WO2004076489A1 - Modified antibody - Google Patents

Modified antibody Download PDF

Info

Publication number
WO2004076489A1
WO2004076489A1 PCT/NO2004/000051 NO2004000051W WO2004076489A1 WO 2004076489 A1 WO2004076489 A1 WO 2004076489A1 NO 2004000051 W NO2004000051 W NO 2004000051W WO 2004076489 A1 WO2004076489 A1 WO 2004076489A1
Authority
WO
WIPO (PCT)
Prior art keywords
recombinant molecule
molecule
derived
recombinant
vaccibodies
Prior art date
Application number
PCT/NO2004/000051
Other languages
French (fr)
Other versions
WO2004076489A8 (en
Inventor
Bjarne Bogen
Agnete Brunsvik Fredriksen
Inger Sandlie
Original Assignee
Medinnova As
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to ES04714520T priority Critical patent/ES2531204T3/en
Application filed by Medinnova As filed Critical Medinnova As
Priority to EP14195614.4A priority patent/EP2862878B1/en
Priority to EP04714520.6A priority patent/EP1599504B1/en
Priority to AU2004215489A priority patent/AU2004215489B2/en
Priority to CA2517054A priority patent/CA2517054C/en
Priority to DK04714520.6T priority patent/DK1599504T3/en
Publication of WO2004076489A1 publication Critical patent/WO2004076489A1/en
Publication of WO2004076489A8 publication Critical patent/WO2004076489A8/en
Priority to US13/353,548 priority patent/US9169322B2/en
Priority to US14/872,290 priority patent/US9796787B2/en
Priority to US15/730,776 priority patent/US20180030155A1/en
Priority to US17/689,076 priority patent/US20230048561A1/en
Priority to US18/163,484 priority patent/US20230303718A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/44Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/34Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Corynebacterium (G)
    • 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
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • 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
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7158Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for chemokines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2833Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against MHC-molecules, e.g. HLA-molecules
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • C12N9/1276RNA-directed DNA polymerase (2.7.7.49), i.e. reverse transcriptase or telomerase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/64Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising a combination of variable region and constant region components
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/40Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/07Nucleotidyltransferases (2.7.7)
    • C12Y207/07049RNA-directed DNA polymerase (2.7.7.49), i.e. telomerase or reverse-transcriptase

Definitions

  • the present invention relates to a recombinant human antibody-based molecule called s Vaccibodies, which are able to trigger both a T cell- and B cell immune response. More particularly, Naccibodies by themselves induce such strong in mune responses that adjuvants are not necessarily required.
  • the present invention also relates to a method of treating e.g. multiple myeloma by means of the said Naccibodies.
  • MM Multiple myeloma
  • Ig monoclonal immunoglobulin
  • Myeloma cells produce monoclonal Ig that is unique for each tumor and thus for each individual patient.
  • Ig is composed of two identical heavy (H) and two identical light 0 (L) chains.
  • L and H chains have highly diversified variable (V) regions, V and V H - V L and V H together form the Fv (fragment variable) that contains unique antigenic determinants called idiotopes (Id).
  • Idiotopes collectively constitute the idiotype of the Fv (of the Ig in casu). Induction of an immune response against the idiotype, so called Id- vaccination is a promising strategy in treatment of B cell lymphomas and MM (Bendandi, Gocke et al.
  • Id is a weak self-Ag in its original context (as part of Ig). Therefore, for vaccine purposes, it is important to enhance the immunogenicity of Id.
  • T helper cells (CD4 + T cells) recognize their antigen (Ag) after it has been processed through engulfment of foreign proteins by APC, proteolytic breakdown into peptide fragments that are loaded onto MHC class II molecules and transported to the surface of the APC where the peptide-MHC complex is presented to T cell receptors (TCRs) of CD4 + T cells.
  • TCRs T cell receptors
  • Activated CD4 + T cells stimulate cytotoxic T cells (CD8 + T cells) and B cells with the corresponding Ag specificity, initiating a broad response against the original Ag.
  • a major problem concerning Id-vaccination of MM patients is that Id-specific CD4 T cells in these patients, as extrapolated from experiments in mice [18], probably are tolerant to Id V-region determinants on the highly abundant myeloma protein.
  • MM patients who have undergone autologous stem cell transplantation (ASCT) may be in an advantageous phase for Id- vaccination for the following two reasons: 1) relief from T cell tolerance to myeloma protein Id and 2) development of new T cells that can respond to Id- accination.
  • Troybodies (Lunde, Munthe et al. 1999), which are equipped with a T cell epitope incorporated in a loop in a constant Ig domain results in increased T cell stimulation by a factor of 100- 100000 (Lunde, Rasmussen et al. 2001).
  • Troybodies do not include the Ag in its native conformation, such as Fv, and are therefore restricted to induction of T cell responses. Therefore, to induce an anti-Id B cell response and anti-Id Abs, it is necessary to include the complete Fv of the M component of the patient.
  • Ig immunoglobulins fused with granulocyte- macrophage colony-stimulating factor (GM-CSF) (Tao and Levy 1993), or CD40 ligand
  • scFv-GM-CSF was effective only when injected as protein and not as a DNA vaccine (Hakim, Levy et al. 1996).
  • DNA vaccination employing scFv fused to IL-l ⁇ did induce tumor immunity (Hakim, Levy et al. 1996).
  • scFv has been genetically fused with fragment C from tetanus toxin and delivered as a DNA vaccine by intramuscular (i. .) injection.
  • Vaccibodies a divalent molecule comprising a flexible hinge, with no FcR binding and that contain the Ag in its native conformation, with the purpose of inducing both strong Id-specific Ab and T cell responses.
  • Vaccibodies are large and complex macromolecules, but, surprisingly, cells were able to produce and export intact molecules.
  • the present invention relates to a novel type of human recombinant antibody-like molecules useful in the treatment of i.e. multiple myeloma. These molecules, called Vaccibodies, bind APC and are able to trigger both T cell and B cell immune response. Moreover, Vaccibodies bind divalently to APC to promote a more efficient initiation of an immune response. Hence, a major purpose of the present invention is to induce a strong immune response to render adjuvants redundant.
  • Vaccibodies comprise a dimer of a monomeric unit that consists of a targeting unit with specificity for a surface molecule on APC, connected through a hinge region and a C ⁇ 3 domain to a antigenic unit in the COOH-terminal end; the latter being of e.g. B cell lymphoma or myeloma origin (Fig 1), although any origin is possible due to the cassette cloning system in the expression vector.
  • the said molecule is capable of inducing an immune response against multiple myeloma, but extension to a general vaccination strategy for any polypeptid should be feasible.
  • the present invention also relates to a DNA sequence coding for this recombinant antibody based molecule, to expression vectors comprising these DNA sequences, cell lines comprising said expression vectors, to treatment of mammals preferentially by immunization by means of Vaccibody DNA, Vaccibody RNA, or Vaccibody protein, and finally to pharmaceuticals and a kit comprising the said molecules.
  • Fig 1 The structure of the Vaccibody.
  • the two scFvs in white target the Vaccibody to surface molecules on APC. They may be replaced by other targeting molecules, e.g. chemokine receptors.
  • the hinge provides flexibility of the relative orientation of the two NH 2 -terminal scFvs and disulfide bridges the monomers.
  • the C H 3 domains (light grey) act as a spacer between the NH -and COOH terminal scFvs and participate in the dimerization through hydrophobic interactions. These dimerization motifs may be replaced by other dimerization or multimerization domains.
  • the two scFvs shown in dark grey are the antigenic moiety of the Vaccibody. These scFvs are derived from the M component, thus harboring idiotypic sequences (black).
  • the antigenic scFv may be replaced by any polypeptide derived from an antigenic source, conferring vaccine strategies towards any antigen.
  • the Vaccibody is targeted to surface molecules on APC, the complex is taken up by receptor-mediated endocytosis, processed and Id-peptides are presented to CD4 + T cells on MHC class II. Simultaneously, the Id may stimulate B cells with an anti-Id BCR. These B cells will also serve as APC for the CD4 + T cells. Thus, T and B cells will cooperate to enhance the response, as indicated. Anti-Id B cells will as a consequence develop into plasma cells that produce anti-Id antibodies.
  • the MHC class Il-specific Vaccibodies bind CD19 + splenocytes (white).
  • the NTP-specific Vaccibodies (negative controls - grey), do not bind to the splenocytes. Binding was detected with streptavidinPE and 9A8bio which is a rat mAb that binds the antigenic Fv of M315 origin (V ⁇ l/2).
  • Fig 4 The NIP-specific control Naccibodies exhibits binding to the hapten NIP. Supernatant from cells transfected with genes encoding NIP-specific Naccibodies was added to ELISA plates coated with ⁇ IP-BSA. 9A8 bio (antiN ⁇ l/2) was used as the secondary Ab. Similar results were obtained with a biotinylated anti IdAb (Ab2.1-4 bio). Fig 5 The Vaccibodies exhibit binding to DNP, hence the antigenic scFv is correctly folded. The M315 Ab, from which the antigenic scFv is derived, is specific for the hapten DNP. ELISA plates were coated with DNP-BSA. Supernatants from cells transfected with genes encoding various Vaccibodies were added. Binding was detected with 9A8 bio.
  • Fig 6 APC pulsed with titrated amounts of MHC class II specific Vaccibodies stimulated polarized M315-specific T cells from TCR-transgenic SCTD mice >100-1000 fold better than the NIP-specific, untargeted control Vaccibodies. There were no significant differences between the Vaccibodies with a long sequence prior to the first disulfide bridge in the hinge (hl+h4) compared to the Vaccibodies with a short hinge sequence above the first disulfide bridge (h4).
  • the MHC class II-specific Vaccibodies induce a strong anti-Id Ab response in the absence of adjuvant.
  • BALB/c mice were injected with Vaccibodies, 20 ⁇ g and 200 ⁇ g, respectively. Blood samples were taken on different time points for sera analysis. Shown is data from sera taken on day 28, 14 days after the second immunization of Vaccibodies.
  • the MHC class II-specific Vaccibodies induced a strong anti-Id Ab response.
  • the Vaccibodies with the longest hinge (hl+h4) induced the strongest anti-Id Ab response reaching 3-4 ⁇ g/ml in sera.
  • Fig 8 Construction of the two hinge-C ⁇ 3 variants derived from hIgG3 by PCR.
  • the templates were from pUC19 containing modified hIgG3 constant regions were the h4 exon were connected to the CH3 domain (A) or the hi exon were connected to the h4 exon further connected to the CH3 domain (B) (Olafsen T et al, 1998).
  • the primers inserted Hindlll (5') and Sfil (3') restriction enzyme sites.
  • the hinge and CH3 domain are connected by a triplicate of the amino acids GlyGlyGlySerSer.
  • Fig 9 Construction of the hinge-C ⁇ 3 segments derived from mIgG2b.
  • the hinge and the CH3 genes were amplified from a pUC18 vector containing the constant region of mIgG2b by PCR with two primers encoding a Hindlll (5') and a Sfil (3') restriction enzyme site. The two PCR fragments were joined by PCR SOEing. In this reaction, the hinge and C H 3 domain were connected by a triplication of the aniino acids Gly-Gly-Gly- Ser-Ser.
  • Fig 10 Construction of the scFv derived from the myeloma protein M315.
  • the cDNA that functioned as a template in the PCR reactions were derived from RNA extracted from MOPC315.4 cells.
  • the V regions were joined by PCR SOEing resulting in a scFv. hi this reaction, the V regions were connected by a triplicate of GlyGlyGlyGlySer.
  • the gene fragments encoding the complete scFv were flanked by Sfil and Sail restriction enzyme sites.
  • Fig 12 Subcloning of the hinge-C ⁇ 3-M315 scFv into pUC19.
  • Three different dimerization motifs were included, derived from mIgG2b or IgG3. In all cases, they consisted of hinge followed by a triplicate of GlyGlyGlySerSer and CH3. Two different hinges were derived from gG3, one consisting of hi linked to h4, and one consisting of h4, only.
  • V regions specific for ND? and MHCII The V regions were amplified and joined by PCR soeing resulting in scFvs.
  • the linker connecting the V regions consists of a triplicate of GlyGlyGlyGlySer.
  • the gene fragments encoding the complete scFvs are flanked by Bsml/Munl and BsiWI sites. Linkers and restriction sites were introduced in the PCR reactions.
  • Fig 19 Detailed figure of Vaccibody gene construct.
  • the targeting unit is inserted between the Bsml/Mfel and BsiWI restriction enzyme sites (The V cassette of the pLNOH2 vector).
  • the hinge-C ⁇ 3-Fv 315 is inserted between the Hindlll and BamHI sites into the C cassette of pLNOH2.
  • the hinge and the C ⁇ 3 domain as well as the two scFv's are connected with (G 4 S) 3 linkers (black boxes).
  • the C ⁇ 3 and the Fv 315 are connected through a GLSGL linker.
  • the Fv 315 is inserted between two nonidentical Sfil restriction enzyme sites.
  • the antigenic unit and dimerization motif may be of any origin appropriate. Also, functional fragments of C ⁇ 3 may be employed, or a sequence which is substantially homologous to the C ⁇ 3 sequence or C ⁇ 3 fragments.
  • Vaccibodies are secreted as functional molecules.
  • Two distinct Vaccibodies were tested, one carrying the hapten specific Fv NIP as targeting unit, while the other carried the MHC class II-specific Fv I_E as targeting unit. Both carried the scFv from M315 (Fv 3 5 ) as antigenic unit, a) 10% SDS-PAGE of metabolically labeled Vaccibodies immunoprecipitated from culture supernatants of transfectants with or without reduction of disulfide bonds by mercaptoethanol (ME), b) DNP-specificity of the Vaccibodies was measured by ELISA. Supernatants from NSO cells transfected with Vaccibodies were added to ELISA plates coated with DNP-BSA.
  • NTP-specificity was measured by ELISA.
  • ELISA plates were coated with NIP-BSA.
  • Vaccibodies in both b) and c) were detected by either 9A8- bio ( ⁇ V ⁇ l/2) or Ab2.1-4 (specific for Id of Fv 315 )
  • Fv 315 carries V ⁇ 2 and Fv N ⁇ > carries V ⁇ l and will both bind 9A8 mAb. Only Fv315 will bind Ab2.1-4 mAb.
  • Fv I_E carries VK and will bind neither of the mAbs.
  • Fig 21 Production of Vaccibodies by intramucular injection of naked DNA plasmids followed by in vivo electroporation.
  • Vaccibody plasmids were injected into I-E d positive BALB/c mice, which were subsequently electroporated.
  • a) Level of Vaccibodies in sera was measured by ELISA, with DNP-BSA and 9A8bio as described previously, b) The same day 14 sera samples were analyzed for anti-Id antibodies by ELISA. Microtiter plates were coated with M315 and 187-bio (anti- mouse K Ab) was used for detection, c) Comparison of detectable Vaccibody levels and anti-Id antibodies. The amount of detectable Vaccibodies in sera is shown on the y-axis and the level of ⁇ ld-Abs is shown on the x-axis.
  • Fig 22 Tumor avoidance BALB/c mice were immunized once with naked plasmids encoding MHC class II specific Vaccibodies (Fv I_E Fv 315 ), nontargeted NIP-specific Vaccibodies (Fv NIP Fv 315 ) or 0.9% NaCl by i.m. immunization into the two quadriceps muscles (25 ⁇ g/muscle) followed by in vivo electroporation. They were challenged with l. ⁇ xlO 5 MOPC315.4 myeloma cells s.c. and the first day tumor take were recorded. A tumor of >3 mm was scored as positive tumor take.
  • mice were immunized once with naked plasmids encoding MHC class II specific Vaccibodies (Fv " Fv ), nontargeted NIP-specific Vaccibodies (Fv Fv ) or 0.9% NaCl by i.m. immunization into the two quadriceps muscles (25 ⁇ g/muscle) followed by in vivo electroporation. They were challenged with 1.6x10 s MOPC315.4 myeloma cells s.c. and their survival were compared.
  • Fv Fv MHC class II specific Vaccibodies
  • Fv Fv Fv nontargeted NIP-specific Vaccibodies
  • 0.9% NaCl 0.9% NaCl
  • M315 in sera samples were measured by ELISA coated with anti-Id-mAb (Ab2.1-4) and detected by biotinylated anti-IgA mAb (8D2).
  • MJJP-l ⁇ Fv 315 has mouse macrophage inflammatory protein 1 ⁇ as the targeting unit and scFv from M315 (Fv 315 ) as the antigenic unit.
  • Functionality of MJP-l ⁇ in Vaccibody format was measured in ELISA. Supernatants from 293E cells transfected with Vaccibodies were added to ELISA plates coated with anti-mouse MlP-l ⁇ mAb (R&D Systems) and detected by 9A8-bio ( ⁇ V ⁇ l/2).
  • the present invention relates to a recombinant human antibody-based molecule, called Vaccibodies, comprising dimers of a monomeric unit that consist of a single chain fragment variable (scFv) of immunoglobulins (Ig) with specificity for surface molecules on Ag presenting cells (APC), connected through a hinge region and a C ⁇ 3 domain to a scFv in the COOH-terminal end, the latter being derived from a myeloma protein (Fig 1), although any origin is possible due to the cassette cloning system of the expression vector.
  • scFv single chain fragment variable
  • Ig immunoglobulins
  • APC Ag presenting cells
  • the dimeric product will include two preferably identical scFvs with binding specificity for the same surface molecules on APC (Fig 2), enabling bivalent binding.
  • Bivalent binding i.e. crosslinking
  • Fig 2 a surface molecules on APC
  • bivalent binding provides increased binding strength due to avidity effects, and increases the likelihood of receptor mediated endocytosis into the APC and subsequent degradation inside the APC.
  • the bivalent binding may provide important receptor mediated intracellular signaling to the APC.
  • the scFvs with a targeting function are either derived from B cell liybridomas expressing monoclonal antibodies (mAbs) that bind to surface molecules on APC, or they may be derived from any source, e.g. phage display libraries.
  • mAbs monoclonal antibodies
  • the use of scFvs from B cell hybridomas as the targeting moiety opens for a great range of possible targets due to the large collection of B cell hybridomas that produce mAbs which bind different surface molecules on APC.
  • one may choose the nature of the signal given to the targeted cell by employing agonistic or antagonistic mAbs.
  • Growing knowledge of Ab-Ag interactions will allow the improvement of the binding affinity of such mAbs to their Ag by amino acid replacements in the binding sites.
  • Vaccibodies can be targeted to said surface molecules by means of targeting units comprising for example antibody binding regions with specificity for e.g HLA, HLA-DP, CD14, CD40, or Tolllike receptor; ligands, e.g.
  • soluble CD40 ligand a compound that has a high affinity for CD40 ligand.
  • chemokines e.g. RANTES or MTP-l ⁇
  • bacterial antigens like e.g. flagellin or a part thereof.
  • the targeting unit is inserted into the V cassette of the expression vector pLNOH 2 (Norderhaug, Olafsen et al. 1997), it is easily exchanged with other units (Fig 17).
  • the crucial dimerization motifs in the Vaccibodies constructed in the examples so far, include hinge regions and an immunoglobulin domain (e.g. C ⁇ 3 domain), e.g. carboxyterminal C domain (C H 3 domain), or a sequence that is substantially homologous to said C domain.
  • the hinge is Ig derived and contributes to the dimerization through the formation of an interchain covalent bond(s), e.g. disulfide bridge(s).
  • it functions as a flexible spacer between the domains allowing the two scFvs with targeting tasks to bind simultaneously to two target molecules expressed with variable distances (Fig 2).
  • the immunoglobulin domains contribute to homodimerization through noncovalent interactions, e.g. hydrophobic interactions, h a preferred embodiment the Cjj3 domain is derived from IgG.
  • These dimerization motifs can be exchanged with other multimerization moieties (e.g from other Ig isotypes/subclasses).
  • the C-terminal scFv derived from the monoclonal Ig produced by myeloma or lymphoma cells also called the myeloma/lymphoma M component, can be genetically exchanged with other scFvs or any antigen because the vector has been constructed with a Sfi I restriction site (Fig 8). Therefore, the scFv derived from the model myeloma protein used in the present example is easily swapped with scFv from the M component of any patient with B cell lymphoma or multiple myeloma. Thus, this vector will allow for rapid construction of individual patient specific vaccines.
  • the dimeric structure of the Vaccibody not only affords crosslinking, but should also allow double loading of the patient Fv to the APC per Vaccibody molecule compared to a single scFv combined to a single targeting moiety. Furthermore, there is in a single Vaccibody duplication of serological idiotypic determinants, which might be of importance for the anti-Id B cell response.
  • the Vaccibodies lack a C H 2 domain and hence all FcR binding sites, and should therefore exclusively be taken up through its target molecules, such as MHC class II in the example used and not by any FcRs, ensuring that a large proportion of the vaccine will arrive at the intended target cells. This is in contrast to vaccines that exert their effect through the binding to an FcR on a target cell (Ravetch and Bolland 2001).
  • Vaccibodies can be extended to a general medical treatment through induction of an immune response against any polypeptide of any origin. It is possible to replace the idiotypic scFv with other antigenic sequences of sufficient length to allow proper folding of the polypeptide.
  • This sequence may be derived from other cancer proteins or infectious agents. Examples of such cancer sequences are telomerase, more specifically hTERT.
  • the infectious agents can be of bacterial, e.g. tuberculosis antigens, or viral origin, more specifically HIV derived sequences like e.g. gpl20 derived sequences.
  • Vaccibodies that are described herein, which comprise the idiotypic scFv.
  • the scFvs on the NH 2 - terminal end of the Vaccibodies target the Vaccibodies to
  • MHC class II is expressed on all professional APC, so the Vaccibodies described herein are able to target B cells, DC and macrophages.
  • Targeting of conventional Ag-Ab complexes to MHC class II induces activation of specific CD4+ T cells (Snider and Segal 1987; Casten and Pierce 1988) .
  • Targeting of Troybodies to MHC class II has previously been shown to enhance Ag presentation and T cell activation in vitro as well as in vivo (Lunde, Western et al. 2002).
  • the Vaccibodies of the examples contain the scFv of the myeloma protein M315 derived from the BALB/c plasmacytoma MOPC315.4.
  • the ⁇ 2 315 light chain of M315 harbors three defined somatic mutations in the CDR3 loop and functions as a model idiotypic T cell epitope in a well defined system (Bogen, Malissen et al. 1986; Bogen and Lambris 1989).
  • the Vaccibodies have been genetically assembled, and the DNA transfected into NSO cells, 293E cells and Cos-7 cells. Transfectants produce and secrete the recombinant Vaccibody molecules.
  • the targeting scFvs at one end of the Vaccibodies exhibit binding to MHC class II (Fig 3).
  • the antigenic scFv at the other end of the Vaccibody binds DNP (di-nitro-phenyl, the specificity of M315) (Fig 5).
  • DNP di-nitro-phenyl, the specificity of M315)
  • Vaccibodies have the ability to induce strong T cell responses through their binding to APC and presentation of Id-peptides on class II molecules to Id- specific CD4+ T cells in vitro (Fig 6). Furthermore, since they have intact Fv of the M- component, they elicit anti-Id antibodies in significant amounts in vivo when injected into BALB/c mice without adjuvants (Fig 7).
  • mice are immunized with DNA encoding patient specific Vaccibodies by in vivo electroporation of muscle cells.
  • Sera from immunized mice are absorbed on anti human Ig-Sepharose to remove crossreactive antibodies and thereafter eluted to obtain purified highly Id-specific antibodies.
  • Sandwich ELISAs specific for each patient's myeloma are performed as follows: The purified anti-Id Ab from mice is coated in wells. Serum from the patient in casu is added. Myeloma protein binding to anti-Id antibodies will be detected by use of Ab specific for human IgG or IgA. The sensitivity of such sandwich ELISAs is usually ⁇ 1 ng/ml, which is > 10 6 times more sensitive than routine assays. Furthermore, to monitor development of new T cells, profile of T cells in blood will be monitored by flow cytometry with V ⁇ -specific mAbs, in combination with other markers.
  • the present invention relates to a pharmaceutical comprising the above described recombinant based antibody, DNA/RNA sequences, or expression vectors according to the invention.
  • this pharmaceutical additionally comprises a pharmaceutically compatible carrier.
  • Suitable carriers and the formulation of such pharmaceuticals are known to a person skilled in the art. Suitable carriers are e.g phosphate-buffered common salt solutions, water, emulsions, e.g. oil/water emulsions, wetting agents, sterile solutions etc.
  • the pharmaceuticals may be administered orally or parenterally.
  • the methods of parenteral administration comprise the topical, intra- arterial, intramuscular, subcutaneous, intramedullary, intrathekal, intraventricular, intravenous, intraperitoneal or intranasal administration.
  • the suitable dose is determined by the attending physician and depends on different factors, e.g. the patient's age, sex and weight, the kind of administration etc.
  • the present invention relates to a vaccine composition against cancer or infectious diseases comprising an immunologically effective amount of the nucleic acid encoding the molecule of the invention or degenerate variants thereof, wherein said composition is able to trigger both a T-cell- and B-cell immuiie response.
  • the present invention also relates to a kit comprising Vaccibody DNA, RNA, or protein for diagnostic, medical or scientific purposes.
  • the invention further relates to a method of preparing the recombinant molecule of the invention comprising, transfecting the vector comprising the molecule of the invention into a cell population; culturing the cell population; collecting recombinant protein expressed from the cell population; and purifying the expressed protein.
  • nucleotide sequences may preferably be inserted into a vector suited for gene therapy, e.g. under the control of a specific promoter, and introduced into the cells.
  • the vector comprising said DNA sequence is a virus, e.g an adenovirus, vaccinia virus or an adeno-associated virus.
  • Retroviruses are particularly preferred.
  • suitable retroviruses are e.g.
  • the DNA/RNA sequences according to the invention can also be transported to the target cells in the form of colloidal dispersions. They comprise e.g. liposomes or lipoplexes.
  • naked Vaccibody DNA construct is injected intra-muscularly into mice, whereupon the site of injection is subject to in vivo electroporation.
  • This DNA vaccination resulted in production of Vaccibody protein which conferred life-saving protective immunity on a majority of the mice.
  • example 4 demonstrates that after a single injection of Vaccibody plasmid in quadriceps and electroporation, F V NIP F v 315 Vaccibody protein was detected in significant amounts in serum, while there was no detectable F V I_E F v 315 (Fig 21a).
  • example 5 shows the presence of anti-Id antibodies in serum after injection of Vaccibody DNA intramuscularly and electroporation.
  • example 6 shows that protection against myeloma can be achieved by i.m. DNA vaccine followed by electroporation and that the protection requires targeting of the tumor-derived scFv to APC.
  • Example 7 demonstrates that chemokines are functional as targeting units in the Vaccibody format.
  • mice BALB/cABom were from Bomholtgaard (Ry, Denmark).
  • the ⁇ 2 315 -specific TCR- transgenic mice on a BALB/c background (Bogen, Gleditsch et al. 1992)were bred in our animal facility.
  • the gene for the hIgG3 hinge and C H 3 domain was cloned from the pUC19 vector containing hinge genetically combined with C ⁇ 3 genes of the hIgG3 subclass (Olafsen, Rasmussen et al. 1998). Two variants of the hinge length in the humanized Vaccibodies were made; one with just the h4 exon connected to the C H 3 domain (sh) and one with both exon hi and h4 connected to the C H 3 domain (Ih) (Fig 8).
  • the primers included restriction enzyme sites (underlined): 5'h4: tag caa get tgg cca gcg cag gga g; 3 5 CH3: cag gee acc Rag gee ttt acc egg aga cag gga.
  • the hi exon were introduced directly upstream of the h4 exon by QuickChange PCR using these primers
  • Qhla ctccaatcttctctgca gag etc aaa acc cca ctt ggt gac aca act cac aca gag ccc aaa tct tgt gac ac
  • Qhlb gt gtc aca aga ttt ggg etc tgt gtg agt tgt gtc acc aag tgg ggt tt gag etc tgcagagagaagattgggag.
  • the murine Vaccibodies have a complete hinge and C R 3 domain of the mIgG2b subclass picked up by PCR from a pUC18 vector containing the C ⁇ 2b genes (Fig 9).
  • the primers included restriction enzyme sites (underlined) or linkers (bold) with the complementary sequences (italic): 5 'hinge: tagcaagctt ca gag ccc age ggg ccc; 3 'hinge: 5' tec acc tec get get tec acc gcc tgg gca ttt gtg aca etc ctt g; 5'C H 3 : gga age age gga ggt gga agt gga ggg eta gtc aga get cca ca; 3'C H 3 : cag gcc acc gag gcc acc egg aga ccg gga gat g.
  • the hinge and the CH3 domain were then joined by PCR SOEing.
  • the Antigenic V region genes were cloned from the plasmacytoma MOPC315.4 (Eisen, Simms et al. 1968). The V regions were obtained by extracting mRNA from the MOPC315.4 cell line with oligo (dT)-coated magnetic Dynabeads (Dynal). First strand cDNA were then made and used as template for PCR amplification of the V region genes using specific primers annealing to the exact ends of the M315 V region sequences. The primers included restriction enzyme sites (underlined) or linkers (bold) with the complementary sequences (italic).
  • the primer sequences were: 5'VH: ggc etc ggt ggc ctg gat gta cag ctt cag gag tea; 3'V H : gcc aga gcc acc tec gcc iga tec gcc tec acc tga gga gac tgt gag agt ggt; 5'V L : ggc gg ⁇ ggt ggc tctggc ggt ggc gga teg cag get gtt gtg act cag gaa; 3'V L : gacg tcgac tag gac agt gac ctt ggt tec.
  • the V H and V L genes were then joined by PCR soeing to a scFv format (Fig 10).
  • Two BamHI restriction enzyme sites inside the V regions of M315 were removed by QuickChange PCR (Fig 13) using primers: BamHI V L l:at gcc aac tgg ata caa gaa aaa cc; BamHI V L 2 gg ttt ttc ttg tat cca gtt ggc at, BamHI V H 1 : tgg aac tgg ata egg cag ttt cc and BamHI H 2 gg aaa ctg ccg tat cca gtt cca.
  • V region genes providing specificity for MHC class II had previously been cloned from the 14-4-4S hybridoma (Lunde, Western et al. 2002), which produces an Ab specific for the E ⁇ chain (determinant la.7) of the I-E MHC class II molecule (Ozato, Mayer et al. 1980).
  • the primer sequences were: 5'V L : gac att caattg aca cag tct tct cct get tec; 3 'V L : gcc aga gcc acc tec gcc aga tec gcc tec acc ttt gat ttc cag ctt ggt gcc; 5' V H : ggc gga ggt ggc tct ggc ggc ggt ggc gga teg cag gtc cag ctg cag cag t; 3'V H : ga cgtacg actcacc tga gga gac ggt gac tga gg.
  • V region genes conferring specificity for the hapten NIP were designed with the similar tag sequences except for the 5'V L primer: 5'V L : ggtg tgcattcc cag get gtt gtg act cag gaa; 3'V L : gcc aga gcc acc tec gcc aga tec gcc tec acc tag gac agt cag ttt ggt acc t; 5'V H : ggc gga ggt ggc tct ggc ggc gga gga teg cag gtc caa ctg cag cag cc; 3'V H : ga cgtacg a etc acc tga gga gac tgt gag agt ggt.
  • N L and N H were then joined by PCR SOEing (Fig 16) and subcloned into the N cassette pL ⁇ OH 2 vector containing the hinge-CH3-scFvM315 genes (Fig 17 and Fig 18).
  • other N genes conferring a desired specificity are isolated from hybridomas or from phage selected from phage display libraries. They are then PCR amplified using primers designed in the same manner as above and subcloned after PCR SOEing in the targeting-cassette (Fig. 17 and 18).
  • Rearranged N H and VK genes conferring specificity for HLA-DP were PCR amplified from cD ⁇ A from the 22C1 hybridoma, which produces an antibody with pan HLA-DP specificity.
  • V genes were reamplified with new primers containing sites for direct cloning into the expression vectors pL ⁇ OK and pL ⁇ OH2 (Norderhaug and Olafsen, 1997); 5 5 -V L, ggt tgeatteeg ⁇ c ⁇ ttgtgctc ce; 3 '-V L , cgtacgttctacteaegtttt ⁇ ttec ⁇ get,- 5 5 -V H , gtgcattccg ⁇ t c ⁇ gctgc ⁇ g ⁇ gtct; 3'-V H , cgtacgaetcacctg ⁇ gg ⁇ g ⁇ ccgt ⁇ gc.
  • scFV was generated by PCR SOEing using the following primers: 5'V L , g gtg tgcattc cga cat tgt get cac c 3'V L : gcc aga gcc acc tec gcc aga tec gcc tec acc gtt tta ttt cca get 5 ' V H : ggc gga ggt ggc tct ggc ggt ggc gga teg gag gtg cag ctg cag gag tct 3'V H , cgtacg act cac ctg agg aga ccg tag c
  • First strand cDNA was made and used as template for PCR amplification of chemokine genes (RANTES and MlP-l ⁇ ) using specific primers: 5'MIP-l ⁇ : ggtg tgcattc cgc gcc ata tgg age tga cac, 3'MTP-l ⁇ : ga cgtacg act cac ctg cat tea gtt cca ggt cag tg 5 'RANTES: ggtg tgcattc c gcc tea cca tat ggc teg g 3 'RANTES: ga cgtacg a etc acc tga cat etc caa ata gtt gat gta ttc.
  • the different targeting unit genes were then digested with Muni and BsiWI or Bsml and BsiWI, respectively and subcloned into the V cassette pLNOH vector containing the hinge-CH3-scFvM315 genes (Fig 17 and Fig 18).
  • CD40 ligand is cloned from T cells that are activated with LPS for 4 hours before mRNA is extracted for preparation of cDNA.
  • the cDNA is used as template in a PCR reaction with primers specific for the CD40 ligand sequence. Furthermore, this sequence is reamplified with primers designed to facilitate subcloning in the targeting cassette as described above.
  • Vaccibodies Production and purification of Vaccibodies
  • the pLNOH 2 vector carrying the Vaccibody genes was transfected into NSO cells by electroporation, and supernatants from single colonies resistant to 800 ⁇ g/ml G418 were analysed for Vaccibody secretion after 2-3 weeks, using ELISA.
  • DNP-BSA was used as coat, and biotinylated rat-anti mouse V ⁇ l/2 (9A8-bio) was used for detection.
  • the NIP- specific Vaccibodies were additionally screened in an ELISA using NIP-BSA as coat.
  • the cells selected for high Vaccibody production were grown in Rollerbottles (VWR) and affinity purified from supernatant using a column made by DNP-lysine (Sigma) coupled to fast flow Sepharose.
  • Vaccibodies were eluted with 0.05M DNP-glycine (Sigma) and the flow-through was run on an ion-exchange Cl " Dowex 1x8 resin column (Sigma). The eluted Vaccibodies were dialyzed against PBS/0.05%NaN3 and sterile PBS, before the vaccibody concentration were calculated from absorbance values at 280nm.
  • Ab and reagents used for flow cytometry were 9A8 biotin, FGK.45 biotin, streptavidin PerCP, anti-CD 19 PE and anti-mIgG2a PE (BD Pharmingen).
  • BALB/c spleen cells were double stained with anti-CD 19 PE and Vaccibodies. Bound Vaccibodies were detected by 9A8-bio and streptavidin PerCP. Twenty thousand cells were run on FACSCalibur (BD Biosciences, Mountain View, CA) and analyzed using the WinMDI software.
  • the beads were washed three times in ice cold PBS with 1%NP40 and resuspended in lO ⁇ l lx sample buffer.
  • the proteins were eluted from the beads by incubating the samples at 95°C for 3 minutes.
  • the Vaccibodies were run on a 10% SDS-PAGE gel, with a 5% stacking gel, at 40mA for lh, using a BIO RAD miniprotean II gel electrophoresis apparatus.
  • the gels were subsequently fixed in 30% methanol and 10% acetic acid for 30 minutes prior to 30 min incubation with Amplify (Amersham), before drying and exposing to BIOMAX-MR film (Eastman Kodak Company, Utah, USA).
  • T cell proliferation assay Irradiated (2000rad) BALB/c splenocytes (5xl0 5 cells/well) were used as a source of APC. Titrated amounts of different MHC CLASS II- and NIP-specific Vaccibodies were added to the splenocytes. A 91-107 ⁇ 2 synthetic peptide were used as a positive control. The assays were put up in 150 ⁇ l cultures in 96-well flat-bottom microtiter wells and incubated for 4 h at 37°C (Lunde, Western et al. 2002). The cultures were then washed three times before addition of 200 ⁇ l polarized ⁇ 2 315 -specific Th2 cells (2xl0 4 ) derived from TCR transgenic SCID mice.
  • mice were injected subcutaneously (s.c) with 200 ⁇ g or 20 ⁇ g purified Vaccibody proteins in PBS on day 0, 14 and 28. Blood samples were taken on day 14 and 28 before revaccination and then on day 35, 42 and 49, before they were sacrificed according to the
  • Anti-idiotypic Abs against M315 were measured by ELISA.
  • the wells were coated with 2 ⁇ g/ml M315.
  • Anti-Id Ab in the sera were detected by a biotinylated anti-mouse VK Ab (187.1 bio), anti-mouse IgGl bio or anti-mouse IgG2a bio (both from BD Pharmingen).
  • Ab2.1-4 an anti-Id mAb that bind ⁇ 2 315 ) was used as standard.
  • Protein vaccination BALB/c mice were injected subcutaneously (s.c) in the right flank region with 20 ⁇ g or 200 ⁇ g purified class II- or NIP specific Vaccibodies (F V I_E F v 315 , F V NIP F v 315 ) in PBS on day 0 and 14. PBS was injected as negative control. Blood samples were collected from the leg vein on different time points after the last immunization. Anti-idiotypic antibodies with specificity with specificity for F v 315 were measured by ELISA. The wells were coated with 2 ⁇ g/ml M315. Anti-Id Ab in the sera were detected by a biotinylated anti-mouse K mAb (187.1 bio). Ab2.1-4 (an anti-Id mAb binding F v 315 (Kristoffersen, Hannestad et al. 1987) was used as standard.
  • mice Five to ten weeks old Balb/c mice were purchased from Bomholtgaard (Ry, Denmark). The animals were anaesthetized by intraperitonal injection with 9 g Pentobarbital/mice and the legs were shaved. Conductive gel was applied at the skin and 50 ⁇ l vector DNA diluted in 0.9% NaCl, was injected into the quadriceps.
  • electroporation was performed, by applying rod electrodes to the skin near the site of the injection and subjecting the site to an electrical potential comprising 10 trains of 1000 pulses each, with a pulse length at two times 200 Sec (positive 200Sec and negative 200 Sec) with 600 s interval between each pulse and with a current limit of 50 mA (about 150-174 V/cm) (Tollefsen, Tjelle et al. 2002).
  • Blood samples were collected from the leg vein on different time points and heart puncture was performed on the day they were sacrificed. Serum samples were analyzed for the presence of correctly folded Vaccibodies.
  • the ELISA was performed with DNP-BSA as coat and 9A8-bio as detected Ab, as described above. In addition, serum samples were analyzed for anti-Id Abs by ELISA as described above.
  • Tumor challenge Protein Vaccibodies-MOPC315.4 BALB/c mice (6-10 weeks old) were injected s.c. with 160 ⁇ g class Il-or NIP-specific Vaccibodies in PBS in the right flank region on day 0 and 14. On day 28, 1.6xl0 5 MOPC315.4 cells were injected s.c. on the right flank. Mice were inspected twice weekly. Tumor size development was monitored by palpation and use of a caliper. A tumor of 3 mm in diameter was scored as tumor take. Mice were killed when tumor size reached 20 mm with no sign of tumor necrosis.
  • DNA Vaccibodies-MOPC315.4 DNA vaccination was performed at day 0 as described above. On day 14, 1.6xl0 5 MOPC315.4 cells were injected s.c. in the right flank region. Tumor size development was monitored by palpation and use of a caliper. The mice were sacrificed when the tumor size reached 20 mm. Blood samples were collected on different time points from the leg vein. Levels of M315 myeloma protein in sera were quantified in a sandwich ELISA with Ab2.1-4 as coat by biotinylated anti-C ⁇ (8D2) mAb as detection Ab. Tumor size, tumor take, survival curves and statistical analyses were calculated by use of Graph Pad Prism 3.0 software (San Diego, CA).
  • Vaccibodies bind APC and are able to trigger both T cell and B cell immune response. Moreover, the following experiments show that Vaccibodies induce a strong immune response rendering adjuvants redundant.
  • the experiments demonstrate that said molecule is capable of inducing an iirimune response against multiple myeloma and, further, the feasibility of treatment of mammals by immunization by means of Vaccibody DNA or Vaccibody protein.
  • the experiments also demonstrate that another attractive approach is to target the Vaccibodies to surface molecules expressed exclusively on subsets of dendritic cells (DC), like e.g. chemokine receptors.
  • DC dendritic cells
  • the following examples are meant to illustrate how to make and use the invention. They are not intended to limit the scope of the invention in any manner or to any degree.
  • Vaccibodies are produced and secreted as functional dimerized molecules and is itself bound by the anti-V ⁇ l/2 antibody (9A8) (Bogen 1989) and the anti-idiotypic antibody Ab2.1-4 (Lauritzsen, Weiss et al. 1994).
  • the M315 mAb binds the hapten di-nitro-phenyl (DNP) (Eisen, Simms et al. 1968). Therefore, to verify that Vaccibodies were produced, secreted and correctly folded as functional molecules, the antigenic units of Vaccibodies were tested in ELISA for their capability to bind DNP, 9A8 and Ab2.1-4 mAbs.
  • Fig 20b shows that both the NTP- specific and the MHCII-specific Vaccibodies, that both have scFv 315 , bind DNP, 9A8 and Ab2.1-4.
  • Vaccibodies were metabolically labelled by growth of transfected cells in medium containing 35 S-methionine, vaccibodies were immunoprecipitated from supernatant using specific antibodies, and analyzed by SDS-
  • MHC class II-specific Vaccibodies enhance ⁇ 2 315 -specific stimulation ofCD4 + T cells.
  • Class II-specific and non-targeting NIP-specific Vaccibodies were mixed with antigen presenting cells (APC) and compared for their ability to induce specific T cell activation.
  • APC antigen presenting cells
  • Irradiated BALB/c splenocytes were used as APC.
  • the BALB/c strain has the H-2 d haplotype, hence they express I-E d molecules necessary for both targeting of the MHC II-specific Vaccibodies and presentation of the ⁇ 2 315 epitope to specific CD4 + T cells.
  • the APC were pulsed with the different Vaccibodies for 4h and subsequently washed. Washing was performed to reduce the chance that I-E -specific Vaccibodies in the culture medium could diminish T cell stimulation by blocking I-E d molecules (Lunde, Western et al. 2002).
  • Polarized Th2 cells from mice transgenic for a ⁇ 2 315 - specific I-E d restricted TCR were added as responder T cells.
  • the dose response curve in fig 6 shows that the ⁇ 2 epitope was presented 100- 1000 times more efficiently to TCR-transgenic Th2 cells when they were carried in the APC-targeted MHC II-specific Vaccibodies (both the ones with short and long human dimerization units) compared to the non-targeted NIP-specific Vaccibodies. It should be noted that Vaccibodies do not include an Fc ⁇ R binding site; hence the NIP-specific Vaccibodies should not be able to enter cells via receptor-mediated endocytosis.
  • Protein Vaccibodies detected in serum after injection of DNA intramuscularly and in vivo electroporation Protein Vaccibodies detected in serum after injection of DNA intramuscularly and in vivo electroporation.
  • Vaccibodies were produced by i.m. plasmid injection and electroporation. Since the F V I_
  • E F v 315 Vaccibodies are specific for I-E d molecules present in BALB/c (H-2 ), these
  • Vaccibodies should be rapidly absorbed by the I-E d positive cells in BALB/c.
  • the non-targeted F V NIP F v 315 Vaccibodies should not be absorbed. Indeed, 14 days after a single injection of 50 ⁇ g Vaccibody plasmid in quadriceps and electroporation, F V NIP F v 315 Vaccibody protein was detected in significant amounts in serum, while there was no detectable (Fig 21a).
  • mice i.m. injected/electroporated with the MHC class II -targetd F V I_E F v 315 Vaccibody DNA had developed antibodies that bound idiotypic Fv from the MOPC315.4 tumor (Fig 21b). This result was in distinct contrast to the lack of any anti- idiotypic antibody response in mice injected with the non-targeted F V NIP F v 315 Vaccibody DNA (Fig 21b).
  • the results demonstrate a daunting effect of targeting to MHC class II (I-E d ) positive cells for development of a strong humoral response.
  • Control mice injected i.m. with 0.9% NaCl followed by electroporation had neither Vaccibodies nor anti-idiotypic Abs in day 14 sera (Fig 21a-c).
  • Chemokines are functional as targeting units in the Vaccibody format
  • a vector carrying the gene encoding a Vaccibody as outlined in Figure 18, but specifically with MTP-la in the targeting unit, long human hinge in the dimerization unit and M315 scFv in the antigenic unit was assembled. Supernatant from cells transfected with this construct was collected and tested in ELISA for the presence of
  • Vaccibodies binding to an anti-mouse MTP-la mAb and 9A8 bio The results showed that such Vaccibodies with desired specificity were produced, and furthermore, that they bound to anti-MTP-l ⁇ mAb, while the NTP-specific Vaccibodies did not (Fig 25).
  • the chemokine RANTES is functional as targeting unit in the Vaccibody format hi the same manner, a vector with a gene encoding Vaccibodies like those described in example 7 was produced, with the exception that the targeting unit was the mouse chemokine RANTES. Supernatant from cells transfected with this construct was collected and tested in ELISA for the presence of Vaccibodies. The experiment showed that this Vaccibody variant was expressed and exported as a functional molecule.
  • Anti-Toll-like-receptor 2 as targeting unit in the Vaccibody format
  • a vector with a gene encoding Vaccibodies like those described in example 7 was produced, with the exception that the targeting unit was a scFv with specificity for toll-like-receptor 2 from the mouse.
  • Supernatant from cells transfected with this construct will be collected and tested in ELISA for the presence of Vaccibodies.
  • Anti-CD14 is functional as targeting units in the Vaccibody format
  • a vector with a gene encoding Vaccibodies like those described in example 7 was produced, with the exception that the targeting unit was scFv with specificity for human CD 14.
  • Supernatant from cells transfected with this construct was collected and tested in ELISA for the presence of Vaccibodies. The results showed that this Vaccibody variant was expressed and exported as a functional molecule.
  • Anti-HLA-DP is functional as targeting units in the Vaccibody format
  • Tuberculosis antigen in the Vaccibody antigenic cassette is Tuberculosis antigen in the Vaccibody antigenic cassette.
  • a nucleic acid encoding a tuberculosis antigen (cattle antigen) will be inserted into the antigenic unit of the Vaccibody construct.
  • telomerase antigen in the Vaccibody antigenic cassette hTERT an antigenic region of the telomerase ribonucleoprotein, will be inserted into the antigenic unit of the Vaccibody construct.
  • a nucleic acid encoding a gpl20 derived molecule will be inserted into the antigenic unit of the Vaccibody construct.
  • Vaccibodies with patient specific scFv of myeloma origin in the antigenic cassette are Vaccibodies with patient specific scFv of myeloma origin in the antigenic cassette.
  • Bone marrow aspirate from patients suffering from multiple myeloma is collected.
  • the mononuclear cells (MNC) are separated using a density gradient solution of Ficoll-
  • RNA is isolated (TRIzol® Reagent from h vitrogenTM Life Technologies) from MNC, and cDNA is made from mRNA (First-Strand cDNA Synthesis Kit from Amersham Biosciences (Not I-d(T)18 bifunctional primer)). This cDNA is used as template in PCR with primers that amplify the V genes of the heavy or light chain of the multiple myeloma Ig.
  • the sense primers are family specific and localized in the leader regions (VH1-7, VK1-6 and VLl-10), and the anti-sense primers are localized in the first part of the C regions (one primer each for IgG, IgA, kappa and lambda).
  • PCR products are ligated into a vector (pGEM®-T Easy Vector from Promega), and transformed into E.colL DNA samples isolated from individual colonies are sequenced. Getting the same sequence from three different colonies originating from three different PCRs confirms that the V regions from the myeloma Ig have been isolated.
  • PCR SOEing is performed and reamplification is done with primers including tags with sites for Sfil as described in Figure 19. For one patient such primers had the sequence: 5'TAVH 5' ACGTAGGCCTCGGTGGCCTGCAGATCACCTTGAAGGAGTCT
  • the resulting vector is transiently transfected into 293E cells.
  • Supernatants are tested in ELISA for the presence of such Vaccibodies. They are also injected into BALB/c mice. The presence of anti-idiotypic antibodies is measured in ELISAs against serum from the mice and serum from the patients.

Abstract

The present invention relates to a novel type of human recombinant antibody-like molecules useful in the treatment of i.e. multiple myeloma. These molecules, called Vaccibodies, bind APC and are able to trigger both T cell and B cell immune response. Moreover, Vaccibodies bind divalently to APC to promote a more efficient initiation of an immune response. Hence, a major purpose of the present invention is to induce a strong immune response to render adjuvants redundant. Vaccibodies comprise a dimer of a monomeric unit that consists of a targeting unit with specificity for a surface molecule on APC, connected through a hinge region and a CϜ3 domain to a antigenic unit in the COOH-terminal end; the latter being of e.g. B cell lymphoma or myeloma origin (Fig 1), although any origin is possible due to the cassette cloning system in the expression vector. The said molecule is capable of inducing an immune response against multiple myeloma, but extension to a general vaccination strategy for any polypeptid should be feasible. The present invention also relates to a DNA sequence coding for this recombinant antibody based molecule, to expression vectors comprising these DNA sequences, cell lines comprising said expression vectors, to treatment of mammals preferentially by immunization by means of Vaccibody DNA, Vaccibody RNA, or Vaccibody protein, and finally to pharmaceuticals and a kit comprising the said molecules.

Description

MODIFIED ANTIBODY
REFERENCE
The present application claim priority back to US provisional application no. 60/450134, filed February 25, 2003. 0
FIELD OF THE INVENTION
The present invention relates to a recombinant human antibody-based molecule called s Vaccibodies, which are able to trigger both a T cell- and B cell immune response. More particularly, Naccibodies by themselves induce such strong in mune responses that adjuvants are not necessarily required. The present invention also relates to a method of treating e.g. multiple myeloma by means of the said Naccibodies.
0
BACKGROUND OF THE INVENTION
Multiple myeloma (MM) is a bone marrow cancer hi which a single plasma cell clone has turned malignant and produces monoclonal immunoglobulin (Ig). MM patients have S a very poor prognosis. Although high response rates and increased survival can be achieved using high dose chemotherapy followed by autologous or allogeneic stem cell grafting, the majority of patients relapse and few, if any, are cured.
Myeloma cells produce monoclonal Ig that is unique for each tumor and thus for each individual patient. Ig is composed of two identical heavy (H) and two identical light 0 (L) chains. L and H chains have highly diversified variable (V) regions, V and VH- VL and VH together form the Fv (fragment variable) that contains unique antigenic determinants called idiotopes (Id). Idiotopes collectively constitute the idiotype of the Fv (of the Ig in casu). Induction of an immune response against the idiotype, so called Id- vaccination is a promising strategy in treatment of B cell lymphomas and MM (Bendandi, Gocke et al. 1999) (Tao and Levy 1993) (Huang, Wu et al. 2004) (Hakim, Levy et al. 1996) (King, Spellerberg et al. 1998) (Biragyn, Tani et al. 1999; Biragyn, Ruffini et al. 2002), and both anti-idiotypic antibodies (Sirisinha and Eisen 1971; Hough, Eady et al. 1976) and Id-specific T cells (Lauritzsen, Weiss et al. 1994) may be of importance. However, Id is a weak self-Ag in its original context (as part of Ig). Therefore, for vaccine purposes, it is important to enhance the immunogenicity of Id.
T helper cells (CD4+ T cells) recognize their antigen (Ag) after it has been processed through engulfment of foreign proteins by APC, proteolytic breakdown into peptide fragments that are loaded onto MHC class II molecules and transported to the surface of the APC where the peptide-MHC complex is presented to T cell receptors (TCRs) of CD4 + T cells. Activated CD4+ T cells stimulate cytotoxic T cells (CD8+ T cells) and B cells with the corresponding Ag specificity, initiating a broad response against the original Ag. A major problem concerning Id-vaccination of MM patients is that Id-specific CD4 T cells in these patients, as extrapolated from experiments in mice [18], probably are tolerant to Id V-region determinants on the highly abundant myeloma protein. MM patients who have undergone autologous stem cell transplantation (ASCT) may be in an advantageous phase for Id- vaccination for the following two reasons: 1) relief from T cell tolerance to myeloma protein Id and 2) development of new T cells that can respond to Id- accination.
Targeting of T cell epitopes to surface molecules on APC with Troybodies (Lunde, Munthe et al. 1999), which are equipped with a T cell epitope incorporated in a loop in a constant Ig domain results in increased T cell stimulation by a factor of 100- 100000 (Lunde, Rasmussen et al. 2001). However, Troybodies do not include the Ag in its native conformation, such as Fv, and are therefore restricted to induction of T cell responses. Therefore, to induce an anti-Id B cell response and anti-Id Abs, it is necessary to include the complete Fv of the M component of the patient. As for induction of an anti-Id T cell response, an inclusion of the entire Fv will greatly increase the chance of including idiotope sequences binding the patient's HLA-molecules, which is a prerequisite for activation of Id-specific T cells. There have been several approaches for rendering idiotypes more immunogenic.
Protein vaccination with complete Id+ immunoglobulins (Ig) fused with granulocyte- macrophage colony-stimulating factor (GM-CSF) (Tao and Levy 1993), or CD40 ligand
(Huang, Wu et al. 2004) enhances the level of anti-Id antibodies and results in protection against B-cell lymphoma in mice. However, scFv-GM-CSF was effective only when injected as protein and not as a DNA vaccine (Hakim, Levy et al. 1996). On the other hand, DNA vaccination employing scFv fused to IL-lβ did induce tumor immunity (Hakim, Levy et al. 1996). In another approach, scFv has been genetically fused with fragment C from tetanus toxin and delivered as a DNA vaccine by intramuscular (i. .) injection. This strategy has resulted in increased levels of anti-Id antibodies, Id-specific CD4+ responses and protection against lymphoma a myelomas in mice. The mechanism of adjuvant activity of tetanus toxoid fragment C is unknown (King, Spellerberg et al. 1998). In a similar approach, scFv has been fused to chemokines like MCP3, IP 10 mDF2β (Biragyn, Tani et al. 1999; Biragyn, Ruffϊni et al. 2002) and has been used both as a DNA and as protein vaccine (Biragyn, Tani et al. 1999). In several of these studies, foreign T cell epitopes corresponding to TT fragment-C or unique fusion sequences could have contributed to responses. Heightened anti-Id antibody responses and tumor protection has been observed. The mechanism of action of scFv-chemokine is unknown. One possibility is that the chemokine moiety targets Fv to chemokine receptors on APC for enhanced delivery of scFv. Alternatively, chemokines attract APC to the site of injection. However, both the Fragment C and chemokine fusion strategies rely on monovalent binding to their target molecules (King, Spellerberg et al. 1998; Biragyn, Tani et al. 1999). This is of concern because crosslinking has been shown to be of importance for optimal stimulation of T cells, e.g. for Troybodies (Lunde, Munthe et al. 1999).
With these considerations in mind, the inventors have designed a novel type of recombinant antibody-like molecules called Vaccibodies, a divalent molecule comprising a flexible hinge, with no FcR binding and that contain the Ag in its native conformation, with the purpose of inducing both strong Id-specific Ab and T cell responses. Vaccibodies are large and complex macromolecules, but, surprisingly, cells were able to produce and export intact molecules. SUMMARY OF THE INVENTION
The present invention relates to a novel type of human recombinant antibody-like molecules useful in the treatment of i.e. multiple myeloma. These molecules, called Vaccibodies, bind APC and are able to trigger both T cell and B cell immune response. Moreover, Vaccibodies bind divalently to APC to promote a more efficient initiation of an immune response. Hence, a major purpose of the present invention is to induce a strong immune response to render adjuvants redundant. Vaccibodies comprise a dimer of a monomeric unit that consists of a targeting unit with specificity for a surface molecule on APC, connected through a hinge region and a Cγ3 domain to a antigenic unit in the COOH-terminal end; the latter being of e.g. B cell lymphoma or myeloma origin (Fig 1), although any origin is possible due to the cassette cloning system in the expression vector. The said molecule is capable of inducing an immune response against multiple myeloma, but extension to a general vaccination strategy for any polypeptid should be feasible. The present invention also relates to a DNA sequence coding for this recombinant antibody based molecule, to expression vectors comprising these DNA sequences, cell lines comprising said expression vectors, to treatment of mammals preferentially by immunization by means of Vaccibody DNA, Vaccibody RNA, or Vaccibody protein, and finally to pharmaceuticals and a kit comprising the said molecules.
DESCRIPTION OF THE FIGURES
Fig 1 The structure of the Vaccibody. The two scFvs in white target the Vaccibody to surface molecules on APC. They may be replaced by other targeting molecules, e.g. chemokine receptors. The hinge provides flexibility of the relative orientation of the two NH2-terminal scFvs and disulfide bridges the monomers. The CH3 domains (light grey) act as a spacer between the NH -and COOH terminal scFvs and participate in the dimerization through hydrophobic interactions. These dimerization motifs may be replaced by other dimerization or multimerization domains. The two scFvs shown in dark grey are the antigenic moiety of the Vaccibody. These scFvs are derived from the M component, thus harboring idiotypic sequences (black). The antigenic scFv may be replaced by any polypeptide derived from an antigenic source, conferring vaccine strategies towards any antigen.
Fig 2 Principle. The Vaccibody is targeted to surface molecules on APC, the complex is taken up by receptor-mediated endocytosis, processed and Id-peptides are presented to CD4+ T cells on MHC class II. Simultaneously, the Id may stimulate B cells with an anti-Id BCR. These B cells will also serve as APC for the CD4+T cells. Thus, T and B cells will cooperate to enhance the response, as indicated. Anti-Id B cells will as a consequence develop into plasma cells that produce anti-Id antibodies.
Fig 3 The MHC class Il-specific Vaccibodies bind CD19+ splenocytes (white). The NTP-specific Vaccibodies (negative controls - grey), do not bind to the splenocytes. Binding was detected with streptavidinPE and 9A8bio which is a rat mAb that binds the antigenic Fv of M315 origin (Vλl/2).
Fig 4 The NIP-specific control Naccibodies exhibits binding to the hapten NIP. Supernatant from cells transfected with genes encoding NIP-specific Naccibodies was added to ELISA plates coated with ΝIP-BSA. 9A8 bio (antiNλl/2) was used as the secondary Ab. Similar results were obtained with a biotinylated anti IdAb (Ab2.1-4 bio). Fig 5 The Vaccibodies exhibit binding to DNP, hence the antigenic scFv is correctly folded. The M315 Ab, from which the antigenic scFv is derived, is specific for the hapten DNP. ELISA plates were coated with DNP-BSA. Supernatants from cells transfected with genes encoding various Vaccibodies were added. Binding was detected with 9A8 bio.
Fig 6 APC pulsed with titrated amounts of MHC class II specific Vaccibodies stimulated polarized M315-specific T cells from TCR-transgenic SCTD mice >100-1000 fold better than the NIP-specific, untargeted control Vaccibodies. There were no significant differences between the Vaccibodies with a long sequence prior to the first disulfide bridge in the hinge (hl+h4) compared to the Vaccibodies with a short hinge sequence above the first disulfide bridge (h4).
Fig 7 The MHC class II-specific Vaccibodies induce a strong anti-Id Ab response in the absence of adjuvant. BALB/c mice were injected with Vaccibodies, 20μg and 200μg, respectively. Blood samples were taken on different time points for sera analysis. Shown is data from sera taken on day 28, 14 days after the second immunization of Vaccibodies. The MHC class II-specific Vaccibodies induced a strong anti-Id Ab response. The Vaccibodies with the longest hinge (hl+h4) induced the strongest anti-Id Ab response reaching 3-4μg/ml in sera.
Fig 8 Construction of the two hinge-Cγ3 variants derived from hIgG3 by PCR. The templates were from pUC19 containing modified hIgG3 constant regions were the h4 exon were connected to the CH3 domain (A) or the hi exon were connected to the h4 exon further connected to the CH3 domain (B) (Olafsen T et al, 1998). The primers inserted Hindlll (5') and Sfil (3') restriction enzyme sites. The hinge and CH3 domain are connected by a triplicate of the amino acids GlyGlyGlySerSer.
Fig 9 Construction of the hinge-Cγ3 segments derived from mIgG2b. The hinge and the CH3 genes were amplified from a pUC18 vector containing the constant region of mIgG2b by PCR with two primers encoding a Hindlll (5') and a Sfil (3') restriction enzyme site. The two PCR fragments were joined by PCR SOEing. In this reaction, the hinge and CH3 domain were connected by a triplication of the aniino acids Gly-Gly-Gly- Ser-Ser.
Fig 10 Construction of the scFv derived from the myeloma protein M315. The cDNA that functioned as a template in the PCR reactions were derived from RNA extracted from MOPC315.4 cells. The V regions were joined by PCR SOEing resulting in a scFv. hi this reaction, the V regions were connected by a triplicate of GlyGlyGlyGlySer. Furthermore, the gene fragments encoding the complete scFv were flanked by Sfil and Sail restriction enzyme sites.
Fig 11 Joining of the hinge-Cγ3 segments and the M315 scFv by PCR SOEing. This reaction introduced the Sfil site 5' of the antigenic scFv encoding region.
Fig 12 Subcloning of the hinge-Cγ3-M315 scFv into pUC19. Three different dimerization motifs were included, derived from mIgG2b or IgG3. In all cases, they consisted of hinge followed by a triplicate of GlyGlyGlySerSer and CH3. Two different hinges were derived from gG3, one consisting of hi linked to h4, and one consisting of h4, only.
Fig 13 Removal of two inconvenient BamHI restriction enzyme sites within the gene fragment encoding the antigenic scFv by QuickChange PCR.
Fig 14 Introduction of stop codon, a Sfil and a BamHI restriction enzyme site downstream of the coding region by QuickChange PCR.
Fig 15 Subcloning into the C cassette of the expression vector pLNOH2 on Hindlll- BamHI
Fig 16 Cloning of the V regions specific for ND? and MHCII. The V regions were amplified and joined by PCR soeing resulting in scFvs. The linker connecting the V regions consists of a triplicate of GlyGlyGlyGlySer. The gene fragments encoding the complete scFvs are flanked by Bsml/Munl and BsiWI sites. Linkers and restriction sites were introduced in the PCR reactions.
Fig 17 Subcloning into the expression vector pLNOH2 on Bsml/Munl and BsiWI
Fig 18 The final Vaccibody construct
Fig 19 Detailed figure of Vaccibody gene construct. The targeting unit is inserted between the Bsml/Mfel and BsiWI restriction enzyme sites (The V cassette of the pLNOH2 vector). The hinge-Cγ3-Fv315 is inserted between the Hindlll and BamHI sites into the C cassette of pLNOH2. The hinge and the Cγ3 domain as well as the two scFv's are connected with (G4S)3 linkers (black boxes). The Cγ3 and the Fv315 are connected through a GLSGL linker. The Fv315 is inserted between two nonidentical Sfil restriction enzyme sites. The antigenic unit and dimerization motif may be of any origin appropriate. Also, functional fragments of Cγ3 may be employed, or a sequence which is substantially homologous to the Cγ3 sequence or Cγ3 fragments.
Fig 20 Vaccibodies are secreted as functional molecules. Two distinct Vaccibodies were tested, one carrying the hapten specific FvNIP as targeting unit, while the other carried the MHC class II-specific FvI_E as targeting unit. Both carried the scFv from M315 (Fv3 5) as antigenic unit, a) 10% SDS-PAGE of metabolically labeled Vaccibodies immunoprecipitated from culture supernatants of transfectants with or without reduction of disulfide bonds by mercaptoethanol (ME), b) DNP-specificity of the Vaccibodies was measured by ELISA. Supernatants from NSO cells transfected with Vaccibodies were added to ELISA plates coated with DNP-BSA. Data are illustrated as mean of triplicates and error bars indicate SEM. c) NTP-specificity was measured by ELISA. ELISA plates were coated with NIP-BSA. Vaccibodies in both b) and c) were detected by either 9A8- bio (αVλl/2) or Ab2.1-4 (specific for Id of Fv315) Fv315 carries Vλ2 and FvNπ> carries Vλl and will both bind 9A8 mAb. Only Fv315 will bind Ab2.1-4 mAb. FvI_E carries VK and will bind neither of the mAbs. Fig 21 Production of Vaccibodies by intramucular injection of naked DNA plasmids followed by in vivo electroporation. Serum samples were collected on day 14. Vaccibody plasmids were injected into I-Ed positive BALB/c mice, which were subsequently electroporated. a) Level of Vaccibodies in sera was measured by ELISA, with DNP-BSA and 9A8bio as described previously, b) The same day 14 sera samples were analyzed for anti-Id antibodies by ELISA. Microtiter plates were coated with M315 and 187-bio (anti- mouse K Ab) was used for detection, c) Comparison of detectable Vaccibody levels and anti-Id antibodies. The amount of detectable Vaccibodies in sera is shown on the y-axis and the level of αld-Abs is shown on the x-axis.
Fig 22 Tumor avoidance. BALB/c mice were immunized once with naked plasmids encoding MHC class II specific Vaccibodies (FvI_EFv315), nontargeted NIP-specific Vaccibodies (FvNIP Fv315) or 0.9% NaCl by i.m. immunization into the two quadriceps muscles (25μg/muscle) followed by in vivo electroporation. They were challenged with l.όxlO5 MOPC315.4 myeloma cells s.c. and the first day tumor take were recorded. A tumor of >3 mm was scored as positive tumor take.
Fig 23 Induction of protective immunity against the MOPC315.4 plasmacytoma.
BALB/c mice were immunized once with naked plasmids encoding MHC class II specific Vaccibodies (Fv " Fv ), nontargeted NIP-specific Vaccibodies (Fv Fv ) or 0.9% NaCl by i.m. immunization into the two quadriceps muscles (25μg/muscle) followed by in vivo electroporation. They were challenged with 1.6x10s MOPC315.4 myeloma cells s.c. and their survival were compared.
Fig 24 Level of M315 myeloma protein in sera of mice on a) day 18 and b) day 24 after MOPC315.4 challenge in BALB/c mice vaccinated i.m. with Vaccibody plasmids followed by in vivo electroporation. M315 in sera samples were measured by ELISA coated with anti-Id-mAb (Ab2.1-4) and detected by biotinylated anti-IgA mAb (8D2).
Fig 25 Chemokine Vaccibodies are secreted as functional molecules. MJJP-lα Fv315 has mouse macrophage inflammatory protein 1 α as the targeting unit and scFv from M315 (Fv315) as the antigenic unit. Functionality of MJP-lα in Vaccibody format was measured in ELISA. Supernatants from 293E cells transfected with Vaccibodies were added to ELISA plates coated with anti-mouse MlP-lα mAb (R&D Systems) and detected by 9A8-bio (αVλl/2).
DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to a recombinant human antibody-based molecule, called Vaccibodies, comprising dimers of a monomeric unit that consist of a single chain fragment variable (scFv) of immunoglobulins (Ig) with specificity for surface molecules on Ag presenting cells (APC), connected through a hinge region and a Cγ3 domain to a scFv in the COOH-terminal end, the latter being derived from a myeloma protein (Fig 1), although any origin is possible due to the cassette cloning system of the expression vector. The hinge region and the Cγ3 domains (carboxyterminal C domain of Ig) contribute to dimerization of the Vaccibody through disulfide bridges in the hinge and strong hydrophobic interactions between the two Cγ3 domains. Hence, the dimeric product will include two preferably identical scFvs with binding specificity for the same surface molecules on APC (Fig 2), enabling bivalent binding. Bivalent binding (i.e. crosslinking) is of importance to trigger activation of the target cell and thereby initiation of an immune response. Also, bivalent binding provides increased binding strength due to avidity effects, and increases the likelihood of receptor mediated endocytosis into the APC and subsequent degradation inside the APC. Furthermore, the bivalent binding may provide important receptor mediated intracellular signaling to the APC. The scFvs with a targeting function are either derived from B cell liybridomas expressing monoclonal antibodies (mAbs) that bind to surface molecules on APC, or they may be derived from any source, e.g. phage display libraries. The use of scFvs from B cell hybridomas as the targeting moiety opens for a great range of possible targets due to the large collection of B cell hybridomas that produce mAbs which bind different surface molecules on APC. Furthermore, one may choose the nature of the signal given to the targeted cell by employing agonistic or antagonistic mAbs. Growing knowledge of Ab-Ag interactions will allow the improvement of the binding affinity of such mAbs to their Ag by amino acid replacements in the binding sites. This can be performed by ordinary site-directed mutagenesis. For vaccine purposes, an attractive approach is to target the Vaccibodies to surface molecules expressed exclusively on subsets of dendritic cells (DC) that are able to initiate a strong, specific immune response towards the patients own Id. Examples of such target surface molecules on APC are HLA, e.g. HLA-DP, CD14, CD40, Toll-like receptors, e.g. Toll-like receptor 2, or chemokine receptors. The Vaccibody can be targeted to said surface molecules by means of targeting units comprising for example antibody binding regions with specificity for e.g HLA, HLA-DP, CD14, CD40, or Tolllike receptor; ligands, e.g. soluble CD40 ligand; natural ligands like chemokines, e.g. RANTES or MTP-lα; or bacterial antigens like e.g. flagellin or a part thereof. Because the targeting unit is inserted into the V cassette of the expression vector pLNOH2 (Norderhaug, Olafsen et al. 1997), it is easily exchanged with other units (Fig 17).
The crucial dimerization motifs in the Vaccibodies constructed in the examples so far, include hinge regions and an immunoglobulin domain (e.g. Cγ3 domain), e.g. carboxyterminal C domain (CH3 domain), or a sequence that is substantially homologous to said C domain. The hinge is Ig derived and contributes to the dimerization through the formation of an interchain covalent bond(s), e.g. disulfide bridge(s). In addition, it functions as a flexible spacer between the domains allowing the two scFvs with targeting tasks to bind simultaneously to two target molecules expressed with variable distances (Fig 2). The immunoglobulin domains contribute to homodimerization through noncovalent interactions, e.g. hydrophobic interactions, h a preferred embodiment the Cjj3 domain is derived from IgG. These dimerization motifs can be exchanged with other multimerization moieties (e.g from other Ig isotypes/subclasses).
The C-terminal scFv derived from the monoclonal Ig produced by myeloma or lymphoma cells, also called the myeloma/lymphoma M component, can be genetically exchanged with other scFvs or any antigen because the vector has been constructed with a Sfi I restriction site (Fig 8). Therefore, the scFv derived from the model myeloma protein used in the present example is easily swapped with scFv from the M component of any patient with B cell lymphoma or multiple myeloma. Thus, this vector will allow for rapid construction of individual patient specific vaccines. The dimeric structure of the Vaccibody not only affords crosslinking, but should also allow double loading of the patient Fv to the APC per Vaccibody molecule compared to a single scFv combined to a single targeting moiety. Furthermore, there is in a single Vaccibody duplication of serological idiotypic determinants, which might be of importance for the anti-Id B cell response. The Vaccibodies lack a CH2 domain and hence all FcR binding sites, and should therefore exclusively be taken up through its target molecules, such as MHC class II in the example used and not by any FcRs, ensuring that a large proportion of the vaccine will arrive at the intended target cells. This is in contrast to vaccines that exert their effect through the binding to an FcR on a target cell (Ravetch and Bolland 2001).
Vaccibodies can be extended to a general medical treatment through induction of an immune response against any polypeptide of any origin. It is possible to replace the idiotypic scFv with other antigenic sequences of sufficient length to allow proper folding of the polypeptide. This sequence may be derived from other cancer proteins or infectious agents. Examples of such cancer sequences are telomerase, more specifically hTERT. The infectious agents can be of bacterial, e.g. tuberculosis antigens, or viral origin, more specifically HIV derived sequences like e.g. gpl20 derived sequences. Insertion of such sequences in a Vaccibody format might also lead to activation of both arms of the immune response, similar to the Vaccibodies that are described herein, which comprise the idiotypic scFv. Imiriunization by means of Vaccibody protein, Vaccibody DNA, or Vaccibody RNA, the latter two executed e.g. by intramuscular injection followed by electroporation (See Examples), are all feasible methods. The scFvs on the NH2- terminal end of the Vaccibodies target the Vaccibodies to
APC through binding to surface molecules (Fig 2), and in the example shown they bind to MHC class II. MHC class II is expressed on all professional APC, so the Vaccibodies described herein are able to target B cells, DC and macrophages. Targeting of conventional Ag-Ab complexes to MHC class II induces activation of specific CD4+ T cells (Snider and Segal 1987; Casten and Pierce 1988) . Targeting of Troybodies to MHC class II has previously been shown to enhance Ag presentation and T cell activation in vitro as well as in vivo (Lunde, Western et al. 2002). In the antigenic region of the molecule, the Vaccibodies of the examples contain the scFv of the myeloma protein M315 derived from the BALB/c plasmacytoma MOPC315.4. The λ2315 light chain of M315 harbors three defined somatic mutations in the CDR3 loop and functions as a model idiotypic T cell epitope in a well defined system (Bogen, Malissen et al. 1986; Bogen and Lambris 1989).
The Vaccibodies have been genetically assembled, and the DNA transfected into NSO cells, 293E cells and Cos-7 cells. Transfectants produce and secrete the recombinant Vaccibody molecules. The targeting scFvs at one end of the Vaccibodies exhibit binding to MHC class II (Fig 3). The antigenic scFv at the other end of the Vaccibody binds DNP (di-nitro-phenyl, the specificity of M315) (Fig 5). Thus, both the targeting and antigenic scFvs retain the same folding pattern as in their original context. We have evidence that Vaccibodies have the ability to induce strong T cell responses through their binding to APC and presentation of Id-peptides on class II molecules to Id- specific CD4+ T cells in vitro (Fig 6). Furthermore, since they have intact Fv of the M- component, they elicit anti-Id antibodies in significant amounts in vivo when injected into BALB/c mice without adjuvants (Fig 7).
To determine if MM patients treated with ASCT achieve remission with a low serum myeloma protein concentration, ELISA should performed for each patient's myeloma protein because routine assays (agarose gel electrophoresis, combined with immunofixation) have only a sensitivity of about 0.2-1 mg/ml, which is far too insensitive. The kinetic data of the serum myeloma protein levels will indicate if and when Id-vaccination may best be performed post ASCT to avoid the problem of T cell tolerance of newly educated thymic emigrants. To achieve this, mice are immunized with DNA encoding patient specific Vaccibodies by in vivo electroporation of muscle cells. Sera from immunized mice are absorbed on anti human Ig-Sepharose to remove crossreactive antibodies and thereafter eluted to obtain purified highly Id-specific antibodies. Sandwich ELISAs specific for each patient's myeloma are performed as follows: The purified anti-Id Ab from mice is coated in wells. Serum from the patient in casu is added. Myeloma protein binding to anti-Id antibodies will be detected by use of Ab specific for human IgG or IgA. The sensitivity of such sandwich ELISAs is usually < 1 ng/ml, which is > 106 times more sensitive than routine assays. Furthermore, to monitor development of new T cells, profile of T cells in blood will be monitored by flow cytometry with Vβ-specific mAbs, in combination with other markers.
The present invention relates to a pharmaceutical comprising the above described recombinant based antibody, DNA/RNA sequences, or expression vectors according to the invention. Where appropriate, this pharmaceutical additionally comprises a pharmaceutically compatible carrier. Suitable carriers and the formulation of such pharmaceuticals are known to a person skilled in the art. Suitable carriers are e.g phosphate-buffered common salt solutions, water, emulsions, e.g. oil/water emulsions, wetting agents, sterile solutions etc. The pharmaceuticals may be administered orally or parenterally. The methods of parenteral administration comprise the topical, intra- arterial, intramuscular, subcutaneous, intramedullary, intrathekal, intraventricular, intravenous, intraperitoneal or intranasal administration. The suitable dose is determined by the attending physician and depends on different factors, e.g. the patient's age, sex and weight, the kind of administration etc. Furthermore, the present invention relates to a vaccine composition against cancer or infectious diseases comprising an immunologically effective amount of the nucleic acid encoding the molecule of the invention or degenerate variants thereof, wherein said composition is able to trigger both a T-cell- and B-cell immuiie response. The present invention also relates to a kit comprising Vaccibody DNA, RNA, or protein for diagnostic, medical or scientific purposes. The invention further relates to a method of preparing the recombinant molecule of the invention comprising, transfecting the vector comprising the molecule of the invention into a cell population; culturing the cell population; collecting recombinant protein expressed from the cell population; and purifying the expressed protein.
The above described nucleotide sequences may preferably be inserted into a vector suited for gene therapy, e.g. under the control of a specific promoter, and introduced into the cells. In a preferred embodiment the vector comprising said DNA sequence is a virus, e.g an adenovirus, vaccinia virus or an adeno-associated virus.
Retroviruses are particularly preferred. Examples of suitable retroviruses are e.g.
MoMuLV or HaMuSV. For the purpose of gene therapy, the DNA/RNA sequences according to the invention can also be transported to the target cells in the form of colloidal dispersions. They comprise e.g. liposomes or lipoplexes.
In a preferred embodiment of the present invention naked Vaccibody DNA construct is injected intra-muscularly into mice, whereupon the site of injection is subject to in vivo electroporation. This DNA vaccination resulted in production of Vaccibody protein which conferred life-saving protective immunity on a majority of the mice. More specifically, example 4 demonstrates that after a single injection of Vaccibody plasmid in quadriceps and electroporation, FV NIP Fv 315 Vaccibody protein was detected in significant amounts in serum, while there was no detectable FV I_E Fv 315 (Fig 21a). Furthermore, example 5 shows the presence of anti-Id antibodies in serum after injection of Vaccibody DNA intramuscularly and electroporation. Taken together the results demonstrate a formidable effect of targeting to MHC class II (I-Ed) positive cells for development of a strong humoral response. Further, example 6 shows that protection against myeloma can be achieved by i.m. DNA vaccine followed by electroporation and that the protection requires targeting of the tumor-derived scFv to APC. Example 7 demonstrates that chemokines are functional as targeting units in the Vaccibody format.
MATERIALS AND METHODS
Mice BALB/cABom were from Bomholtgaard (Ry, Denmark). The λ2315-specific TCR- transgenic mice on a BALB/c background (Bogen, Gleditsch et al. 1992)were bred in our animal facility.
Cell lines The 14-4-4S Hybridoma (Ozato, Mayer et al. 1980) and NSO cells were purchased from American Type Culture Collection (ATCC, Manassas, VA). 293E cells, a variant of the 293 cell line expressing the Epstein-Barr virus EBNA1 protein.
Construction of Vaccibodies The gene for the hIgG3 hinge and CH3 domain was cloned from the pUC19 vector containing hinge genetically combined with Cγ3 genes of the hIgG3 subclass (Olafsen, Rasmussen et al. 1998). Two variants of the hinge length in the humanized Vaccibodies were made; one with just the h4 exon connected to the CH3 domain (sh) and one with both exon hi and h4 connected to the CH3 domain (Ih) (Fig 8). The primers included restriction enzyme sites (underlined): 5'h4: tag caa get tgg cca gcg cag gga g; 35CH3: cag gee acc Rag gee ttt acc egg aga cag gga. The hi exon were introduced directly upstream of the h4 exon by QuickChange PCR using these primers Qhla: ctccaatcttctctctgca gag etc aaa acc cca ctt ggt gac aca act cac aca gag ccc aaa tct tgt gac ac and Qhlb: gt gtc aca aga ttt ggg etc tgt gtg agt tgt gtc acc aag tgg ggt ttt gag etc tgcagagagaagattgggag.
The murine Vaccibodies have a complete hinge and CR3 domain of the mIgG2b subclass picked up by PCR from a pUC18 vector containing the Cγ2b genes (Fig 9). The primers included restriction enzyme sites (underlined) or linkers (bold) with the complementary sequences (italic): 5 'hinge: tagcaagctt ca gag ccc age ggg ccc; 3 'hinge: 5' tec acc tec get get tec acc gcc tgg gca ttt gtg aca etc ctt g; 5'CH3 : gga age age gga ggt gga agt gga ggg eta gtc aga get cca ca; 3'CH3 : cag gcc acc gag gcc acc egg aga ccg gga gat g. The hinge and the CH3 domain were then joined by PCR SOEing. The Antigenic V region genes were cloned from the plasmacytoma MOPC315.4 (Eisen, Simms et al. 1968). The V regions were obtained by extracting mRNA from the MOPC315.4 cell line with oligo (dT)-coated magnetic Dynabeads (Dynal). First strand cDNA were then made and used as template for PCR amplification of the V region genes using specific primers annealing to the exact ends of the M315 V region sequences. The primers included restriction enzyme sites (underlined) or linkers (bold) with the complementary sequences (italic). The primer sequences were: 5'VH: ggc etc ggt ggc ctg gat gta cag ctt cag gag tea; 3'VH: gcc aga gcc acc tec gcc iga tec gcc tec acc tga gga gac tgt gag agt ggt; 5'VL: ggc ggα ggt ggc tctggc ggt ggc gga teg cag get gtt gtg act cag gaa; 3'VL: gacg tcgac tag gac agt gac ctt ggt tec. The VH and VL genes were then joined by PCR soeing to a scFv format (Fig 10).
The complementary sequences in the tags 3' of the Cγ3 coding region and 5' of the M315 VH coding region enabled the M315 scFv to be combined with the three different hinge-CπS genes by PCR SOEing (Fig 11). The products of this reaction were then digested with Hindlll and Sail and subcloned into a pUC19 vector (Fig 12). Two BamHI restriction enzyme sites inside the V regions of M315 were removed by QuickChange PCR (Fig 13) using primers: BamHI VLl:at gcc aac tgg ata caa gaa aaa cc; BamHI VL2 gg ttt ttc ttg tat cca gtt ggc at, BamHI VH1 : tgg aac tgg ata egg cag ttt cc and BamHI H2 gg aaa ctg ccg tat cca gtt cca. A following QuickChange PCR using primers: 3'VL stopl gtc act gtc eta tga ggcctgcagggcc ggatcc gtcgactctag and 3'VL stop2: eta gag teg ac ggatec ggccctgcaggcc tea tag gac agt gac, were then performed to introduce a stop codon (bold), a Sfil and a BamHI restriction enzyme site (underlined) downstream of the coding region (Fig 14). The final construct is then digested with Hindlll and BamHI and subcloned into the expression vector pLNOH2 (Fig 15) (Norderhaug, Olafsen et al. 1997).
The V region genes providing specificity for MHC class II had previously been cloned from the 14-4-4S hybridoma (Lunde, Western et al. 2002), which produces an Ab specific for the Eα chain (determinant la.7) of the I-E MHC class II molecule (Ozato, Mayer et al. 1980). Specific primers annealing to the exact ends of the V region sequences with tags designed to include restriction enzyme sites (underlined) or linker sequences (bold) with the complementary sequences (italic). The primer sequences were: 5'VL: gac att caattg aca cag tct tct cct get tec; 3 'VL: gcc aga gcc acc tec gcc aga tec gcc tec acc ttt gat ttc cag ctt ggt gcc; 5' VH: ggc gga ggt ggc tct ggc ggt ggc gga teg cag gtc cag ctg cag cag t; 3'VH: ga cgtacg actcacc tga gga gac ggt gac tga gg. The V region genes conferring specificity for the hapten NIP (Neuberger 1983) were designed with the similar tag sequences except for the 5'VL primer: 5'VL: ggtg tgcattcc cag get gtt gtg act cag gaa; 3'VL: gcc aga gcc acc tec gcc aga tec gcc tec acc tag gac agt cag ttt ggt acc t; 5'VH: ggc gga ggt ggc tct ggc ggt ggc gga teg cag gtc caa ctg cag cag cc; 3'VH: ga cgtacg a etc acc tga gga gac tgt gag agt ggt. Both NL and NH were then joined by PCR SOEing (Fig 16) and subcloned into the N cassette pLΝOH2 vector containing the hinge-CH3-scFvM315 genes (Fig 17 and Fig 18). Likewise, other N genes conferring a desired specificity are isolated from hybridomas or from phage selected from phage display libraries. They are then PCR amplified using primers designed in the same manner as above and subcloned after PCR SOEing in the targeting-cassette (Fig. 17 and 18). Rearranged NH and VK genes conferring specificity for HLA-DP were PCR amplified from cDΝA from the 22C1 hybridoma, which produces an antibody with pan HLA-DP specificity. The V genes were reamplified with new primers containing sites for direct cloning into the expression vectors pLΝOK and pLΝOH2 (Norderhaug and Olafsen, 1997); 55-VL, ggt tgeatteegαcαttgtgctc ce; 3 '-VL, cgtacgttctacteaegttttαtttecαget,- 55-VH, gtgcattccgα t cαgctgcΩ gαgtct; 3'-VH, cgtacgaetcacctgαggαgαccgtαgc. Furthermore, scFV was generated by PCR SOEing using the following primers: 5'VL, g gtg tgcattc cga cat tgt get cac c 3'VL: gcc aga gcc acc tec gcc aga tec gcc tec acc gtt tta ttt cca get 5 ' VH: ggc gga ggt ggc tct ggc ggt ggc gga teg gag gtg cag ctg cag gag tct 3'VH, cgtacg act cac ctg agg aga ccg tag c
In 3'NL and 5'NH the sequences in bold+italics are complementary and antiparallell, thus hybridising to generate the gene fragment encoding the linker region. Anti CD 14 N regions are cloned from the mouse hybridoma 3C10 (ATCC). The chemokine genes were cloned from thioglycolate stimulated peritoneal macrophages. 4ml 2% thioglycolate were injected i.p. into Balb/c mice. 3 days later peritoneal macrophages were collected and mRΝA was extracted with oligo (dT)-coated magnetic Dynabeads. First strand cDNA was made and used as template for PCR amplification of chemokine genes (RANTES and MlP-lα) using specific primers: 5'MIP-lα: ggtg tgcattc cgc gcc ata tgg age tga cac, 3'MTP-lα: ga cgtacg act cac ctg cat tea gtt cca ggt cag tg 5 'RANTES: ggtg tgcattc c gcc tea cca tat ggc teg g 3 'RANTES: ga cgtacg a etc acc tga cat etc caa ata gtt gat gta ttc. The different targeting unit genes were then digested with Muni and BsiWI or Bsml and BsiWI, respectively and subcloned into the V cassette pLNOH vector containing the hinge-CH3-scFvM315 genes (Fig 17 and Fig 18). CD40 ligand is cloned from T cells that are activated with LPS for 4 hours before mRNA is extracted for preparation of cDNA. The cDNA is used as template in a PCR reaction with primers specific for the CD40 ligand sequence. Furthermore, this sequence is reamplified with primers designed to facilitate subcloning in the targeting cassette as described above.
Production and purification of Vaccibodies The pLNOH2 vector carrying the Vaccibody genes was transfected into NSO cells by electroporation, and supernatants from single colonies resistant to 800μg/ml G418 were analysed for Vaccibody secretion after 2-3 weeks, using ELISA. DNP-BSA was used as coat, and biotinylated rat-anti mouse Vλl/2 (9A8-bio) was used for detection. The NIP- specific Vaccibodies were additionally screened in an ELISA using NIP-BSA as coat. The cells selected for high Vaccibody production were grown in Rollerbottles (VWR) and affinity purified from supernatant using a column made by DNP-lysine (Sigma) coupled to fast flow Sepharose. The Vaccibodies were eluted with 0.05M DNP-glycine (Sigma) and the flow-through was run on an ion-exchange Cl" Dowex 1x8 resin column (Sigma). The eluted Vaccibodies were dialyzed against PBS/0.05%NaN3 and sterile PBS, before the vaccibody concentration were calculated from absorbance values at 280nm.
Ab and flow cytometry
Ab and reagents used for flow cytometry were 9A8 biotin, FGK.45 biotin, streptavidin PerCP, anti-CD 19 PE and anti-mIgG2a PE (BD Pharmingen). BALB/c spleen cells were double stained with anti-CD 19 PE and Vaccibodies. Bound Vaccibodies were detected by 9A8-bio and streptavidin PerCP. Twenty thousand cells were run on FACSCalibur (BD Biosciences, Mountain View, CA) and analyzed using the WinMDI software.
Metabolic labelling and immunoprecipitation 2xl06 cells were labelled for 6h at 37°C in RPMI lacking methionine, cysteine (BioWhittaker) containing lOOμCi 35[S]-methionine, cysteine (Amersham). The SN was harvested and immunoprecipitated with rat anti-mouse Vλl/2 (9A8) on a wheel ON at 4°C. lOμl Dynabeads coated with sheep anti-rat IgG (Dynal AS, Oslo, Norway) were incubated with the precipitate for lh on a wheel and the Dynabeads were collected with a Dynal Magnetic Particle Concentrator rack (Dynal MPC). The beads were washed three times in ice cold PBS with 1%NP40 and resuspended in lOμl lx sample buffer. The proteins were eluted from the beads by incubating the samples at 95°C for 3 minutes. The Vaccibodies were run on a 10% SDS-PAGE gel, with a 5% stacking gel, at 40mA for lh, using a BIO RAD miniprotean II gel electrophoresis apparatus. The gels were subsequently fixed in 30% methanol and 10% acetic acid for 30 minutes prior to 30 min incubation with Amplify (Amersham), before drying and exposing to BIOMAX-MR film (Eastman Kodak Company, Utah, USA).
T cell proliferation assay Irradiated (2000rad) BALB/c splenocytes (5xl05cells/well) were used as a source of APC. Titrated amounts of different MHC CLASS II- and NIP-specific Vaccibodies were added to the splenocytes. A 91-107 λ2 synthetic peptide were used as a positive control. The assays were put up in 150μl cultures in 96-well flat-bottom microtiter wells and incubated for 4 h at 37°C (Lunde, Western et al. 2002). The cultures were then washed three times before addition of 200μl polarized λ2315-specific Th2 cells (2xl04) derived from TCR transgenic SCID mice. After 48h, the cultures were pulsed for 16-24h with lμCi 3[H] dThd. The cultures were harvested and, and incorporated 3[H] dThd was measured using a TopCount NXT scintillation counter (Packard, Meriden, CT).
In vivo experiments
BALB/c mice were injected subcutaneously (s.c) with 200μg or 20μg purified Vaccibody proteins in PBS on day 0, 14 and 28. Blood samples were taken on day 14 and 28 before revaccination and then on day 35, 42 and 49, before they were sacrificed according to the
Humane End Point procedure.
Measurement of antibody responses Anti-idiotypic Abs against M315 were measured by ELISA. The wells were coated with 2μg/ml M315. Anti-Id Ab in the sera were detected by a biotinylated anti-mouse VK Ab (187.1 bio), anti-mouse IgGl bio or anti-mouse IgG2a bio (both from BD Pharmingen). Ab2.1-4 (an anti-Id mAb that bind λ2315) was used as standard.
Vaccination
Protein vaccination: BALB/c mice were injected subcutaneously (s.c) in the right flank region with 20μg or 200μg purified class II- or NIP specific Vaccibodies (FV I_E Fv 315, FV NIP Fv 315 ) in PBS on day 0 and 14. PBS was injected as negative control. Blood samples were collected from the leg vein on different time points after the last immunization. Anti-idiotypic antibodies with specificity with specificity for Fv 315 were measured by ELISA. The wells were coated with 2μg/ml M315. Anti-Id Ab in the sera were detected by a biotinylated anti-mouse K mAb (187.1 bio). Ab2.1-4 (an anti-Id mAb binding Fv 315 (Kristoffersen, Hannestad et al. 1987) was used as standard.
DNA vaccination and electroporation
Five to ten weeks old Balb/c mice were purchased from Bomholtgaard (Ry, Denmark). The animals were anaesthetized by intraperitonal injection with 9 g Pentobarbital/mice and the legs were shaved. Conductive gel was applied at the skin and 50 μl vector DNA diluted in 0.9% NaCl, was injected into the quadriceps. Following injection, electroporation was performed, by applying rod electrodes to the skin near the site of the injection and subjecting the site to an electrical potential comprising 10 trains of 1000 pulses each, with a pulse length at two times 200 Sec (positive 200Sec and negative 200 Sec) with 600 s interval between each pulse and with a current limit of 50 mA (about 150-174 V/cm) (Tollefsen, Tjelle et al. 2002). Blood samples were collected from the leg vein on different time points and heart puncture was performed on the day they were sacrificed. Serum samples were analyzed for the presence of correctly folded Vaccibodies. The ELISA was performed with DNP-BSA as coat and 9A8-bio as detected Ab, as described above. In addition, serum samples were analyzed for anti-Id Abs by ELISA as described above.
Tumor challenge Protein Vaccibodies-MOPC315.4: BALB/c mice (6-10 weeks old) were injected s.c. with 160 μg class Il-or NIP-specific Vaccibodies in PBS in the right flank region on day 0 and 14. On day 28, 1.6xl05 MOPC315.4 cells were injected s.c. on the right flank. Mice were inspected twice weekly. Tumor size development was monitored by palpation and use of a caliper. A tumor of 3 mm in diameter was scored as tumor take. Mice were killed when tumor size reached 20 mm with no sign of tumor necrosis.
DNA Vaccibodies-MOPC315.4: DNA vaccination was performed at day 0 as described above. On day 14, 1.6xl05 MOPC315.4 cells were injected s.c. in the right flank region. Tumor size development was monitored by palpation and use of a caliper. The mice were sacrificed when the tumor size reached 20 mm. Blood samples were collected on different time points from the leg vein. Levels of M315 myeloma protein in sera were quantified in a sandwich ELISA with Ab2.1-4 as coat by biotinylated anti-Cα (8D2) mAb as detection Ab. Tumor size, tumor take, survival curves and statistical analyses were calculated by use of Graph Pad Prism 3.0 software (San Diego, CA).
EXAMPLES
By way of example the following experiments demonstrate that Vaccibodies bind APC and are able to trigger both T cell and B cell immune response. Moreover, the following experiments show that Vaccibodies induce a strong immune response rendering adjuvants redundant. The experiments demonstrate that said molecule is capable of inducing an iirimune response against multiple myeloma and, further, the feasibility of treatment of mammals by immunization by means of Vaccibody DNA or Vaccibody protein. The experiments also demonstrate that another attractive approach is to target the Vaccibodies to surface molecules expressed exclusively on subsets of dendritic cells (DC), like e.g. chemokine receptors. The following examples are meant to illustrate how to make and use the invention. They are not intended to limit the scope of the invention in any manner or to any degree.
Example 1
Vaccibodies are produced and secreted as functional dimerized molecules and is itself bound by the anti-Vλl/2 antibody (9A8) (Bogen 1989) and the anti-idiotypic antibody Ab2.1-4 (Lauritzsen, Weiss et al. 1994).
The M315 mAb binds the hapten di-nitro-phenyl (DNP) (Eisen, Simms et al. 1968). Therefore, to verify that Vaccibodies were produced, secreted and correctly folded as functional molecules, the antigenic units of Vaccibodies were tested in ELISA for their capability to bind DNP, 9A8 and Ab2.1-4 mAbs. Fig 20b shows that both the NTP- specific and the MHCII-specific Vaccibodies, that both have scFv315, bind DNP, 9A8 and Ab2.1-4. This was the case with all Vaccibodies containing scFv315, both the ones with long human dimerization unit, short human dimerization unit and murine dimerization unit (Fig 5). We next tested the targeting units of the Vaccibodies. These were found to be correct; first, the NIP-specific Vaccibodies bound NTP-BSA in ELISA (Fig 4 and Fig 20c), while the MHCII-specific Vaccibodies did not (Fig 20c). Second, the MHCII-specific Vaccibodies bound to I-E expressing BALB/c splenic B cells (H-2d) as detected by flow cytometry, whereas the NIP-specific Vaccibodies did not (Fig 3). To check for correct homodimerization, the Vaccibodies were metabolically labelled by growth of transfected cells in medium containing 35S-methionine, vaccibodies were immunoprecipitated from supernatant using specific antibodies, and analyzed by SDS-
PAGE. As would be expected from the theoretical consideration of Fig.2, both the FV NIP
Fv 315 and ¥v l'B Fv 315 transfectomas secreted dimeric Vaccibodies of ~ 130 kDa. After reduction of disulfide bonds, the Vaccibodies are degraded to monomeric chains of- 65 kDa (Fig 20a).
Example 2
MHC class II-specific Vaccibodies enhance λ2315 -specific stimulation ofCD4+ T cells. Class II-specific and non-targeting NIP-specific Vaccibodies were mixed with antigen presenting cells (APC) and compared for their ability to induce specific T cell activation. Irradiated BALB/c splenocytes were used as APC. The BALB/c strain has the H-2d haplotype, hence they express I-Ed molecules necessary for both targeting of the MHC II-specific Vaccibodies and presentation of the λ2315 epitope to specific CD4+ T cells.
The APC were pulsed with the different Vaccibodies for 4h and subsequently washed. Washing was performed to reduce the chance that I-E -specific Vaccibodies in the culture medium could diminish T cell stimulation by blocking I-Ed molecules (Lunde, Western et al. 2002). Polarized Th2 cells from mice transgenic for a λ2315- specific I-Ed restricted TCR (Lauritzsen, Weiss et al. 1993) were added as responder T cells. The dose response curve in fig 6 shows that the λ2 epitope was presented 100- 1000 times more efficiently to TCR-transgenic Th2 cells when they were carried in the APC-targeted MHC II-specific Vaccibodies (both the ones with short and long human dimerization units) compared to the non-targeted NIP-specific Vaccibodies. It should be noted that Vaccibodies do not include an FcγR binding site; hence the NIP-specific Vaccibodies should not be able to enter cells via receptor-mediated endocytosis. Example 3
Level of anti-idiotypic antibodies in sera of mice that received Vaccibodies as proteins in saline s.c. in the absence of adjuvant.
In the protein vaccination protocol, BALB/c mice were immunized twice, spaced two weeks apart, with 20 or 200μg MHC II- specific Vaccibodies or NIP-specific Vaccibodies in PBS. Note that no adjuvant was employed. Sera from immunized mice taken at various time points after the second vaccination were then analyzed for anti- idiotypic antibodies binding M315 in ELISA. The MHC II-specific Vaccibodies elicited significant higher anti-idiotypic antibody responses after 14 days after the second immunization than did NIP-specific Vaccibodies. Vaccibodies with a long human dimerization unit induced best anti-idiotypic Ab responses (Fig 7). Thus targeting of Vaccibodies enhanced anti-Id immuneresponses, however, by this route of immunization, also the non-targeted Vaccibodies induced some responses.
Example 4
Protein Vaccibodies detected in serum after injection of DNA intramuscularly and in vivo electroporation.
It has recently been described that skeletal muscle can produce antibodies after injection of Ig genes and electroporation (Tjelle 2004). We therefore investigated if functional
Vaccibodies were produced by i.m. plasmid injection and electroporation. Since the FV I_
E Fv 315 Vaccibodies are specific for I-Ed molecules present in BALB/c (H-2 ), these
Vaccibodies should be rapidly absorbed by the I-Ed positive cells in BALB/c. By contrast, the non-targeted FV NIP Fv 315 Vaccibodies should not be absorbed. Indeed, 14 days after a single injection of 50μg Vaccibody plasmid in quadriceps and electroporation, FV NIP Fv 315 Vaccibody protein was detected in significant amounts in serum, while there was no detectable
Figure imgf000028_0001
(Fig 21a). Example 5
Anti-Id antibodies in serum after injection of Vaccibody DNA intramuscularly and electropor'ation.
Analysis of the same day 14 sera samples for anti-idiotypic antibodies demonstrated that mice i.m. injected/electroporated with the MHC class II -targetd FV I_E Fv 315 Vaccibody DNA, had developed antibodies that bound idiotypic Fv from the MOPC315.4 tumor (Fig 21b). This result was in distinct contrast to the lack of any anti- idiotypic antibody response in mice injected with the non-targeted FV NIP Fv 315 Vaccibody DNA (Fig 21b). Taken together with the results described in example 4, the results demonstrate a formidable effect of targeting to MHC class II (I-Ed) positive cells for development of a strong humoral response. Control mice injected i.m. with 0.9% NaCl followed by electroporation had neither Vaccibodies nor anti-idiotypic Abs in day 14 sera (Fig 21a-c).
Example 6
Induction of protective immunity against the MOPC315.4 myeloma: Vaccibody DNA injection/electroporation.
Intramuscular vaccination with MHC class Il-targeted FV I_E Fv 315 Vaccibody plasmids and subsequent electroporation induced strong protection against a challenge with MOPC315.4 myeloma cells, pO.OOl, compared to control mice injected with 0.9% NaCl and electroporated (Fig 23). By contrast, non-targeted FV NIP Fv 315 Vaccibody plasmid immunization was ineffective compared to the saline control group, p= 0.2739 (Fig 23). The appearance of tumor was delayed in mice vaccinated with FV I_E Fv 315 compared to FV NIPFV 315 (Fig 22). One of the
Figure imgf000029_0001
Fv 315 vaccinated mice developed a tumor of maximum 6mm (day 20) in diameter that regressed and was completely undetectable from day 28 (data not shown). The presence of M315 myeloma protein in sera confirmed the tumor size measurements (Fig 24). These results show that protection against the MOPC315.4 myeloma can be achieved by i.m. DNA vaccine followed by electroporation and that the protection requires targeting of the tumor- derived scFv to MHC class II (I-Ed) positive cells. Example 7
Chemokines are functional as targeting units in the Vaccibody format
A vector carrying the gene encoding a Vaccibody as outlined in Figure 18, but specifically with MTP-la in the targeting unit, long human hinge in the dimerization unit and M315 scFv in the antigenic unit was assembled. Supernatant from cells transfected with this construct was collected and tested in ELISA for the presence of
Vaccibodies binding to an anti-mouse MTP-la mAb and 9A8 bio. The results showed that such Vaccibodies with desired specificity were produced, and furthermore, that they bound to anti-MTP-lα mAb, while the NTP-specific Vaccibodies did not (Fig 25).
Example 8
The chemokine RANTES is functional as targeting unit in the Vaccibody format hi the same manner, a vector with a gene encoding Vaccibodies like those described in example 7 was produced, with the exception that the targeting unit was the mouse chemokine RANTES. Supernatant from cells transfected with this construct was collected and tested in ELISA for the presence of Vaccibodies. The experiment showed that this Vaccibody variant was expressed and exported as a functional molecule.
Example 9 Flaggelin as targeting unit in the Vaccibody format
In the same manner, a vector with a gene encoding Vaccibodies like those described in example 7 was produced, with the exception that the targeting unit was flaggelin. Supernatant from cells transfected with this construct will be collected and tested in ELISA for the presence of Vaccibodies.
Example 10
Soluble CD40 ligand as targeting unit in the Vaccibody format
In the same manner, a vector with a gene encoding Vaccibodies like those described in example 7 was produced, with the exception that the targeting unit was soluble CD 40 ligand from the mouse. Supernatant from cells transfected with this construct will be collected and tested in ELISA for the presence of Vaccibodies. Example 11
Anti-Toll-like-receptor 2 as targeting unit in the Vaccibody format In the same manner, a vector with a gene encoding Vaccibodies like those described in example 7 was produced, with the exception that the targeting unit was a scFv with specificity for toll-like-receptor 2 from the mouse. Supernatant from cells transfected with this construct will be collected and tested in ELISA for the presence of Vaccibodies.
Example 12
Anti-CD14 is functional as targeting units in the Vaccibody format In the same manner, a vector with a gene encoding Vaccibodies like those described in example 7 was produced, with the exception that the targeting unit was scFv with specificity for human CD 14. Supernatant from cells transfected with this construct was collected and tested in ELISA for the presence of Vaccibodies. The results showed that this Vaccibody variant was expressed and exported as a functional molecule.
Example 13
Anti-HLA-DP is functional as targeting units in the Vaccibody format
In the same manner, a vector with a gene encoding Vaccibodies like those described in example 7 was produced, with the exception that the targeting unit was scFv with specificity for HLA-DP. Supernatant from cells transfected with this construct was collected and tested in ELISA for the presence of Vaccibodies. The results showed that this Vaccibody variant was expressed and exported as a functional molecule.
Example 14
Tuberculosis antigen in the Vaccibody antigenic cassette.
A nucleic acid encoding a tuberculosis antigen (cattle antigen) will be inserted into the antigenic unit of the Vaccibody construct.
Example 15
Telomerase antigen in the Vaccibody antigenic cassette hTERT, an antigenic region of the telomerase ribonucleoprotein, will be inserted into the antigenic unit of the Vaccibody construct.
Example 16
HIV Gpl20 antigenic in the Vaccibody antigenic cassette
A nucleic acid encoding a gpl20 derived molecule will be inserted into the antigenic unit of the Vaccibody construct.
Example 17
Vaccibodies with patient specific scFv of myeloma origin in the antigenic cassette.
Bone marrow aspirate from patients suffering from multiple myeloma is collected. The mononuclear cells (MNC) are separated using a density gradient solution of Ficoll-
Isopaque (LymphoprepTM from Axis-Shield PoC AS). Total RNA is isolated (TRIzol® Reagent from h vitrogenTM Life Technologies) from MNC, and cDNA is made from mRNA (First-Strand cDNA Synthesis Kit from Amersham Biosciences (Not I-d(T)18 bifunctional primer)). This cDNA is used as template in PCR with primers that amplify the V genes of the heavy or light chain of the multiple myeloma Ig. The sense primers are family specific and localized in the leader regions (VH1-7, VK1-6 and VLl-10), and the anti-sense primers are localized in the first part of the C regions (one primer each for IgG, IgA, kappa and lambda). PCR products are ligated into a vector (pGEM®-T Easy Vector from Promega), and transformed into E.colL DNA samples isolated from individual colonies are sequenced. Getting the same sequence from three different colonies originating from three different PCRs confirms that the V regions from the myeloma Ig have been isolated. PCR SOEing is performed and reamplification is done with primers including tags with sites for Sfil as described in Figure 19. For one patient such primers had the sequence: 5'TAVH 5' ACGTAGGCCTCGGTGGCCTGCAGATCACCTTGAAGGAGTCT
3'TAVK 5' GATCCGGCCCTGCAGGCCTCATTTGATCTCCAGCTTGGTCCC
The resulting vector is transiently transfected into 293E cells. Supernatants are tested in ELISA for the presence of such Vaccibodies. They are also injected into BALB/c mice. The presence of anti-idiotypic antibodies is measured in ELISAs against serum from the mice and serum from the patients.
REFERENCES
Bendandi, M., C. D. Gocke, et al. (1999). "Complete molecular remissions induced by patient-specific vaccination plus granulocyte-monocyte colony-stimulating factor against lymphoma." Nat Med 5(10): 1171-7. Biragyn, A., P. A. Ruffini, et al. (2002). "Toll-like receptor 4-dependent activation of dendritic cells by beta-defensin 2." Science 298(5595): 1025-9. Biragyn, A., K. Tani, et al. (1999). "Genetic fusion of chemokines to a self tumor antigen induces protective, T-cell dependent antitumor immunity." Nat Biotechnol 17(3):
253-8. Bogen, B. (1989). "Monoclonal antibodies specific for variable and constant domains of murine lambda chains." Scand J Immunol 29(3): 273-9. Bogen, B., Peripheral T cell tolerance as a tumor escape mechanism: deletion of CD4+ T cells specific for a monoclonal immunoglobulin idiotype secreted by a plasmacytoma. Eur J Immunol. 1996 Nov;26(l l):2671-9. Bogen, B., L. Gleditsch, et al. (1992). "Weak positive selection of transgenic T cell receptor-bearing thymocytes: importance of major histocompatibility complex class II, T cell receptor and CD4 surface molecule densities." Eur J Immunol 22(3): 703-9.
Bogen, B. and J. D. Lambris (1989). "Minimum length of an idiotypic peptide and a model for its binding to a major histocompatibility complex class II molecule." Embo J 8(7): 1947-52. Bogen, B., B. Malissen, et al. (1986). "Idiotope-specific T cell clones that recognize syngeneic immunoglobulin fragments in the context of class II molecules." Eur J
Immunol 16(11): 1373-8. Casten, L. A. and S. K. Pierce (1988). "Receptor-mediated B cell antigen processing. Increased antigenicity of a globular protein covalently coupled to antibodies specific for B cell surface structures." J Immunol 140(2): 404-10. Eisen, H. N, E. S. Simms, et al. (1968). "Mouse myeloma proteins with antihapten antibody acitivity. The protein produced by plasma cell tumor MOPC-315." Biochemistry 7(11): 4126-34. Hakim, I., S. Levy, et al. (1996). "A nine-amino acid peptide from IL-lbeta augments antitumor immune responses induced by protein and DNA vaccines." J hnmunol 157(12): 5503-11. Hough, D. W., R. P. Eady, et al. (1976). "Anti-idiotype sera raised against surface immunoglobulin of human neoplastic lymphocytes." J Exp Med 144(4): 960-9.
Huang, H. L, P. Y. Wu, et al. (2004). "Improved immunogenicity of a self tumor antigen by covalent linkage to CD40 ligand." Int J Cancer 108(5): 696-703. King, C. A., M. B. Spellerberg, et al. (1998). "DNA vaccines with single-chain Fv fused to fragment C of tetanus toxin induce protective immunity against lymphoma and myeloma." Nat Med 4(11): 1281-6.
Kristoffersen, G., K. Haimestad, et al. (1987). "Two M315 idiotopes defined by isologous monoclonal antibodies: one depends on germline and the other on mutated murine lambda 2 light chain sequences." Scand J Immunol 26(5): 535- 46. Lauritzsen, G. F., S. Weiss, et al. (1993). "Anti-tumour activity of idiotype-specific, MHC-restricted Thl and Th2 clones in vitro and in vivo." Scand J Immunol 37(1): 77-85. Lauritzsen, G. F., S. Weiss, et al. (1994). "Naive idiotype-specific CD4+ T cells and immunosurveillance of B-cell tumors." Proe Natl Acad Sci U S A 91(12): 5700-4. Lunde, E., L. A. Munthe, et al. (1999). "Antibodies engineered with IgD specificity efficiently deliver integrated T-cell epitopes for antigen presentation by B cells." Nat Biotechnol 17(7): 670-5. Lunde, E., I. B. Rasmussen, et al. (2001). "Troy-bodies': antibodies as vector proteins for T cell epitopes." Biomol Eng 18(3): 109-16. Lunde, E., K. H. Western, et al. (2002). "Efficient delivery of T cell epitopes to APC by use of MHC class II-specific Troybodies." J hmnunol 168(5): 2154-62. Neuberger, M. S. (1983). "Expression and regulation of immunoglobulin heavy chain gene transfected into lymphoid cells." Embo J 2(8): 1373-8. Norderhaug, L., T. Olafsen, et al. (1997). "Versatile vectors for transient and stable expression of recombinant antibody molecules in mammalian cells." J Immunol
Methods 204(1): 77-87. Olafsen, T., I. B. Rasmussen, et al. (1998). "IgM secretory tailpiece drives multimerisation of bivalent scFv fragments in eukaryotic cells." Immunotechnology 4(2): 141-53. Ozato, K., N. Mayer, et al. (1980). "Hybridoma cell lines secreting monoclonal antibodies to mouse H-2 and la antigens." J Immunol 124(2): 533-40.
Ravetch, J. V. and S. Bolland (2001). "IgG Fc receptors." Annu Rev Immunol 19: 275-
90. Sirisinha, S. and H. N. Eisen (1971). "Autoimmune-like antibodies to the ligand-binding sites of myeloma proteins." Proc Natl Acad Sci U S A 68(12): 3130-5. Snider, D. P. and D. M. Segal (1987). "Targeted antigen presentation using crosslmked antibody heteroaggregates." J Immunol 139(5): 1609-16. Tao, M. H. and R. Levy (1993). "Idiotype/granulocyte-macrophage colony-stimulating factor fusion protein as a vaccine for B-cell lymphoma." Nature 362(6422): 755- 8. Tjelle, T., Corthay, A., Lunde, E., Sandlie, L, Michaelsen, TE., Mathiesen, I and Bogen, B. (2004). "Monoclonal antibodies produced by muscle after plasmid injection and electroporation." J Mol Ther. Tollefsen, S., T. Tjelle, et al. (2002). "Improved cellular and humoral immune responses against Mycobacterium tuberculosis antigens after intramuscular DNA immunisation combined with muscle electroporation." Vaccine 20(27-28): 3370-
8.

Claims

P a t e n t C l a i m s:
1. A method of treating multiple myeloma or lymphoma in a patient, the method comprising administering to the patient, a recombinant antibody-based molecule comprising two targeting units and two antigenic units connected through a dimerization motif, or a nucleic acid encoding said recombinant antibody-based molecule
2. The method of claim 1, wherein administering the nucleic acid comprises delivering the nucleic acid by electroporation.
3. The method of claim 1, wherein said targeting unit(s) is/are a single chain fragment variable of Ig (scFv).
4. The method of claim 3, wherein said scFv is anti-HLA, anti-CD 14, anti-CD40, or anti-toll-like receptor.
5. The method of claim 4, wherein said anti-HLA is anti-HLA-DP.
6. The method of claim 4, wherein said anti-toll-like receptor is anti-toll-like receptor 2.
7. The method of claim 1, wherein said targeting unit(s) is/are a ligand.
8. The method of claim 7, wherein said ligand is soluble CD40 ligand or a chemokine.
9. The method of claim 7, wherein said ligand is a chemokine.
10. The method of claim 8 or 9, wherein said chemokine is RANTES or MlP-lα.
11. The method of claim 8 or 9, wherein said chemokine is MlP-lα.
12. The method of claim 1, wherein said targeting unit(s) is/are a bacterial antigen.
13. The method of claim 12, wherein the bacterial antigen is a flaggelin.
14. The method of claim 1, wherein the targeting units have the ability to target antigen presenting cells (APC).
15. The method of claim 1, wherein the targeting units have the ability to target HLA- DP, CD14, CD40, toll-like receptors, or a chemokine receptors.
16. The method of claim 15, wherein said HLA is HLA-DP
17. The method of claim 1, wherein the targeting units have the ability to target chemokine receptors.
18. The method of claim 1, wherein the antigenic unit(s) is/are an antigenic scFv.
19. The method of claim 18, wherein the antigenic scFv is derived from a monoclonal Ig produced by myeloma or lymphoma.
20. The method of claim 18, wherein the antigenic unit(s) is/are a telomerase, or a functional part thereof.
21. The method of claim 20, wherein said telomerase is hTERT.
22. The method of claim 1, wherein the antigenic unit(s) is/are derived from a bacterium.
23. The method of claim 22, wherein the bacterium derived antigenic unit(s) is/are a tuberculosis antigen.
24. The method of claim 1, wherein the antigenic unit(s) is/are derived from a virus.
25. The method of claim 24, wherein the virus derived antigenic unit(s) is/are derived from HIV.
26. The method of claim 25, wherein the HIV derived antigenic unit(s) is/are derived
Figure imgf000038_0001
27. The method of claim 1, wherein the dimerization motif comprises a hinge region and an immunoglobulin domain.
28. The method of claim 27, wherein the hinge region is Ig derived.
29. The method of claim 27, wherein the hinge region has the ability to form one or several covalent bonds.
30. The method of claim 29, wherein the covalent bond is a disulphide bridge.
31. The method of claim 27, wherein the immunoglobulin domain is a carboxyterminal C domain, or a sequence that is substantially homologous to said C domain.
32. The method of claim 31, wherein the carboxyterminal C domain is derived from IgG.
33. The method of claim 27, wherein the immunoglobulin domain has the ability to homodimerize.
34. The method of claim 33, wherein said immunoglobulin domain has the ability to homodimerize via noncovalent interactions.
35. The method of claim 34, wherein said noncovalent interactions are hydrophobic interactions.
36. The method of claim 1 comprising administering the nucleic acid to the patient to induce production of the recombinant antibody-based molecule.
37. The method of claim 1 comprising administering a vector comprising the nucleic acid.
38. A recombinant antibody-based molecule comprising two targeting units and two antigenic units connected through a dimerization motif, or a nucleic acid encoding said recombinant antibody-based molecule.
39. The recombinant molecule of claim 38, wherein said targeting unit(s) is/are a single chain fragment variable of Ig (scFv).
40. The recombinant molecule of claim 39, wherein said scFv is anti-HLA, anti-CD14, anti-CD40, or anti-toll-like receptor.
41. The recombinant molecule of claim 40, wherein said anti-HLA is anti-HLA-DP.
42. The recombinant molecule of claim 40, wherein said anti-toll-like receptor is anti- toll-like receptor 2.
43. The recombinant molecule of claim 38, wherein said targeting unit(s) is/are a ligand.
44. The recombinant molecule of claim 43, wherein said ligand is soluble CD40 ligand or a chemokine.
45. The recombinant molecule of claim 43, wherein said ligand is a chemokine.
46. The recombinant molecule of claim 45, wherein said chemokine is RANTES or MTP-la.
47. The recombinant molecule of claim 45 wherein said chemokine is MTP-la.
48. The recombinant molecule of claim 38, wherein said targeting unit(s) is/are a bacterial antigen.
49. The recombinant molecule of claim 48, wherein the bacterial antigen is a flaggelin.
50. The recombinant molecule of claim 38, wherein the targeting units have the ability to target antigen presenting cells (APC).
51. The recombinant molecule of claim 38, wherein the targeting units have the ability to target HLA, CD 14, CD40, toll-like receptors, or chemokine receptors.
52. The recombinant molecule of claim 51, wherein said HLA is HLA-DP.
53. The recombinant molecule of claim 38, wherein the targeting units have the ability to target chemokine receptors.
54. The recombinant molecule of claim 38, wherein the antigenic unit(s) is/are an antigenic scFv.
55. The recombinant molecule of claim 54, wherein the antigenic scFv is derived from a monoclonal Ig produced by myeloma or lymphoma.
56. The recombinant molecule of claim 38, wherein the antigenic unit(s) is/are a telomerase, or a functional part thereof.
57. The recombinant molecule of claim 56, wherein said telomerase is hTERT.
58. The recombinant molecule of claim 38, wherein the antigenic unit(s) is/are derived from a bacterium.
59. The recombinant molecule of claim 58, wherein the bacterium derived antigenic unit(s) is/are a tuberculosis antigen.
60. The recombinant molecule of claim 38, wherein the antigenic unit(s) is/are derived from a virus.
61. The recombinant molecule of claim 60, wherein the virus derived antigenic unit(s) is/are derived from HIV.
62. The recombinant molecule of claim 61, wherein the HIV derived antigenic unit(s) is/are derived from gpl20.
63. The recombinant molecule of claim 38, wherein the dimerization motif comprises a hinge region and an immunoglobulin domain.
64. The recombinant molecule of claim 63, wherein the hinge region is Ig derived.
65. The recombinant molecule of claim 63, wherein the hinge region has the ability to form one or several covalent bonds.
66. The recombinant molecule of claim 65, wherein the covalent bond is a disulphide bridge.
67. The recombinant molecule of claim 63, wherein the immunoglobulin domain is a carboxyterminal C domain, or a sequence that is substantially homologous to said C domain.
68. The recombinant molecule of claim 67, wherein the carboxyterminal C domain is derived from IgG.
69. The recombinant molecule of claim 63, wherein the immunoglobulin domain has the ability to homodimerize.
70. The recombinant molecule of claim 63, wherein said immunoglobulin domain has the ability to homodimerize via noncovalent interactions.
71. The recombinant molecule of claim 70, wherein said noncovalent interactions are hydrophobic interactions.
72. The recombinant molecule of claim 38 comprising a nucleic acid formulated for administration to a patient to induce production of the recombinant antibody-based molecule.
73. The recombinant molecule of claim 38, wherein the nucleic acid is comprised by a vector.
74. The recombinant molecule of claim 73, wherein said vector is comprised by a cell line.
75. The recombinant molecule of claim 38, wherein the nucleic acid is comprised by a cell line.
76. A pharmaceutical composition comprising a recombinant molecule of claim 38 and a physiologically acceptable diluent or carrier.
77. A method of preparing the recombinant molecule of claim 38 comprising a. transfecting the vector of claim 73 into a cell population; b. culturing the cell population; c. collecting recombinant protein expressed from the cell population; and d. purifying the expressed protein.
78. A vaccine composition against cancer or infectious diseases comprising an immunologically effective amount of the nucleic acid of claim 38 or degenerate variants thereof, wherein said composition is able to trigger both a T-cell- and B-cell immune response.
79. The composition of claim 78 further comprising a pharmaceutically acceptable carrier.
80. The composition of claim 78, wherein said cancer is multiple myeloma or lymphoma.
81. The composition of claim 78, wherein said infectious disease is AIDS or tuberculosis.
82. A kit for preparation of a recombinant antibody-based molecule of anyone of claims 38 to 75.
PCT/NO2004/000051 2003-02-25 2004-02-25 Modified antibody WO2004076489A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
DK04714520.6T DK1599504T3 (en) 2003-02-25 2004-02-25 Modified antibody
EP14195614.4A EP2862878B1 (en) 2003-02-25 2004-02-25 Modified antibody
EP04714520.6A EP1599504B1 (en) 2003-02-25 2004-02-25 Modified antibody
AU2004215489A AU2004215489B2 (en) 2003-02-25 2004-02-25 Modified antibody
CA2517054A CA2517054C (en) 2003-02-25 2004-02-25 Use of nucleic acids encoding antibody-like molecules for use in medical treatment
ES04714520T ES2531204T3 (en) 2003-02-25 2004-02-25 Modified antibody
US13/353,548 US9169322B2 (en) 2003-02-25 2012-01-19 Modified antibody
US14/872,290 US9796787B2 (en) 2003-02-25 2015-10-01 Modified antibody
US15/730,776 US20180030155A1 (en) 2003-02-25 2017-10-12 Modified Antibody
US17/689,076 US20230048561A1 (en) 2003-02-25 2022-03-08 Modified Antibody
US18/163,484 US20230303718A1 (en) 2003-02-25 2023-02-02 Modified Antibody

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US45013403P 2003-02-25 2003-02-25
US60/450,134 2003-02-25
US10/786,907 US8932603B2 (en) 2003-02-25 2004-02-25 Modified antibody
US10/786,907 2004-02-25

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/786,907 Division US8932603B2 (en) 2003-02-25 2004-02-25 Modified antibody

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10/786,907 Division US8932603B2 (en) 2003-02-25 2004-02-25 Modified antibody
US13/353,548 Division US9169322B2 (en) 2003-02-25 2012-01-19 Modified antibody

Publications (2)

Publication Number Publication Date
WO2004076489A1 true WO2004076489A1 (en) 2004-09-10
WO2004076489A8 WO2004076489A8 (en) 2005-05-19

Family

ID=32930542

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NO2004/000051 WO2004076489A1 (en) 2003-02-25 2004-02-25 Modified antibody

Country Status (8)

Country Link
US (6) US8932603B2 (en)
EP (2) EP2862878B1 (en)
AU (1) AU2004215489B2 (en)
CA (1) CA2517054C (en)
DK (1) DK1599504T3 (en)
ES (1) ES2531204T3 (en)
PT (1) PT1599504E (en)
WO (1) WO2004076489A1 (en)

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007146968A2 (en) * 2006-06-12 2007-12-21 Trubion Pharmaceuticals, Inc. Single-chain multivalent binding proteins with effector function
WO2011059478A1 (en) 2009-11-16 2011-05-19 United States Of America As Represented By The Secretary Of The Navy Th1/th2 polarizing vaccines
WO2011161244A1 (en) 2010-06-25 2011-12-29 Vaccibody As Homodimeric protein constructs
WO2012064792A2 (en) * 2010-11-09 2012-05-18 Altimab Therapeutics, Inc. Protein complexes for antigen binding and methods of use
WO2013041966A1 (en) 2011-09-23 2013-03-28 University Of Oslo Vaccibodies targeted to cross-presenting dendritic cells
WO2013092875A1 (en) 2011-12-21 2013-06-27 Vaccibody As Vaccines against hpv
EP2762496A1 (en) 2013-02-05 2014-08-06 EngMab AG Method for the selection of antibodies against BCMA
WO2014122143A1 (en) 2013-02-05 2014-08-14 Engmab Ag Method for the selection of antibodies against bcma
WO2014140176A1 (en) * 2013-03-15 2014-09-18 Vaccibody As Targeting vaccines for veterinary use
US8853366B2 (en) 2001-01-17 2014-10-07 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
EP2789630A1 (en) 2013-04-09 2014-10-15 EngMab AG Bispecific antibodies against CD3e and ROR1
US9101609B2 (en) 2008-04-11 2015-08-11 Emergent Product Development Seattle, Llc CD37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof
WO2016055592A1 (en) 2014-10-09 2016-04-14 Engmab Ag Bispecific antibodies against cd3epsilon and ror1
WO2016169971A1 (en) 2015-04-20 2016-10-27 Pantec Biosolutions Ag Laser-assisted intradermal administration of active substances
WO2017118695A1 (en) * 2016-01-08 2017-07-13 Vaccibody As Therapeutic anticancer neoepitope vaccine
WO2018083204A1 (en) 2016-11-02 2018-05-11 Engmab Sàrl Bispecific antibody against bcma and cd3 and an immunological drug for combined use in treating multiple myeloma
US10143748B2 (en) 2005-07-25 2018-12-04 Aptevo Research And Development Llc B-cell reduction using CD37-specific and CD20-specific binding molecules
WO2019048928A1 (en) * 2017-09-07 2019-03-14 University Of Oslo Vaccine molecules
WO2019048936A1 (en) 2017-09-07 2019-03-14 University Of Oslo Vaccine molecules
EP3670535A1 (en) 2015-08-03 2020-06-24 EngMab Sàrl Monoclonal antibodies against bcma
US10940195B2 (en) 2014-01-13 2021-03-09 Baylor Research Institute Vaccines against HPV and HPV-related diseases
US10980869B2 (en) 2009-03-10 2021-04-20 Baylor Research Institute Fusion proteins comprising an anti-CD40 antibody and cancer antigens
WO2021219897A1 (en) 2020-05-01 2021-11-04 Vaccibody As Betacoronavirus prophylaxis and therapy
US11352426B2 (en) 2015-09-21 2022-06-07 Aptevo Research And Development Llc CD3 binding polypeptides
WO2022200590A1 (en) 2021-03-26 2022-09-29 Nykode Therapeutics ASA Therapeutic combination for treating cancer
WO2022233851A1 (en) 2021-05-03 2022-11-10 Nykode Therapeutics ASA Immunogenic constructs and vaccines for use in the prophylactic and therapeutic treatment of infectious diseases
WO2022238402A1 (en) 2021-05-10 2022-11-17 Nykode Therapeutics ASA Tolerance-inducing constructs and composition and their use for the treatment of immune disorders
WO2022238432A2 (en) 2021-05-10 2022-11-17 Nykode Therapeutics ASA Co-expression of constructs and immunoinhibitory compounds
WO2022238420A2 (en) 2021-05-10 2022-11-17 Nykode Therapeutics ASA Co-expression of constructs and immunostimulatory compounds
WO2022238395A1 (en) 2021-05-10 2022-11-17 Nykode Therapeutics ASA Tolerance-inducing constructs and compositions and their use for the treatment of immune disorders
WO2022238363A1 (en) 2021-05-10 2022-11-17 Nykode Therapeutics ASA Immunogenic constructs and vaccines for use in the prophylactic and therapeutic treatment of infectious diseases
WO2023079001A1 (en) 2021-11-03 2023-05-11 Nykode Therapeutics ASA Immunogenic constructs and vaccines for use in the prophylactic and therapeutic treatment of diseases caused by sars-cov-2
WO2024092025A1 (en) 2022-10-25 2024-05-02 Nykode Therapeutics ASA Constructs and their use

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2862878B1 (en) * 2003-02-25 2022-11-02 Nykode Therapeutics ASA Modified antibody
HUE025945T2 (en) 2005-02-15 2016-07-28 Univ Duke Anti-cd19 antibodies and uses in oncology
US20060263357A1 (en) 2005-05-05 2006-11-23 Tedder Thomas F Anti-CD19 antibody therapy for autoimmune disease
EP1999148B8 (en) 2006-03-06 2014-03-05 Medlmmune, LLC Humanized anti-cd22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
BRPI0811466A2 (en) 2007-05-07 2014-10-14 Medimmune Llc ISOLATED ANTIBODY, NUCLEIC ACID, VECTOR, ISOLATED CELL, METHODS FOR PRODUCTING AN ANTIBODY, TO TREAT DISEASE OR DISORDER, TO TREAT OR PREVENT REJECTION IN A HUMAN TRANSPLANT PATIENT TO BE TURNED TO HUMAN, TO EXHAUST T CELLS EXPRESSING ICOS IN A HUMAN PATIENT, TO BREAK THE GERMINAL CENTER ARCHITECTURE IN A PRIMARY SECONDARY LYMPHID ORGAN, TO DEPLETE GERMAN CENTRAL LYMPHIDE ORGAN B CELLS WITH A PRIMATE BULTA IN CURRENT CLASSES IN A PRIMATE, AND, PHARMACEUTICAL COMPOSITION.
EP2235064B1 (en) * 2008-01-07 2015-11-25 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
JP5933975B2 (en) 2008-11-12 2016-06-15 メディミューン,エルエルシー Antibody preparation
KR20130009760A (en) 2010-02-10 2013-01-23 이뮤노젠 아이엔씨 Cd20 antibodies and uses thereof
AU2011288412A1 (en) 2010-08-13 2013-02-21 Medimmune Limited Monomeric polypeptides comprising variant Fc regions and methods of use
WO2012022734A2 (en) 2010-08-16 2012-02-23 Medimmune Limited Anti-icam-1 antibodies and methods of use
US20180230218A1 (en) 2017-01-04 2018-08-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
WO2020014306A1 (en) 2018-07-10 2020-01-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6099846A (en) * 1992-10-14 2000-08-08 The Board Of Trustees Of The Leland Stanford Junior University Enhancement of B cell lymphoma and tumor resistance using idiotype/cytokine conjugates

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5703055A (en) * 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
US20050069549A1 (en) * 2002-01-14 2005-03-31 William Herman Targeted ligands
US20040052773A1 (en) 2002-01-18 2004-03-18 Inovio As Production of multi-chain protein from muscle
EP2862878B1 (en) * 2003-02-25 2022-11-02 Nykode Therapeutics ASA Modified antibody
KR102044948B1 (en) * 2010-06-25 2019-11-14 백시바디 에이에스 Homodimeric protein constructs
US20160031991A1 (en) * 2013-03-15 2016-02-04 Vaccibody As Targeting vaccines for veterinary use
KR20180100659A (en) * 2016-01-08 2018-09-11 백시바디 에이에스 Therapeutic anti-cancer neo-epitope vaccine

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6099846A (en) * 1992-10-14 2000-08-08 The Board Of Trustees Of The Leland Stanford Junior University Enhancement of B cell lymphoma and tumor resistance using idiotype/cytokine conjugates

Non-Patent Citations (47)

* Cited by examiner, † Cited by third party
Title
BENDANDI, M.; C. D. GOCKE ET AL.: "Complete molecular remissions induced by patient-specific vaccination plus granulocyte-monocyte colony-stimulating factor against lymphoma.", NAT MED, vol. 5, no. 10, 1999, pages 1171 - 7, XP003007895, DOI: doi:10.1038/13928
BIRAGYN A., ET AL.: "Genetic fusion of chemokines to a self tumor antigen induces protective, T-cell dependent antitumor immunity", NATURE BIOTECHNOLOGY, vol. 17, March 1999 (1999-03-01), pages 253 - 258, XP002108131 *
BIRAGYN A., ET AL.: "Toll-Like Receptor 4-Dependent Activation of Dentritic Cells by beta-Defensin 2", SCIENCE, vol. 298, 1 November 2002 (2002-11-01), pages 1025 - 1029, XP002979405 *
BIRAGYN, A.; K. TANI ET AL.: "Genetic fusion of chemokines to a self tumor antigen induces protective, T-cell dependent antitumor immunity.", NAT BIOTECHNOL, vol. 17, no. 3, 1999, pages 253 - 8, XP002108131, DOI: doi:10.1038/6995
BIRAGYN, A.; P. A. RUFFINI ET AL.: "Toll-like receptor 4-dependent activation of dendritic cells by beta-defensin 2.", SCIENCE, vol. 298, no. 5595, 2002, pages 1025 - 9, XP002979405, DOI: doi:10.1126/science.1075565
BOGEN, B.: "Monoclonal antibodies specific for variable and constant domains of murine lambda chains.", SCAND J IMMUNOL, vol. 29, no. 3, 1989, pages 273 - 9
BOGEN, B.; B. MALISSEN ET AL.: "Idiotope-specific T cell clones that recognize syngeneic immunoglobulin fragments in the context of class II molecules.", EUR J IMMUNOL, vol. 16, no. 11, 1986, pages 1373 - 8
BOGEN, B.; J. D. LAMBRIS: "Minimum length of an idiotypic peptide and a model for its binding to a major histocompatibility complex class II molecule.", EMBO J, vol. 8, no. 7, 1989, pages 1947 - 52
BOGEN, B.; L. GLEDITSCH ET AL.: "Weak positive selection of transgenic T cell receptor-bearing thymocytes: importance of major histocompatibility complex class II, T cell receptor and CD4 surface molecule densities.", EUR J IMMUNOL, vol. 22, no. 3, 1992, pages 703 - 9
BOGEN, B.; PERIPHERAL T: "cell tolerance as a tumor escape mechanism: deletion of CD4+ T cells specific for a monoclonal immunoglobulin idiotype secreted by a plasmacytoma", EUR J IMMUNOL., vol. 26, no. 11, November 1996 (1996-11-01), pages 2671 - 9
BRUNSVIK A: "Vaccibodies: future vaccines for B cell lymphoma and myeloma?", 5 February 2003
BRUNSVIK ET AL.: "Construction of tetrabodies for cancer vaccines", BANFF 2003, XP003005727 *
BRUNSVIK ET AL.: "Construction of tetrabodies for cancer vaccines", BANFF 2003, XP003005728 *
BRUNSVIK ET AL.: "Vaccibodies: future vaccines for 8 celllymphoma and myeloma?", BANFF 2003, XP003005729 *
CASTEN, L. A.; S. K. PIERCE: "Receptor-mediated B cell antigen processing. Increased antigenicity of a globular protein covalently coupled to antibodies specific for B cell surface structures.", J IMMUNOL, vol. 140, no. 2, 1988, pages 404 - 10
CHEN H.-W.: "Linkage of CD40L to a self-tumor antigen enhances the antitumor immune responses of dendritic cell-based treatment", CANCER IMMUNOL IMMUNOTHER, vol. 51, 2002, pages 341 - 348, XP002979403 *
EISEN, H. N.; E. S. SIMMS ET AL.: "Mouse myeloma proteins with antihapten antibody acitivity. The protein produced by plasma cell tumor MOPC-315.", BIOCHEMISTRY, vol. 7, no. 11, 1968, pages 4126 - 34
HAKIM, I.; S. LEVY ET AL.: "A nine-amino acid peptide from IL-lbeta augments antitumor immune responses induced by protein and DNA vaccines.", J IMMUNOL, vol. 157, no. 12, 1996, pages 5503 - 11, XP002167434
HOOGENBOOM H. R.: "Mix and match: Building manifold binding sites", NATURE BIOTECHNOLOGY, vol. 15, February 1997 (1997-02-01), pages 125 - 126, XP002110046 *
HOUGH, D. W.; R. P. EADY ET AL.: "Anti-idiotype sera raised against surface immunoglobulin of human neoplastic lymphocytes.", J EXP MED, vol. 144, no. 4, 1976, pages 960 - 9, XP000992837, DOI: doi:10.1084/jem.144.4.960
HU S.-Z., ET AL.: "Minibody: A Novel Engineered Anti-Carcinoembryonic Antigen Antibody Fragment (Single-Chain Fv-Ch3) Which Exhibits Rapid, High-level Targeting of Xenografts", CANCER RESEARCH, vol. 56, 1 July 1996 (1996-07-01), pages 3055 - 3061, XP002026337 *
HUANG H.-I.: "Improved immunogenicity of a self tumor antigen by covalent linkage to CD40 ligand", INT. J. CANCER, vol. 108, 2004, pages 696 - 703, XP002979404 *
HUANG T.-H., ET AL.: "Enhanced antitumor immunity by fusion of CTLA-4 to a self tumor antigen", BLOOD, vol. 96, no. 12, 1 December 2000 (2000-12-01), pages 3663 - 3670, XP002955727 *
HUANG, H. I.; P. Y. WU ET AL.: "Improved immunogenicity of a self tumor antigen by covalent linkage to CD40 ligand.", INT J CANCER, vol. 108, no. 5, 2004, pages 696 - 703, XP002979404, DOI: doi:10.1002/ijc.11612
KING, C. A.; M. B. SPELLERBERG ET AL.: "DNA vaccines with single-chain Fv fused to fragment C of tetanus toxin induce protective immunity against lymphoma and myeloma.", NAT MED, vol. 4, no. 11, 1998, pages 1281 - 6, XP002339736, DOI: doi:10.1038/3266
KRIANGKUM J., ET AL.: "Bispecific and bifunctional single chain recombinant antibodies", BIOMOLECULAR ENGINEERING, vol. 18, 2001, pages 31 - 40, XP004304464 *
KRISTOFFERSEN, G.; K. HANNESTAD ET AL.: "Two M315 idiotopes defined by isologous monoclonal antibodies: one depends on germline and the other on mutated murine lambda 2 light chain sequences.", SCAND J IMMUNOL, vol. 26, no. 5, 1987, pages 535 - 46
LAURITZSEN, G. F.; S. WEISS ET AL.: "Anti-tumour activity of idiotype-specific, MHC-restricted Thl and Th2 clones in vitro and in vivo.", SCAND J IMMUNOL, vol. 37, no. 1, 1993, pages 77 - 85
LAURITZSEN, G. F.; S. WEISS ET AL.: "Naive idiotype-specific CD4+ T cells and immunosurveillance of B-cell tumors.", PROC NATL ACAD SCI U S A, vol. 91, no. 12, 1994, pages 5700 - 4
LUNDE E., ET AL.: ""Troy-bodies": Recombinant Antibodies that Target T Cell Epitopes to Antigen Presenting Cells", INTERN. REV. IMMUNOL., vol. 20, 2001, pages 647 - 673, XP002979406 *
LUNDE E., ET AL.: "Troybodies and Pepbodies", BIOCHEMICAL SOCIETY, vol. 30, 2002, pages 500 - 506, XP002227237 *
LUNDE, E.; I. B. RASMUSSEN ET AL.: "Troy-bodies': antibodies as vector proteins for T cell epitopes.", BIOMOL EN, vol. 18, no. 3, 2001, pages 109 - 16, XP004305908, DOI: doi:10.1016/S1389-0344(01)00091-0
LUNDE, E.; K. H. WESTERN ET AL.: "Efficient delivery of T cell epitopes to APC by use of MHC class 11-specific Troybodies.", J IMMUNOL, vol. 168, no. 5, 2002, pages 2154 - 62
LUNDE, E.; L. A. MUNTHE ET AL.: "Antibodies engineered with IgD specificity efficiently deliver integrated T-cell epitopes for antigen presentation by B cells.", NAT BIOTECHNOL, vol. 17, no. 7, 1999, pages 670 - 5, XP002241695, DOI: doi:10.1038/10883
NEUBERGER, M. S.: "Expression and regulation of immunoglobulin heavy chain gene transfected into lymphoid cells.", EMBO J, vol. 2, no. 8, 1983, pages 1373 - 8, XP002713612
NORDERHAUG, L.; T. OLAFSEN ET AL.: "Versatile vectors for transient and stable expression of recombinant antibody molecules in mammalian cells.", J IMMUNOL METHODS, vol. 204, no. 1, 1997, pages 77 - 87, XP004107719, DOI: doi:10.1016/S0022-1759(97)00034-3
OLAFSEN, T.; 1. B. RASMUSSEN ET AL.: "IgM secretory tailpiece drives multimerisation of bivalent scFv fragments in eukaryotic cells.", IMMUNOTECHNOLOGY, vol. 4, no. 2, 1998, pages 141 - 53, XP004153638, DOI: doi:10.1016/S1380-2933(98)00014-1
OZATO, K.; N. MAYER ET AL.: "Hybridoma cell lines secreting monoclonal antibodies to mouse H-2 and Ia antigens.", J IMMUNOL, vol. 124, no. 2, 1980, pages 533 - 40
PLUCKTHUN A., ET AL.: "New protein engineering approaches to multivalent and bispecific antibody fragments", IMMUNOTECHNOLOGY, vol. 3, 1997, pages 83 - 105, XP004126672 *
RAVETCH, J. V.; S. BOLLAND: "IgG Fc receptors.", ANNU REV IMMUNOL, vol. 19, 2001, pages 275 - 90
RUFFINI P. A., ET AL.: "Idiotypic vaccination for B-cell malignancies as a model for therapeutic cancer vaccines: from prototype protein to second generation vaccines", HAEMATOLOGICA, vol. 87, 2002, pages 989 - 1001, XP002979407 *
SIRISINHA, S.; H. N. EISEN: "Autoimmune-like antibodies to the ligand-binding sites of myeloma proteins.", PROC NATL ACAD SCI U S A, vol. 68, no. 12, 1971, pages 3130 - 5
SNIDER, D. P.; D. M. SEGAL: "Targeted antigen presentation using crosslinked antibody heteroaggregates.", J IMMUNOL, vol. 139, no. 5, 1987, pages 1609 - 16, XP002667340
TAO, M. H.; R. LEVY: "Idiotype/granulocyte-macrophage colony-stimulating factor fusion protein as a vaccine for B-cell lymphoma.", NATURE, vol. 362, no. 6422, 1993, pages 755 - 8, XP002031411, DOI: doi:10.1038/362755a0
TJELLE, T.; CORTHAY, A.; LUNDE, E.; SANDLIE, I.; MICHAELSEN, TE.; MATHIESEN, I; BOGEN, B.: "Monoclonal antibodies produced by muscle after plasmid injection and electroporation.", J MOL THER, 2004
TOLLEFSEN, S.; T. TJELLE ET AL.: "Improved cellular and humoral immune responses against Mycobacterium tuberculosis antigens after intramuscular DNA immunisation combined with muscle electroporation.", VACCINE, vol. 20, no. 27-28, 2002, pages 3370 - 8, XP004378532, DOI: doi:10.1016/S0264-410X(02)00289-X
VAN SPRIEL A. B., ET AL.: "Immunotherapeutic perspective for bispecific antibodies", IMMUNOLOGY TODAY, vol. 21, no. 8, August 2000 (2000-08-01), pages 391 - 397, XP004215167 *

Cited By (76)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8853366B2 (en) 2001-01-17 2014-10-07 Emergent Product Development Seattle, Llc Binding domain-immunoglobulin fusion proteins
US10143748B2 (en) 2005-07-25 2018-12-04 Aptevo Research And Development Llc B-cell reduction using CD37-specific and CD20-specific binding molecules
US10307481B2 (en) 2005-07-25 2019-06-04 Aptevo Research And Development Llc CD37 immunotherapeutics and uses thereof
US8409577B2 (en) 2006-06-12 2013-04-02 Emergent Product Development Seattle, Llc Single chain multivalent binding proteins with effector function
JP2009539413A (en) * 2006-06-12 2009-11-19 トゥルビオン・ファーマシューティカルズ・インコーポレーテッド Single-chain multivalent binding protein with effector function
WO2007146968A3 (en) * 2006-06-12 2008-06-19 Trubion Pharmaceuticals Inc Single-chain multivalent binding proteins with effector function
CN105837690A (en) * 2006-06-12 2016-08-10 新兴产品开发西雅图有限公司 Single-chain multivalent binding proteins with effector function
WO2007146968A2 (en) * 2006-06-12 2007-12-21 Trubion Pharmaceuticals, Inc. Single-chain multivalent binding proteins with effector function
EP2418223A3 (en) * 2006-06-12 2013-01-16 Emergent Product Development Seattle, LLC Single-chain multivalent binding proteins with effector function
KR101571027B1 (en) 2006-06-12 2015-11-23 이머전트 프로덕트 디벨롭먼트 시애틀, 엘엘씨 Single-chain multivalent binding proteins with effector function
EP3805269A1 (en) * 2006-06-12 2021-04-14 Aptevo Research and Development LLC Single-chain multivalent binding proteins with effector function
JP2015070842A (en) * 2006-06-12 2015-04-16 エマージェント プロダクト デベロップメント シアトル, エルエルシー Single-chain multivalent binding proteins
RU2487888C2 (en) * 2006-06-12 2013-07-20 ЭМЕРДЖЕНТ ПРОДАКТ ДИВЕЛОПМЕНТ СИЭТЛ, ЭлЭлСи Single-chain multivalent binding proteins with effector function
US9101609B2 (en) 2008-04-11 2015-08-11 Emergent Product Development Seattle, Llc CD37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof
US11806390B2 (en) 2009-03-10 2023-11-07 Baylor Research Institute Fusion proteins comprising an anti-CD40 antibody and cancer antigens
US10980869B2 (en) 2009-03-10 2021-04-20 Baylor Research Institute Fusion proteins comprising an anti-CD40 antibody and cancer antigens
EP2488197A4 (en) * 2009-11-16 2013-05-15 Us Navy Th1/th2 polarizing vaccines
EP2488197A1 (en) * 2009-11-16 2012-08-22 The United States Of America As Represented By The Secretary Of The Navy Th1/th2 polarizing vaccines
WO2011059478A1 (en) 2009-11-16 2011-05-19 United States Of America As Represented By The Secretary Of The Navy Th1/th2 polarizing vaccines
US9795661B2 (en) 2009-11-16 2017-10-24 The United States Of America As Represented By The Secretary Of The Navy TH1/TH2 polarizing vaccines
US11479605B2 (en) 2010-06-25 2022-10-25 Nykode Therapeutics ASA Homodimeric protein constructs
JP2013532971A (en) * 2010-06-25 2013-08-22 バッシボディ アクスイェ セルスカプ Homodimeric protein construct
JP2016202185A (en) * 2010-06-25 2016-12-08 バッシボディ アクスイェ セルスカプ Homodimeric protein constructs
KR102044948B1 (en) 2010-06-25 2019-11-14 백시바디 에이에스 Homodimeric protein constructs
WO2011161244A1 (en) 2010-06-25 2011-12-29 Vaccibody As Homodimeric protein constructs
US10590195B2 (en) 2010-06-25 2020-03-17 Vaccibody As Homodimeric protein constructs
EP3441085A1 (en) 2010-06-25 2019-02-13 Vaccibody AS Homodimeric protein constructs
KR20180099918A (en) * 2010-06-25 2018-09-05 백시바디 에이에스 Homodimeric protein constructs
EP2638073A2 (en) * 2010-11-09 2013-09-18 Altimab Therapeutics, Inc. Protein complexes for antigen binding and methods of use
US9518132B2 (en) 2010-11-09 2016-12-13 Altimab Therapeutics, Inc. Protein complexes for antigen binding and methods of use
CN103328514B (en) * 2010-11-09 2015-12-02 阿尔蒂单抗治疗公司 The albumen composition combined for antigen and using method thereof
CN103328514A (en) * 2010-11-09 2013-09-25 阿尔蒂单抗治疗公司 Protein complexes for antigen binding and methods of use
WO2012064792A3 (en) * 2010-11-09 2012-07-19 Altimab Therapeutics, Inc. Protein complexes for antigen binding and methods of use
WO2012064792A2 (en) * 2010-11-09 2012-05-18 Altimab Therapeutics, Inc. Protein complexes for antigen binding and methods of use
EP2638073A4 (en) * 2010-11-09 2014-05-07 Altimab Therapeutics Inc Protein complexes for antigen binding and methods of use
JP2014534807A (en) * 2011-09-23 2014-12-25 ユニバーシティ オブ オスロUniversity of Oslo Vaccibody targeting dendritic cells for cross-presentation
WO2013041966A1 (en) 2011-09-23 2013-03-28 University Of Oslo Vaccibodies targeted to cross-presenting dendritic cells
CN104136039A (en) * 2011-09-23 2014-11-05 奥斯陆大学 Vaccibodies targeted to cross-presenting dendritic cells
CN104039833B (en) * 2011-12-21 2018-01-30 瓦西博迪公司 For hpv vaccine
EP3533462A1 (en) 2011-12-21 2019-09-04 Vaccibody AS Vaccines against hpv
RU2644201C2 (en) * 2011-12-21 2018-02-08 Вэксибоди Ас Vaccines against hpv
US9901635B2 (en) 2011-12-21 2018-02-27 Vaccibody As Vaccines against HPV
WO2013092875A1 (en) 2011-12-21 2013-06-27 Vaccibody As Vaccines against hpv
AU2012356969B2 (en) * 2011-12-21 2017-05-04 Nykode Therapeutics ASA Vaccines against HPV
CN104039833A (en) * 2011-12-21 2014-09-10 瓦西博迪公司 Vaccines against HPV
JP2015508284A (en) * 2011-12-21 2015-03-19 バッシボディ アクスイェ セルスカプ Vaccine against HPV
EP2762496A1 (en) 2013-02-05 2014-08-06 EngMab AG Method for the selection of antibodies against BCMA
WO2014122143A1 (en) 2013-02-05 2014-08-14 Engmab Ag Method for the selection of antibodies against bcma
EP3620468A1 (en) 2013-02-05 2020-03-11 EngMab Sàrl Method for the selection of antibodies against bcma
WO2014140176A1 (en) * 2013-03-15 2014-09-18 Vaccibody As Targeting vaccines for veterinary use
EP2789630A1 (en) 2013-04-09 2014-10-15 EngMab AG Bispecific antibodies against CD3e and ROR1
US11717567B2 (en) 2014-01-13 2023-08-08 Baylor Research Institute Vaccines against HPV and HPV-related diseases
US10940195B2 (en) 2014-01-13 2021-03-09 Baylor Research Institute Vaccines against HPV and HPV-related diseases
WO2016055592A1 (en) 2014-10-09 2016-04-14 Engmab Ag Bispecific antibodies against cd3epsilon and ror1
US11952421B2 (en) 2014-10-09 2024-04-09 Bristol-Myers Squibb Company Bispecific antibodies against CD3EPSILON and ROR1
WO2016169971A1 (en) 2015-04-20 2016-10-27 Pantec Biosolutions Ag Laser-assisted intradermal administration of active substances
US10828354B2 (en) 2015-04-20 2020-11-10 Pantec Biosolutions Ag Laser-assisted intradermal administration of active substances
EP3670535A1 (en) 2015-08-03 2020-06-24 EngMab Sàrl Monoclonal antibodies against bcma
US11352426B2 (en) 2015-09-21 2022-06-07 Aptevo Research And Development Llc CD3 binding polypeptides
WO2017118695A1 (en) * 2016-01-08 2017-07-13 Vaccibody As Therapeutic anticancer neoepitope vaccine
CN108495649A (en) * 2016-01-08 2018-09-04 瓦西博迪公司 The new epiposition vaccine of therapeutic anti-cancer
US11124577B2 (en) 2016-11-02 2021-09-21 Engmab Sàrl Bispecific antibody against BCMA and CD3 and an immunological drug for combined use in treating multiple myeloma
EP4295918A2 (en) 2016-11-02 2023-12-27 Bristol-Myers Squibb Company Bispecific antibody against bcma and cd3 and an immunological drug for combined use in treating multiple myeloma
WO2018083204A1 (en) 2016-11-02 2018-05-11 Engmab Sàrl Bispecific antibody against bcma and cd3 and an immunological drug for combined use in treating multiple myeloma
WO2019048936A1 (en) 2017-09-07 2019-03-14 University Of Oslo Vaccine molecules
WO2019048928A1 (en) * 2017-09-07 2019-03-14 University Of Oslo Vaccine molecules
WO2021219897A1 (en) 2020-05-01 2021-11-04 Vaccibody As Betacoronavirus prophylaxis and therapy
WO2022200590A1 (en) 2021-03-26 2022-09-29 Nykode Therapeutics ASA Therapeutic combination for treating cancer
WO2022233851A1 (en) 2021-05-03 2022-11-10 Nykode Therapeutics ASA Immunogenic constructs and vaccines for use in the prophylactic and therapeutic treatment of infectious diseases
WO2022238402A1 (en) 2021-05-10 2022-11-17 Nykode Therapeutics ASA Tolerance-inducing constructs and composition and their use for the treatment of immune disorders
WO2022238432A2 (en) 2021-05-10 2022-11-17 Nykode Therapeutics ASA Co-expression of constructs and immunoinhibitory compounds
WO2022238420A2 (en) 2021-05-10 2022-11-17 Nykode Therapeutics ASA Co-expression of constructs and immunostimulatory compounds
WO2022238395A1 (en) 2021-05-10 2022-11-17 Nykode Therapeutics ASA Tolerance-inducing constructs and compositions and their use for the treatment of immune disorders
WO2022238363A1 (en) 2021-05-10 2022-11-17 Nykode Therapeutics ASA Immunogenic constructs and vaccines for use in the prophylactic and therapeutic treatment of infectious diseases
WO2023079001A1 (en) 2021-11-03 2023-05-11 Nykode Therapeutics ASA Immunogenic constructs and vaccines for use in the prophylactic and therapeutic treatment of diseases caused by sars-cov-2
WO2024092025A1 (en) 2022-10-25 2024-05-02 Nykode Therapeutics ASA Constructs and their use

Also Published As

Publication number Publication date
US20230303718A1 (en) 2023-09-28
US20230048561A1 (en) 2023-02-16
US9169322B2 (en) 2015-10-27
WO2004076489A8 (en) 2005-05-19
US20160039946A1 (en) 2016-02-11
ES2531204T3 (en) 2015-03-11
US20180030155A1 (en) 2018-02-01
AU2004215489A1 (en) 2004-09-10
US20040253238A1 (en) 2004-12-16
US20120171242A1 (en) 2012-07-05
US9796787B2 (en) 2017-10-24
EP1599504A1 (en) 2005-11-30
US8932603B2 (en) 2015-01-13
EP1599504B1 (en) 2014-12-03
EP2862878B1 (en) 2022-11-02
EP2862878A1 (en) 2015-04-22
CA2517054A1 (en) 2004-09-10
AU2004215489B2 (en) 2010-07-15
PT1599504E (en) 2015-03-02
CA2517054C (en) 2016-05-10
DK1599504T3 (en) 2015-03-09

Similar Documents

Publication Publication Date Title
US20230048561A1 (en) Modified Antibody
EP3265575B1 (en) Cd20 binding molecules and uses thereof
US20200362047A1 (en) Single domain antibodies that bind to cd137
WO2020063787A1 (en) Anti-b7-h3 monoclonal antibody and use thereof in cell therapy
US7563445B2 (en) CD40 binding molecules and CTL peptides for treating tumors
US9249217B2 (en) Bispecific EGFRvIII x CD3 antibody engaging molecules
CA2448831A1 (en) Multi-stage cascade boosting vaccine
US20150132306A1 (en) HUMAN BISPECIFIC EGFRvIII ANTIBODY ENGAGING MOLECULES
WO2020199860A1 (en) Binder against programmed death-ligand and application thereof
US20160168263A1 (en) Certain Improved Human Bispecific EGFRvIII Antibody Engaging Molecules
WO1998056416A1 (en) Immunogenic compositions for induction of anti-tumor immunity
CN112513088A (en) anti-OX 40 antibodies, antigen-binding fragments thereof, and medical uses thereof
Rinaldi et al. Antibodies elicited by naked DNA vaccination against the complementary-determining region 3 hypervariable region of immunoglobulin heavy chain idiotypic determinants of B-lymphoproliferative disorders specifically react with patients’ tumor cells
AU2002225230B2 (en) Polypeptides capable of binding to CD64 and comprising one or more heterologous T cell epitopes, and their uses
Tunheim et al. Human receptors of innate immunity (CD14, TLR2) are promising targets for novel recombinant immunoglobulin-based vaccine candidates
Fló et al. Codelivery of DNA coding for the soluble form of CD86 results in the down-regulation of the immune response to DNA vaccines
EP1355946B1 (en) Dna vaccines expressing hypervariable vh-cdr3 idiotipic determinants
JP2021523688A (en) Anti-CD27 antibody and its use
WO2003090513A2 (en) Bi-specific antigen-binding compositions and related methods

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WR Later publication of a revised version of an international search report
WWE Wipo information: entry into national phase

Ref document number: 2004215489

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2517054

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2004215489

Country of ref document: AU

Date of ref document: 20040225

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004215489

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2004714520

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2004714520

Country of ref document: EP