Gin 30 35 40 45
GAA GAC CAC CTC AGG 3GA CAG CAC TAT CAT CAG AAG GGG CAA AAT GGT 240 Glu Asp His Leu Arg Gly Gin His Tyr His Gin Lys Gly Gin Asn Gly 50 " 55 60
TCT TTC GAC GCC CCC AAT GAA AGG CCC TAT TCC CTG AAG ATC CGA AAC 288 Ser Phe Asp Ala Pro Asn Glu Arg Pro Tyr Ser Leu Lys lie Arg Asn 65 70 75
ACT ACC AGC TGC AAC ICG GGG ACA TAC AGG TGC ACT CTG CAG GAC CCG 336 Thr Thr Ser Cys Asn Ser Gly Thr Tyr Arg Cys Thr Leu Gin Asp Pro 80 85 90 GAT GGG CAG AGA AAC CTA AGT GGC AAG GTG ATC TTG AGA GTG ACA GGA 384 Asp Gly Gin Arg Asn Leu Ser Gly Lys Val lie Leu Arg Val Thr Gly 95 100 105 TGC CCT GCA CAG CGT \AA GAA GAG ACT TTT AAG AAA TAC AGA GCG GAG 432 Cys Pro Ala Gin Arg Lys Glu Glu Thr Phe Lys Lys Tyr Arg Ala Glu 110 115 120 125 ATT GTC CTG CTG CTG GCT CTG GTT ATT TTC TAC TTA ACA CTC ATC ATT 480 lie Val Leu Leu Leu 41a Leu Val lie Phe Tyr Leu Thr Leu lie lie 130 135 140
TTC ACT TGT AAG TTT GCA CGG CTA CAG AGT ATC TTC CCA GAT TTT TCT 528 Phe Thr Cys Lys Phe Ala Arg Leu Gin Ser lie Phe Pro Asp Phe Ser 145 150 155
AAA GCT GGC ATG GAA CGA GCT TTT CTC CCA GTT ACC TCC CCA AAT AAG 576 Lys Ala Gly Met Glu \rg Ala Phe Leu Pro Val Thr Ser Pro Asn Lys 160 165 170
CAT TTA GGG CTA GTG ACT CCT CAC AAG ACA GAA CTG GTA TGA 618
His Leu Gly Leu Val Thr Pro His Lys Thr Glu Leu Val * 175 180 185
(2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS: (A) LE 3TH: 206 amino acids
(B) TYPE: amino acid (D) TOl'DLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2 :
Met Ser Arg Gly Leu Gin Leu Leu Leu Leu Ser Cys Ala Tyr Ser Leu -19 -15 -10 -5
Ala Pro Ala Thr Pre Glu Val Lys Val Ala Cys Ser Glu Asp Val Asp 1 "5 10
Leu Pro Cys Thr Aid rro Trp Asp Pro Gin Val Pro Tyr Thr Val Ser 15 20 25
Trp Val Lys Leu Leu Glu Gly Gly Glu Glu Arg Met Glu Thr Pro Gin
30 35 " 40 45 Glu Asp His Leu Arg Gly Gin His Tyr His Gin Lys Gly Gin Asn Gly
50 55 60
Ser Phe Asp Ala Pro Asn Glu Arg Pro Tyr Ser Leu Lys lie Arg Asn 65 70 75
Thr Thr Ser Cys Asn Ser Gly Thr Tyr Arg Cys Thr Leu Gin Asp Pro 80 85 90
Asp Gly Gin Arg Asn Leu Ser Gly Lys Val lie Leu Arg Val Thr Gly 95 100 105
Cys Pro Ala Gin Arg Lys Glu Glu Thr Phe Lys Lys Tyr Arg Ala Glu
110 115 120 125 lie Val Leu Leu Leu Ala Leu Val lie Phe Tyr Leu Thr Leu lie lie
130 135 140 Phe Thr Cys Lys Phe Ala Arg Leu Gin Ser lie Phe Pro Asp Phe Ser 145 150 155
Lys Ala Gly Met Glu Arg Ala Phe Leu Pro Val Thr Ser Pro Asn Lys 160 165 170
His Leu Gly Leu Val Thr Pro His Lys Thr Glu Leu Val 175 180 185
(2) INFORMATION FOR SEQ ID NO: 3 :
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 699 base pairs
(B) TYPE: nucleic acid
(C) STRANLEDNESS : single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: NO
(vii) IMMEDIATE SOURCE:
(B) CLONE: HulgG Fc mutein (ix) FEATURE:
(A) NAME/ EY: CDS
(B) LOCATION: 1..699
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3 :
GAG CCC AGA TCT TGT GAC AAA ACT CAC ACA TGC CCA CCG TGC CCA GCA 48
Glu Pro Arg Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala
1 5 10 15
CCT GAA GCC GAG GGC GCG CCG TCA GTC TTC CTC TTC CCC CCA AAA CCC 96 Pro Glu Ala Glu Gly Ala Pro Ser Val Phe Leu Phe Pro Pro Lys Pro 20 " 25 30 AAG GAC ACC CTC ATG ATC TCC CGG ACC CCT GAG GTC ACA TGC GTG GTG 144 Lys Asp Thr Leu Met lie Ser Arg Thr Pro Glu Val Thr Cys Val Val 35 40 45
GTG GAC GTG AGC CAC GAA GAC CCT GAG GTC AAG TTC AAC TGG TAC GTG 192 Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val 50 55 60
GAC GGC GTG GAG GTG CAT AAT GCC AAG ACA AAG CCG CGG GAG GAG CAG 240 Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gin 65 70 75 80
TAC AAC AGC ACG TAC CGG GTG GTC AGC GTC CTC ACC GTC CTG CAC CAG 288 Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gin 85 90 95 GAC TGG CTG AAT GGC AAG GAC TAC AAG TGC AAG GTC TCC AAC AAA GCC 336
Asp Trp Leu Asn Gly Lys Asp Tyr Lys Cys Lys Val Ser Asn Lys Ala 100 105 110 CTC CCA GCC CCC ATG CAG AAA ACC ATC TCC AAA GCC AAA GGG CAG CCC 384
Leu Pro Ala Pro Met Gin Lys Thr lie Ser Lys Ala Lys Gly Gin Pro 115 120 125
CGA GAA CCA CAG GTG TAC ACC CTG CCC CCA TCC CGG GAT GAG CTG ACC 432 Arg Glu Pro Gin Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr 130 135 140
AAG AAC CAG GTC AGC CTG ACC TGC CTG GTC AAA GGC TTC TAT CCC AGG 480
Lys Asn Gin Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Arg 145 150 155 160
CAC ATC GCC GTG GAG TGG GAG AGC AAT GGG CAG CCG GAG AAC AAC TAC 528
His lie Ala Val Glu Trp Glu Ser Asn Gly Gin Pro Glu Asn Asn Tyr 165 170 175
AAG ACC ACG CCT CCC GTG CTG GAC TCC GAC GGC TCC TTC TTC CTC TAC 576
Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr 180 185 190 AGC AAG CTC ACC GTG GAC AAG AGC AGG TGG CAG CAG GGG AAC GTC TTC 624
Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gin Gin Gly Asn Val Phe 195 200 205
TCA TGC TCC GTG ATG CAT GAG GCT CTG CAC AAC CAC TAC ACG CAG AAG 672 Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gin Lys 210 215 220
AGC CTC TCC CTG TCT CCG GGT AAA TGA 699
Ser Leu Ser Leu Ser Pro Gly Lys * 225 230
(2) INFORMATION FOR SEQ ID NO: : (i) SEQUENCE CHARACTERISTICS:
(A) LEI-3TH: 232 amino acids
(B) TY"E: amino acid (D) T - LOGY: linear (ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: :
Glu Pro Arg Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala 1 5 10 15
Pro Glu Ala Glu Gl_ Ala Pro Ser Val Phe Leu Phe Pro Pro Lys Pro 20 25 30 Lys Asp Thr Leu Met He Ser Arg Thr Pro Glu Val Thr Cys Val Val 35 40 45
Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val 50 55 60 Asp Gly Val Glu Val s Asn Ala Lys Thr Lys Pro Arg Glu Glu Gin 65 70 75 80
Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gin 85 90 95
Asp Trp Leu Asn Gly Lys Asp Tyr Lys Cys Lys Val Ser Asn Lys Ala 100 105 110 Leu Pro Ala Pro Met Gin Lys Thr He Ser Lys Ala Lys Gly Gin Pro 115 120 125
Arg Glu Pro Gin Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr 130 135 140
Lys Asn Gin Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Arg 145 150 155 160
His He Ala Val Glu Tro Glu Ser Asn Gly Gin Pro Glu Asn Asn Tyr 165 170 175
Lys Thr Thr Pro Pro /al Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr 180 185 190 Ser Lys Leu Thr Val ^sp Lys Ser Arg Trp Gin Gin Gly Asn Val Phe 195 200 205
Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gin Lys 210 215 220
Ser Leu Ser Leu Ser Pro Gly Lys 225 230
(2) INFORMATION FOR 3EQ ID NO: 5 :
(i) SEQUENCE Ci ARACTERISTICS :
(A) LENGT) : 5 a ino acids
(B) TYPE: a ino acid (D) TOPOLi GY : linear
(u) MOLECULE T ?L: peptide
(vii) IMMEDIATE OURCE : (B) CLONE FLAG® peptide
(xi) SEQUENCE D1 SCRIPTION: SEQ ID NO:5: Asp Tyr Lys Asp Asp Asp Asp Lys
1 5

Claims

CLAIMS We claim:
1. A method of stimulating a humoral immune response, comprising administering a CD83 DNA and an antigen, in a pharmaceutically acceptable carrier.
2. The method according to claim 1 , wherein the DNA is selected from the group consisting of:
(a) a DNA having a nucleotide sequence encoding an amino acid sequence of amino acids 1 through 124 of SEQ ID NO: 2; and
(b) DNA molecules capable of hybridization to the DNA of (a) under stringent conditions and which encode biologically active CD83.
3. The method according to claim 1 wherein the DNA encodes the extracellular domain of CD83, which comprises amino acids 1 through 124 of SEQ ID NO: 1.
4. The method according to claim 3, wherein the DNA is administered via intradermal injection.
5. A method of stimulating a humoral immune response, comprising administering a CD83 protein and an antigen, in a pharmaceutically acceptable carrier.
6. The method according to claim 5, wherein the CD83 protein is selected from the group consisting of:
(a) a CD83 peptide having an amino acid sequence of amino acids 1 through
124 of SEQ ID NO: 2;
(b) fragment of a peptide according to (a) that have CD83 biological activity; and
(c) peptides encoded by DNA molecules capable of hybridization to a DNA encoding the peptide of (a) under stringent conditions, which are biologically active.
7. The method according to claim 6, wherein the CD83 protein comprises the extracellular domain of CD83.
8. The method according to claim 6, which further comprises administering a cytokine selected from he group consisting of Interleukins 1, 2, 4, 5, 6, 7, 10, 12 and 15; granulocyte-macrophage colony stimulating factor, granulocyte colony stimulating factor, a fusion protein comprising Interleukin-3 and granulocyte-macrophage colony stimulating factor; Interferon-γ; TNF; TGF-β; flt-3 ligand: soluble CD40 ligand; biologically active derivatives of these cytokines; and combinations thereof.
9. A vaccine composition comprising a CD83 reagent selected from the group consisting of a DNA encoding CD83 and a CD83 protein, and an antigen, in a suitable diluent or carrier.
10. The vaccine composition according to claim 9, further comprising a cytokine selected from the group consisting of Interleukins 1, 2, 4, 5, 6, 7, 10, 12 and 15; granulocyte-macrophage colony stimulating factor, granulocyte colony stimulating factor, a fusion protein comprising lnterleukin-3 and granulocyte-macrophage colony stimulating factor; Interferon-γ; TNF; TGF-ß; flt-3 ligand; soluble CD40 ligand; biologically active derivatives of these cytokines; and combinations thereof.
1 1. The vaccine composition according to claim 9, wherein the DNA encoding
CD83 is selected from the group consisting of:
(a) a DNA having a nucleotide sequence encoding an amino acid sequence of amino acids 1 through 1 24 of SEQ ID NO: 2: and
(b) DNA molecules capable of hybridization to the DNA of (a) under stringent conditions and which encode biologically active CD83.
1 2. The vaccine composition according to claim 10, wherein the DNA encoding CD83 is selected from the group consisting of:
(a) a DNA having a nucleotide sequence encoding an amino acid sequence of amino acids 1 through 124 of SEQ ID NO: 2; and
(b) DNA molecules capable of hybridization to the DNA of (a) under stringent conditions and which encode biologically active CD83.
1 3. The vaccine composition according to claim 9, wherein the CD83 protein is selected from the group consisting of:
(a) a CD83 peptide having an amino acid sequence of amino acids 1 through 124 of SEQ ID NO: 2;
(b) fragment of a peptide according to (a) that have CD83 biological activity; and
(c) peptides encoded by DNA molecules capable of hybridization to a DNA encoding the peptide of (a) under stringent conditions, and which encode biologically active CD83.
14. The vaccine composition according to claim 10, wherein the CD83 protein is selected from the group consisting of:
(a) a CD83 peptide having an amino acid sequence of amino acids 1 through 124 of SEQ ID NO: 2; (b) fragments of a peptide according to (a) that have CD83 biological activity; and
(c) peptides encoded by DNA molecules capable of hybridization to a DNA encoding the peptide of (a) under stringent conditions, and which encode biologically active CD83.
PCT/US1997/002350 1996-02-15 1997-02-13 Methods and compositions for modulating an immune response WO1997029781A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU22733/97A AU2273397A (en) 1996-02-15 1997-02-13 Methods and compositions for modulating an immune response

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US60195496A 1996-02-15 1996-02-15
US08/601,954 1996-02-15
US67375396A 1996-06-27 1996-06-27
US08/673,753 1996-06-27
US72028496A 1996-09-26 1996-09-26
US08/720,284 1996-09-26

Publications (1)

Publication Number Publication Date
WO1997029781A1 true WO1997029781A1 (en) 1997-08-21

Family

ID=27416839

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/002350 WO1997029781A1 (en) 1996-02-15 1997-02-13 Methods and compositions for modulating an immune response

Country Status (2)

Country Link
AU (1) AU2273397A (en)
WO (1) WO1997029781A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000030680A1 (en) * 1998-11-23 2000-06-02 Idec Pharmaceuticals Corporation Tumor antigen-specific antibody-gp39 chimeric protein constructs
GB2370273A (en) * 2000-12-20 2002-06-26 Viaxxel Biotech Gmbh Compounds that affect CD83 expression
EP1389480A1 (en) * 2002-08-14 2004-02-18 Mondobiotech Interferon SA Therapeutical use of guanylhydrazones for the inhibition of CD83 dependent processes and dendritic cell maturation
EP1422241A1 (en) * 2002-11-19 2004-05-26 Alexander Steinkasserer Use of soluble forms of CD83 and nucleic acids encoding them for the treatment or prevention of diseases
EP1480598A2 (en) * 2001-11-21 2004-12-01 Celltech R &amp; D, Inc. Manipulation of cytokine levels using cd83 gene products
EP1572976A2 (en) * 2002-11-21 2005-09-14 Celltech R &amp; D, Inc. Modulating immune responses
WO2008024242A3 (en) * 2006-08-18 2008-10-23 Argos Therapeutics Inc Use of cd83 in combination therapies
US7700740B2 (en) 2001-11-21 2010-04-20 Celltech R&D Ltd Antibodies to CD83
US8124099B2 (en) 2002-12-04 2012-02-28 Argos Therapeutics, Inc. Soluble CD83 proteins and use thereof for the treatment and prevention of a disease or medical condition caused by dysfunction or undesired function of a cellular immune response involving T cells
US9102726B2 (en) 2002-12-04 2015-08-11 Argos Therapeutics, Inc. Nucleic acid of recombination expression vector encoding soluble forms of CD83, host cells transformed/transfected therewith and pharmaceutical compositions containing same
CN109320610A (en) * 2015-01-27 2019-02-12 中国科学技术大学先进技术研究院 Anti-human CD83 monoclonal antibody and its preparation, identification and application

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993021318A1 (en) * 1992-04-17 1993-10-28 Dana-Farber Cancer Institute, Inc. Lymphocyte activation antigen hb15, a member of the immunoglobulin superfamily
WO1995029236A2 (en) * 1994-04-25 1995-11-02 Dana-Farber Cancer Institute, Inc. Lymphocyte activation antigens and antibodies thereto

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993021318A1 (en) * 1992-04-17 1993-10-28 Dana-Farber Cancer Institute, Inc. Lymphocyte activation antigen hb15, a member of the immunoglobulin superfamily
WO1995029236A2 (en) * 1994-04-25 1995-11-02 Dana-Farber Cancer Institute, Inc. Lymphocyte activation antigens and antibodies thereto

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BLOOD, 01 November 1995, Vol. 86, No. 9, ZHOU et al., "A Distinct Pattern of Cytokine Gene Expression by Human CD83+ Blood Dendritic Cells", pages 3295-3301. *
LEUK. LYMPHOMA, 1994, Vol. 13, Suppl. 1, ENGEL et al., "New CD from the B Cell Section of the Fifth International Workshop on Human Leukocyte Differentiation Antigens", pages 61-64. *
THE JOURNAL OF IMMUNOLOGY, 15 July 1992, Vol. 149, No. 2, ZHOU et al., "A Novel Cell-Surface Molecule Expressed by Human Interdigitating Reticulum Cells, Langerhans Cells and Activated Lymphocytes is a New Member of the Ig Superfamily", pages 735-742. *

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000030680A1 (en) * 1998-11-23 2000-06-02 Idec Pharmaceuticals Corporation Tumor antigen-specific antibody-gp39 chimeric protein constructs
GB2370273A (en) * 2000-12-20 2002-06-26 Viaxxel Biotech Gmbh Compounds that affect CD83 expression
WO2002049625A2 (en) * 2000-12-20 2002-06-27 Viaxxel Biotech Gmbh Compounds that affect cd83 expression, pharmaceutical compositions comprising said compounds and methods for identifying said compounds
WO2002049625A3 (en) * 2000-12-20 2003-11-27 Viaxxel Biotech Gmbh Compounds that affect cd83 expression, pharmaceutical compositions comprising said compounds and methods for identifying said compounds
US7850968B2 (en) 2001-11-21 2010-12-14 Celltech R&D Limited Manipulation of cytokine levels using CD83 gene products
US7618629B2 (en) 2001-11-21 2009-11-17 Celltech R&D, Inc. Manipulation of cytokine levels using CD83 gene products
US7872103B2 (en) 2001-11-21 2011-01-18 Celltech R & D, Limited Modulating immune responses
EP1480598A2 (en) * 2001-11-21 2004-12-01 Celltech R &amp; D, Inc. Manipulation of cytokine levels using cd83 gene products
US7700740B2 (en) 2001-11-21 2010-04-20 Celltech R&D Ltd Antibodies to CD83
EP1480598A4 (en) * 2001-11-21 2006-03-22 Celltech R & D Inc Manipulation of cytokine levels using cd83 gene products
EP1389480A1 (en) * 2002-08-14 2004-02-18 Mondobiotech Interferon SA Therapeutical use of guanylhydrazones for the inhibition of CD83 dependent processes and dendritic cell maturation
JP2014195455A (en) * 2002-11-19 2014-10-16 アルゴス セラピューティクス,インコーポレイティド Use of soluble forms of cd83 protein and nucleic acids encoding them for treatment or prevention of diseases
EP3031824A1 (en) 2002-11-19 2016-06-15 Argos Therapeutics, Inc. Nucleic acids encoding soluble forms of cd83
US9732140B2 (en) 2002-11-19 2017-08-15 Argos Therapeutics, Inc. Use of soluble forms of CD83 and nucleic acids encoding them for the treatment or prevention of diseases
JP2006523432A (en) * 2002-11-19 2006-10-19 アルゴス セラピューティクス,インコーポレイティド Use of soluble CD83 protein and nucleic acid encoding the same for treatment or prevention of diseases
US8759505B2 (en) 2002-11-19 2014-06-24 Argos Therapeutics, Inc. Use of soluble forms of CD83 and nucleic acids encoding them for the treatment or prevention of diseases
WO2004046182A3 (en) * 2002-11-19 2004-10-14 Alexander Steinkasserer Use of soluble forms of cd83 and nucleic acids encoding them for the treatment or prevention of diseases
WO2004046182A2 (en) 2002-11-19 2004-06-03 Argos Therapeutics, Inc. Use of soluble forms of cd83 and nucleic acids encoding them for the treatment or prevention of diseases
US7893200B2 (en) 2002-11-19 2011-02-22 Argos Therapeutics, Inc. Use of soluble forms of CD83 and nucleic acids encoding them for the treatment or prevention of diseases
EP2345670A1 (en) 2002-11-19 2011-07-20 Argos Therapeutics, Inc. Use of soluble forms of CD83 and nucleic acids encoding them for the treatment or prevention of diseases
JP4740595B2 (en) * 2002-11-19 2011-08-03 アルゴス セラピューティクス,インコーポレイティド Use of soluble CD83 protein and nucleic acid encoding the same for treatment or prevention of diseases
EP1422241A1 (en) * 2002-11-19 2004-05-26 Alexander Steinkasserer Use of soluble forms of CD83 and nucleic acids encoding them for the treatment or prevention of diseases
EP2692733A1 (en) 2002-11-19 2014-02-05 Argos Therapeutics, Inc. Nucleic acids encoding soluble forms of CD83
EP1572976A4 (en) * 2002-11-21 2007-05-09 Celltech R & D Inc Modulating immune responses
EP1572976A2 (en) * 2002-11-21 2005-09-14 Celltech R &amp; D, Inc. Modulating immune responses
US9102726B2 (en) 2002-12-04 2015-08-11 Argos Therapeutics, Inc. Nucleic acid of recombination expression vector encoding soluble forms of CD83, host cells transformed/transfected therewith and pharmaceutical compositions containing same
US8124099B2 (en) 2002-12-04 2012-02-28 Argos Therapeutics, Inc. Soluble CD83 proteins and use thereof for the treatment and prevention of a disease or medical condition caused by dysfunction or undesired function of a cellular immune response involving T cells
EP2789625A1 (en) 2006-08-18 2014-10-15 Argos Therapeutics, Inc. Use of soluble CD83 for perfusing a tissue for transplantation
AU2007288368B2 (en) * 2006-08-18 2013-07-04 Coimmune, Inc. Use of CD83 in combination therapies
WO2008024242A3 (en) * 2006-08-18 2008-10-23 Argos Therapeutics Inc Use of cd83 in combination therapies
CN109320610A (en) * 2015-01-27 2019-02-12 中国科学技术大学先进技术研究院 Anti-human CD83 monoclonal antibody and its preparation, identification and application
CN109369807A (en) * 2015-01-27 2019-02-22 中国科学技术大学先进技术研究院 Anti-human CD83 monoclonal antibody and its preparation, identification and application
CN109369807B (en) * 2015-01-27 2021-07-27 中国科学技术大学先进技术研究院 Anti-human CD83 monoclonal antibody and preparation, identification and application thereof
CN109320610B (en) * 2015-01-27 2021-07-27 中国科学技术大学先进技术研究院 Anti-human CD83 monoclonal antibody and preparation, identification and application thereof

Also Published As

Publication number Publication date
AU2273397A (en) 1997-09-02

Similar Documents

Publication Publication Date Title
US6410711B1 (en) DNA encoding CD40 ligand, a cytokine that binds CD40
AU696775B2 (en) IL-17 receptor
US7309489B1 (en) Mouse CD40 ligand-specific antibodies and hybridomas
AU678787B2 (en) Methods of preparing soluble, oligomeric proteins
KR100689739B1 (en) Fc fusion proteins for enhancing the immunogenicity of protein and peptide antigens
US7875709B2 (en) Nucleic acids encoding interleukin-18 mutants
EP0897983B1 (en) Antibody against CD40-L
WO1994010308A9 (en) Methods of preparing soluble, oligomeric proteins
HU215949B (en) Process for preparation of purified nucleic-acid sequence corresponding to human gp39, of corresponding soluble gp39, and pharmaceutical composition containing thereof, and promoting of b-cell proliferation
JP2001503014A (en) Methods for enhancing a protective immune response
WO1997017368A1 (en) Cd100 antigen and uses therefor
WO1997029781A1 (en) Methods and compositions for modulating an immune response
US7405270B2 (en) CD40-Ligand lacking native-pattern glycosylation
AU2002324249A1 (en) Interleukin-18 mutants, their production and use
US20030059439A1 (en) Th1 inducing natural adjuvant for heterologous antigens
US5716623A (en) Isolated Herpesvirus saimiri proteins that bind MHC Class II molecules
US7432237B2 (en) Methods of treating asthma by administering interleukin-17 receptor
US20040005321A1 (en) Use of anti-ceacam antibodies for stimulating b cells in the production of monoclonal antibodies or in immunotheraphy
ES2352745T3 (en) FELINO CD86.
EP1181320A2 (en) Somatic transgene immunization and related methods
KR100301796B1 (en) Immunosuppressive Anti-CD40 Ligand Monoclonal Antibodies
IE911109A1 (en) Interleukin-4 binding protein-gamma
Seder et al. of June 13, 2013.

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA NZ

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
122 Ep: pct application non-entry in european phase