EP1412390A2 - Agents pouvant activer ou inhiber le recepteur 9 de type toll - Google Patents

Agents pouvant activer ou inhiber le recepteur 9 de type toll

Info

Publication number
EP1412390A2
EP1412390A2 EP02795487A EP02795487A EP1412390A2 EP 1412390 A2 EP1412390 A2 EP 1412390A2 EP 02795487 A EP02795487 A EP 02795487A EP 02795487 A EP02795487 A EP 02795487A EP 1412390 A2 EP1412390 A2 EP 1412390A2
Authority
EP
European Patent Office
Prior art keywords
tlr9
molecule
binds
antibody
cpg
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP02795487A
Other languages
German (de)
English (en)
Inventor
Ling-Ling An
Herren Wu
Michael S. C. Fung
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Tanox Inc
Original Assignee
Tanox Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Tanox Inc filed Critical Tanox Inc
Publication of EP1412390A2 publication Critical patent/EP1412390A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • 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
    • 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
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55516Proteins; Peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55561CpG containing adjuvants; Oligonucleotide containing adjuvants

Definitions

  • the present invention is related to the identification of an epitope on the toll-like receptor, TLR9, responsible for interaction with the CpG motif.
  • An organism's immunity includes all of the mechanisms it uses for protection against foreign environmental agents, e.g., microorganisms, foods, chemicals, drugs, or pollens. In vertebrates, immunity may be either innate or acquired (adaptive).
  • Innate immunity is conferred by elements with which the organism is born. These include physical barriers, such as skin and mucosal membranes, internal elements, such as fever and coughs, a variety of chemical elements, such as interferon, serum proteins, e.g., lysozyme and polyamines, and cellular elements, e.g., macrophages, dendritic cells, and granulocytes.
  • physical barriers such as skin and mucosal membranes
  • internal elements such as fever and coughs
  • chemical elements such as interferon, serum proteins, e.g., lysozyme and polyamines
  • cellular elements e.g., macrophages, dendritic cells, and granulocytes.
  • the immune response to any antigen can be either Th1 or Th2, and each induces different cytokines, antibodies, and cellular responses.
  • the immune response to any antigen can be either Th1 or Th2, and each induces different cytokines, antibodies, and cellular responses.
  • CpG unmethylated CpG dinucleotides
  • Unmethylated CpG dinucleotides are abundant in all bacterial genomes, as well as viral and invertebrate eukaryotic genomes (Bird, A.P., Nuc. Acids Res.
  • CpG motifs can be found only at low frequency, and about 70% of vertebrate CpG motifs found are methylated. Id. Only about one-quarter of the predicted number of CG sequences are present in vertebrates as would be expected if base frequency were random (Bird, A.P., Trends in Genetics 3:342-347 (1987)). In contrast to bacterial CGs, human CG sequences are most frequently preceded by a C or followed by a G.
  • CpG-DNA is a potent Th1-like adjuvant triggering a strong Th1 biased antibody response, and concomitantly suppressing the Th2 response (Davis, H.L. et al., J. Immunol.160:870-876 (1998)).
  • CpG-DNA is also a potent single B-cell mitogen which is capable of driving more than 95% of B-cells into an activated state (Krieg, A.M. et al., Nature 374:546-549 (1995)).
  • CpG-ODNs synthesized unmethylated CpG- containing oligodeoxynucleotides
  • B-cells activated by bacterial CpG sequences or CpG-ODNs show increased expression of surface class-ll major histocompatibility complex (MHC) molecules and the co-stimulatory molecules B7-1 and B7-2 (Krieg, A.M., In: Delivery Strategies for Antisense Oli ⁇ onucleotide Therapeutics. Ed. Akhtar, S., CRC Press, Inc., pp 177-190; Davis, H.L. et al., J. Immunol. 160: 870-876 (1998)).
  • MHC major histocompatibility complex
  • CpG-ODNs strongly stimulate NK lytic activity and IFN- ⁇ production (Tokunaga, T et al., J. Natl. Cancer Institute 72:955-962 (1984); Yamamoto S., J. Immunol. 148:4072-4076 (1992)).
  • Antigen-presenting cells such as monocytes and dendritic cells, are activated by CpG-ODNs resulting in the production of Th1 cytokines (Jakob, T et al., J. Immunol. 161 :3042-3049 (1998)), as well as MHC- class II molecules and co-stimulatory B7-1 and B7-2 molecules (Stacy, K.J. et al., J. Immunol. 157:2116-2122 (1996); Sparwasser, T. et al., Eur. J. Immunol. 28:2045-2054 (1998)).
  • CpG-ODNs are, therefore, potent inducers and stimulators of a wide range of antigen-dependent and antigen-independent immune responses, and can be used for developing vaccines against cancer and infectious diseases. Since CpG-ODNs activate NK cells, they are useful for enhancing the antibody dependent cellular cytotoxicity (ADCC) of anti-tumor antibodies. CpG-ODNs can shift the T-cell response from a Th2-type response to a Th1 -type response, which can result in down-modulating of the allergic responses (Kline, J.N. et al., J. Immunol. 160:2555-2559 (1998); Sur, S. et al., J. Immunol.
  • the CpG-DNA driven innate immune response can be ablated by changing the CpG motif to a GC dinucleotide or by methylating the cytosine.
  • This clear structure-function relationship implied the existence of a receptor with specificity for the unmethylated CpG motif.
  • CpG function was completely dependent on the recently discovered toll-like receptor 9 (TLR9) (Hemmi, H. et al., Nature 408:740-745 (2000)). Based on in vitro studies, it is thought that the CpG motif interacts with a TLR9 after CpG-DNA has been internalized in endosomes (Wagner, H., Immunity 14:499-502 (2001)).
  • the TLR family consists of phylogenetically conserved transmembrane proteins that mediate innate immunity and are essential for microbial recognition.
  • the extracellular region of these receptors contains multiple leucine-rich repeats (LRRs) and a carboxy-terminal cysteine-rich domain having homology to the cytoplasmic domain of IL1 R.
  • LRRs leucine-rich repeats
  • dToll The first TLR, was found in Drosophila, and plays an important role in innate immune responses to fungal infection (Anderson, K.V., Curr. Opin. Immunol. 12:13-19 (2000); Means, T.K., Life Sci. 68:241 -258 (2000)).
  • Other TLR members were subsequently found in other organisms.
  • TLR2 mediates immune responses to peptidoglycan (PGN) and TLR4 mediates immune responses to lipopolysaccharides (LPS).
  • PPN peptidoglycan
  • LPS lipopolysaccharides
  • the present invention includes molecules that bind to a peptidic segment on TLR9 and mimic the effects of the CpG motif.
  • the CpG mimicking agents include, but are not limited to, antibodies, small-molecule compounds, peptides, peptide mimetics, and nucleic acids.
  • the invention includes compositions comprising molecules that bind to a peptidic segment on TLR9 and mimic the effects of the CpG motif suitable for administering to a patient in need of treatment, optionally in combination with, for example, an excipient, diluant, or carrier.
  • the present invention includes those molecules which bind to the TLR9's CXXC motifs at 255 Cys-Arg-Arg- 258 Cys (as CRRC) or at 265 Cys-Met- Glu- 268 Cys (as CMEC).
  • the present invention also includes methods of making molecules that bind to TLR9 and mimic the effects of the CpG motif. These methods include making monoclonal antibodies to the CpG epitope of TLR9.
  • the present invention includes a method of treating TLR9- mediated diseases comprising administering a CpG-mimicking agent including, but are not limited to, antibodies, small-molecule compounds, peptides, peptide mimetics, and nucleic acids.
  • Nucleic acids include oligonucleotides. These molecules may be used to treat, prevent, or ameliorate disorders such as tumors, cancer, or pathogenic infections, such as those caused by viruses, fungi, bacteria, or parasites.
  • the present invention also includes compositions suitable for administering to a patient suffering from an allergic disease comprising molecules that bind to TLR9 and mimic CpG function, optionally in combination with, for example, an excipient, diluant, or carrier.
  • the present invention includes a method of modulating the immune response by inducing a Th1-type response comprising administering molecules that bind to TLR9 and mimic CpG function. These molecules also shift the host cellular response away from a Th2-type response toward the Th1-type response.
  • administering the molecules of the present invention that bind to TLR9 may avoid the risk of Th2-mediated, immunization-induced anaphylaxis, making this method useful in immunotherapy and asthma treatment.
  • the molecules of the present invention may be administered in combination with a particular allergen.
  • the present invention includes administering molecules that bind to TLR9 and mimic CpG function as an artificial adjuvant in a mammal such as a mouse or a human.
  • the present invention includes a method of vaccination in a subject by administering a vaccine antigen or an antigen encoded in a DNA vaccine and a molecule or composition that binds to a peptidic segment on TLR9 and mimics the effects of the CpG motif.
  • the present invention also includes immunogens comprising synthetic peptides, recombinant proteins or a DNA encoding the peptide or the recombinant protein derived from TLR9 containing CRRC and/or CMEC, which induce the production of antibodies that bind to the epitope of TLR9 responsible for binding CpG, particularly at CRRC and/or CMEC, and activate the receptor's functions.
  • immunogens can be administered to a subject in order to immunize against cancer or allergic reactions.
  • Another embodiment of the invention includes molecules that bind TLR9 and inhibit or antagonize the function of the receptor.
  • the present invention includes molecules that antagonize the effects of CpG.
  • Another embodiment of the present invention includes gene constructs of antibodies or fragments thereof that bind to TLR9 for therapy.
  • the TLR9 antibody gene constructs will direct the synthesis of an antibody (or its fragments) capable of binding to TLR9 and either mimicking CpG function or inhibiting the function of the TLR9 receptor.
  • the gene construct to be expressed may also encode the epitope of TLR9 responsible for interaction with CpG.
  • These constructs include genes for whole antibody molecules as well as modified or derived forms thereof, including immunoglobulin fragments like Fab, single chain Fv (scFv) and F(ab') 2 .
  • the gene construct can be introduced into a host with conventional gene therapy techniques, including naked DNA, DNA incorporated in liposomes, DNA conjugated to lipids or to lipid derivatives, or via suitable plasmids or recombinant viral vectors.
  • Figure 1 shows the human cDNA sequence of toll-like receptor 9.
  • Figure 2 shows the protein sequence for human toll-like receptor 9.
  • This invention also covers the making and use molecules that bind to this peptidic segment on TLR9 and mimic the effects of CpG.
  • the CpG-mimicking agents could be, but not limited to, antibodies, small-molecule compounds, peptides, peptide mimetics, and nucleic acids.
  • These novel agents could be used in humans to treat cancer and infectious diseases, such as those caused by intracellular pathogens like Leishmania, Listeria, Francisella, Schistosoma, ebola, anthrax, and malaria.
  • these molecules could be used to treat allergic diseases, such as but not limited to, allergic rhinitis and asthma.
  • These novel agents could be used either alone or in combination or as conjugates when administered to humans.
  • Monoclonal antibodies to TLR9 that mimic CpG functions can be made by immunizing animals such as rodents, with synthetic peptides or recombinant proteins encompassing a peptidic segment derived from human TLR9 containing, e.g., CRRC or CMEC or both.
  • the immunogen could be DNA encoding the peptide segment derived from human TLR9.
  • Antibody molecules of the present invention include polyclonal or monoclonal antibodies, single chain antibodies, as well as functional fragments thereof.
  • Monoclonal antibodies include chimeric or humanized antibodies, human antibodies, or DelmmunisedTM antibodies.
  • Fragments of these antibodies include Fv, Fab, F(ab') 2 , single or double chain Fv fragments which retain the antigen binding function of the parent antibody.
  • the antibody may be produced by any recombinant method known in the art and may be produced in vitro or in vivo.
  • Single chain antibodies (“ScFv”) and the method of their construction are described in U.S. Patent No. 4,946,778.
  • Monoclonal antibodies may be made using the hybridoma method first described by Kohler et al., Nature, 256:495 (1975), or may be made by recombinant DNA methods (U.S. Pat. No. 4,816,567).
  • lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization.
  • lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp.59-103 (Academic Press, 1986)).
  • the hybridoma cells thus prepared are seeded and grown in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
  • Preferred myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium.
  • myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC- 11 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, Calif. USA, and SP2/0 or X63-Ag8-653 cells available from the American Type Culture Collection, Rockville, Md. USA.
  • Human myeloma and mouse- human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor, J. Immunol.
  • mice myeloma cell line NSO may also be used (European Collection of Cell Cultures, Salisbury, Wiltshire UK).
  • Culture medium in which hybridoma cells are grown is assayed for production of monoclonal antibodies directed against the antigen.
  • the binding specificity of monoclonal antibodies produced by hybridoma cells may be determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA).
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, pp.59-103 (Academic Press, 1986)). Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium.
  • the hybridoma cells may be grown in vivo as ascites tumors in an animal.
  • the monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by conventional immunoglobulin purification procedures such as, for example, protein A- Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (Innis M. et al. In PCR Protocols. A Guide to Methods and Applications. Academic, San Diego, CA (1990), Sanger, F.S, et al. Proc. Nat. Acad. Sci. 74:5463-5467 (1977)).
  • the hybridoma cells serve as a source of such DNA.
  • the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. Recombinant production of antibodies will be described in more detail below.
  • host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein
  • antibodies or antibody fragments can be isolated from antibody phage libraries generated using the techniques described in McCafferty et al., Nature 348:552-554 (1990). Clackson et al., Nature 352:624- 628 (1991) and Marks et al., J. Mol. Biol. 222:581-597 (1991) describe the isolation of murine and human antibodies, respectively, using phage libraries.
  • the DNA also may be modified, for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous murine sequences (U.S. Pat. No. 4,816,567; Morrison, et al., Proc. Nat. Acad. Sci. USA 81 :6851 (1984)), or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non- immunoglobulin polypeptide.
  • non-immunoglobulin polypeptides are substituted for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen.
  • Another alternative is to use electrical fusion rather than chemical fusion to form hybridomas. This technique is well established. Instead of fusion, one can also transform a B-cell to make it immortal using, for example, an Epstein Barr Virus, or a transforming gene. (See, e.g., "Continuously Proliferating Human Cell Lines Synthesizing Antibody of Predetermined Specificity," Zurawaki, V. R. et al, in Monoclonal Antibodies, ed. by Kennett R. H. et al, Plenum Press, N.Y. 1980, pp 19-33.)
  • Hybridomas producing specific anti-TLR9 monoclonal antibodies may be identified by ELISA using TLR9-derived antigen and by cell binding assays using cells expressing human TLR9.
  • the specific activity, either agonistic (mimicking CpG) or antagonistic (inhibiting CpG), of the antibodies will be tested by examining their effects on cell-surface molecule expression and Th1-type cytokine production in primary cultures of B-cells, T-cells, NK cells, monocytes/macrophages, and dendritic cells.
  • the functionally interesting antibodies will further be tested in animal models, such as those for allergy and asthma, tumor, intracellular pathogenic diseases, and vaccines.
  • a humanized antibody has one or more amino acid residues introduced into it from a source, which is non-human. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an "import” variable domain. Humanization can be essentially performed following the method of Winter and co-workers (Jones et al., Nature 321 :522-525 (1986); Riechmann et al., Nature 332:323-327 (1988); Verhoeyen, et al., Science, 239:1534-1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody.
  • humanized antibodies have substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species.
  • humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
  • variable domains both light and heavy
  • the choice of human variable domains, both light and heavy, to be used in making the humanized antibodies is very important to reduce antigenicity.
  • the sequence of the variable domain of a rodent antibody is screened against the entire library of known human variable-domain sequences.
  • the human sequence which is closest to that of the rodent is then accepted as the human framework (FR) for the humanized antibody (Sims et al., J. Immunol., 151 :2296 (1993); Chothia et al., J. Mol. Biol, 196:901 (1987)).
  • Another method uses a particular framework derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains.
  • humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences. Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences.
  • transgenic animals e.g., mice
  • transgenic mice are available from Abgenix, Inc., Fremont, California, and Medarex, Inc., Annandale, New Jersey. It has been described that the homozygous deletion of the antibody heavy-chain joining region (JH) gene in chimeric and germ-line mutant mice results in complete inhibition of endogenous antibody production. Transfer of the human germ-line immunoglobulin gene array in such germ-line mutant mice will result in the production of human antibodies upon antigen challenge. See, e.g., Jakobovits et al., Proc. Natl.
  • Human antibodies can also be derived from phage-display libraries (Hoogenboom et al., J. Mol. Biol. 227:381 (1991); Marks et al., J. Mol. Biol. 222:581-597 (1991); Vaughan, et al., Nature Biotech 14:309 (1996)). DelmmunisedTM Antibodies
  • DelmmunisedTM antibodies are antibodies in which the potential T cell epitopes have been eliminated, as described in International Patent Application PCT/GB98/01473. Therefore, immunogenicity in humans is expected to be eliminated or substantially reduced when they are applied in vivo.
  • antibodies can be chemically modified by covalent conjugation to a polymer to increase their circulating half-life, for example.
  • Preferred polymers, and methods to attach them to peptides are shown in U.S. Pat. Nos. 4,766,106; 4,179,337; 4,495,285; and 4,609,546 which are all hereby incorporated by reference in their entireties.
  • Preferred polymers are polyoxyethylated polyols and polyethylene glycol (PEG).
  • PEG is soluble in water at room temperature and has a preferred average molecular weight between 1000 and 40,000, more preferably between 2000 and 20,000, most preferably between 3,000 and 12,000. Other Molecules
  • the gene constructs of the present invention may be incorporated into a viral genome and subsequently packaged into suitable viral particles, which allows for a highly efficient gene delivery through viral infection.
  • Exemplary viral vectors commonly used for gene therapy include retrovirus vectors, adenovirus vectors, and adeno-associated virus (AAV) vectors.
  • the more recently developed viral vectors suitable for genetic therapy include lentivirus (HIV-1 or HIV-2 based vectors), and alphavirus vectors (based on Sindbis virus and Semliki Forest virus).
  • the gene constructs can be incorporated into viral genomes of retroviruses, lentiviruses or AAV vectors by subcloning of the transcriptional units into appropriate cassette vectors containing necessary sequences for virus packing. Upon DNA transfection of the resulting constructs into appropriate packaging cell lines that produce viral components, the recombinant viral genomes can be properly packaged into viable viral particles.
  • adenoviral genome is approximately 36 Kbp long, it is not convenient to directly insert the antibody gene into the genome through restriction endonuclease digestion and ligation. Instead, genes are inserted in a cassette vector such as pAvCvSv (Kobayashi K et al. (1996) J. Biol. Chem. 22:6852 - 60).
  • the vector has a pBR322 backbone and contains adenovirus type 5 (Ad5) 5' inverted terminal repeats (ITR), the Ad5 origin of replication, the Ad5 encapsidation signal, the E1 a enhancer, multiple cloning sites, and Ad5 sequence from nucleotide positions 3328 to 6246, which serve as a homologous recombination fragment.
  • Ad5 adenovirus type 5
  • ITR inverted terminal repeats
  • Homologous recombination between two DNAs in overlapping regions would allow the generation of a recombinant viral genome harboring the anti-TLR9 genes. This recombinant genome will be subsequently packaged into viable infectious viral particles in the 293 host cells. Incorporation of genes into the genome of alphaviruses or other viruses with a large genome can be similarly carried out to generate recombinant virus.
  • These gene constructs can be prepared as plasmids, which can be delivered to host cells or tissues, either directly or as naked DNA, or as DNA incorporated in liposomes, conjugated with appropriate lipid components, or incorporated in viral vectors. They are preferably injected for administration.
  • the gene constructs will be expected to direct the synthesis of molecules that bind to TLR9 or antibody fragments thereof, which will gradually enter the blood stream to interact with TLR9.
  • the recombinant virus constructs can be administered into an individual with allergic diseases via intra-muscular, intravenous, or subcutaneous routes. The dosage can be determined by extrapolating from animal experiments or determined in human clinical trials.
  • cells are transfected to express intrabodies that specifically target, bind to, or inhibit the TLR9 receptor.
  • An "intrabody” as used herein is an antibody that is expressed and active inside a cell. Intrabodies are typically not secreted and instead are directed to intracellularly expressed targets. The intrabodies typically bind to targets within the cell and thereby trap the targets in an intracellular compartment (e.g., the ER). Intrabodies are well known to those of skill in the art (see, e.g., Chen et al., Hum. Gene Therap. 7: 1515- 1525 (1996); Marasco Immunotech. 1 : 1-19 (1995); and Maciejewski et al. Nature Med. 1 : 667-673 (1995)).
  • intrabodies can be used to modulate cellular physiology and metabolism by a wide variety of mechanisms.
  • binding of an intrabody may be used to block or stabilize macromolecular interactions, modulate enzyme function by occluding an active site, sequestering substrate or fixing the enzyme in an active or an inactive conformation as the need may be.
  • Intrabodies may also be used to divert proteins from their usual cellular compartment for example by sequestering transcription factors in the cytoplasm, or by retention in the ER of the proteins destined for the cell surface.
  • intrabodies may be useful in conjunction with the present invention to trigger signal transduction mimicking CpG.
  • Additional pharmaceutical vehicles could be used to control the duration of action of the molecules of the invention. They could be entrapped in microcapsules prepared by coacervation techniques or by interracial polymerization (hydroxymethylcellulose or gelatin microcapsules) in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Methods of preparing liposome delivery systems are discussed in Gabizon et al., Cancer Research 42:4734 (1982); Cafiso, Biochem Biophys Ada 649:129(1981 ); and Szoka, Ann Rev Biophys Eng 9:467 (1980).
  • Liquid pharmaceutical compositions may be lyophilized to prevent degradation and to preserve sterility. Methods for lyophilizing liquid compositions are known to those of ordinary skill in the art. Just prior to use, the composition may be reconstituted with a sterile diluent (Ringer's solution, distilled water, or sterile saline, for example), which may include additional ingredients. Upon reconstitution, the composition is administered to subjects.
  • a sterile diluent Finger's solution, distilled water, or sterile saline, for example
  • the molecules of the present invention can be administered by any of a number of routes and are administered at a concentration that is therapeutically effective in the indication or for the purpose sought.
  • the antibodies may be formulated using a variety of acceptable excipients known in the art.
  • the antibodies are administered by injection, either intravenously or intraperitoneally. Methods to accomplish this administration are known to those of ordinary skill in the art. It may also be possible to obtain compositions which may be topically or orally administered, or which may be capable of transmission across mucous membranes.
  • the dosage and mode of administration will depend on the individual and the agent to be administered.
  • the dosage can be determined by routine experimentation in clinical trials or extrapolation from animal models in which the antibody was effective.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne des molécules qui se lient à un segment peptidique sur TLR9, et simulent les effets du motif CpG. Les agents simulant CpG comprennent, mais pas exclusivement, des anticorps, des composés à petites molécules, des peptides, des mimétiques de peptides, et des acides nucléiques, y compris des compositions comprenant des molécules qui se lient à un segment peptidique sur TLR9 et simulent les effets du motif CpG, et qui peuvent être administrées à un patient nécessitant un traitement, éventuellement en association avec, par exemple, un excipient, un diluant ou un support. L'invention concerne également celles des molécules qui se lient aux motifs CXXC de TLR9, au niveau de 255Cys-Arg-Arg-258Cys (comme CRRC) ou au niveau de 265Cys-Met-Glu-268Cys (comme CMEC). L'invention concerne en outre des méthodes de modulation d'une réponse immunitaire par induction d'une réponse de type Th1, qui consiste à administrer des molécules qui se lient à TLR9 et simulent la fonction de CpG. Ces molécules déplacent également la réponse cellulaire hôte et l'éloigne d'une réponse de type Th2 vers la réponse de type Th1. Ainsi, l'administration des molécules de l'invention, qui se lient à TLR9, peut éviter le risque d'une anaphylaxie dépendante de Th2 et induite par l'immunisation, ce qui rend la méthode utile pour le traitement de l'immunothérpie et de l'asthme. Les molécules de l'invention peuvent être adminsistrées en association avec un allergène particulier.
EP02795487A 2001-07-26 2002-07-25 Agents pouvant activer ou inhiber le recepteur 9 de type toll Withdrawn EP1412390A2 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US30806801P 2001-07-26 2001-07-26
US308068P 2001-07-26
PCT/US2002/023645 WO2003035695A2 (fr) 2001-07-26 2002-07-25 Agents pouvant activer ou inhiber le recepteur 9 de type toll

Publications (1)

Publication Number Publication Date
EP1412390A2 true EP1412390A2 (fr) 2004-04-28

Family

ID=23192407

Family Applications (1)

Application Number Title Priority Date Filing Date
EP02795487A Withdrawn EP1412390A2 (fr) 2001-07-26 2002-07-25 Agents pouvant activer ou inhiber le recepteur 9 de type toll

Country Status (4)

Country Link
EP (1) EP1412390A2 (fr)
CN (1) CN1642982A (fr)
AU (1) AU2002360243A1 (fr)
WO (1) WO2003035695A2 (fr)

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002022809A2 (fr) 2000-09-15 2002-03-21 Coley Pharmaceutical Gmbh Procede de criblage a haut rendement d'immuno-agoniste/antagoniste base sur cpg
NZ531612A (en) 2001-09-07 2006-01-27 Univ Boston Method and composition for treating immune complex associated disorders
BRPI0411514A (pt) * 2003-06-20 2006-08-01 Coley Pharm Gmbh antagonistas de receptor toll-like de molécula pequena
US20050208470A1 (en) * 2003-12-16 2005-09-22 University Of Massachusetts Toll-like receptor assays
JP2008507530A (ja) * 2004-07-20 2008-03-13 シェーリング コーポレイション Toll様受容体を発現する腫瘍細胞におけるアポトーシスの誘導
EP1829895A1 (fr) * 2006-03-03 2007-09-05 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Molécule bispécifique anti-TLR9 et anti-CD32 contenant un épitope de lymphocyte T pour le traitement des allergies
US9636415B2 (en) 2006-03-03 2017-05-02 S-Target Therapeutics Gmbh Bispecific molecule binding TLR9 and CD32 and comprising a T cell epitope for treatment of allergies
CN101240271B (zh) * 2006-08-28 2012-08-29 长春华普生物技术有限公司 Toll样受体调节性寡核苷酸及其用途
CN101134046B (zh) * 2006-09-01 2011-10-05 中国医学科学院药物研究所 Tlr4和tlr9激动剂复方在抑制肿瘤转移中的应用
US20110105383A1 (en) * 2008-09-10 2011-05-05 Magnus Hook Methods and compositions for stimulation of mammalian innate immune resistance to pathogens
BRPI1009842B8 (pt) 2009-03-25 2021-05-25 Univ Texas uso de agonista de tlr9 e agonista de tlr2/6 na preparação de uma composição farmacêutica para tratamento, inibição ou atenuação de uma infecção microbiana, bem como composição farmaceuticamente aceitável compreendendo os referidos agonistas
EP2437772B1 (fr) 2009-06-05 2017-12-06 13Therapeutics, Inc. Peptides immunorégulateurs et procédés d'utilisation
US20120282276A1 (en) * 2009-11-05 2012-11-08 The Regents Of The University Of Michigan Biomarkers predictive of progression of fibrosis
CN101712957B (zh) * 2009-11-25 2011-11-23 中国人民解放军第三军医大学第一附属医院 Toll样受体9阻断剂及应用
FR2975600B1 (fr) 2011-05-24 2013-07-05 Assist Publ Hopitaux De Paris Agents pour le traitement de tumeurs
AR095882A1 (es) 2013-04-22 2015-11-18 Hoffmann La Roche Terapia de combinación de anticuerpos contra csf-1r humano con un agonista de tlr9
WO2015173342A1 (fr) 2014-05-16 2015-11-19 Ablynx Nv Procédés de détection et/ou de mesure d'anticorps anti-médicament, en particulier d'anticorps anti-médicament émergents dans un traitement
HUE050007T2 (hu) 2014-05-16 2020-11-30 Ablynx Nv Immunglobulin variábilis domének
WO2016044839A2 (fr) 2014-09-19 2016-03-24 The Board Of Regents Of The University Of Texas System Compositions et méthodes pour traiter des infections virales par le biais de l'immunité innée stimulée en combinaison avec des composés antiviraux
WO2019066571A2 (fr) 2017-09-28 2019-04-04 연세대학교 산학협력단 Procédé de production de cellules suppressives d'origine myéloïde, cellules suppressives d'origine myéloïde ainsi produites et leurs utilisations
US20210317454A1 (en) * 2020-04-10 2021-10-14 Idera Pharmaceuticals, Inc. Use of a tlr9 agonist in methods for treating covid-19
JP2024518616A (ja) * 2021-05-18 2024-05-01 ホン,ミェン―チェ ワクチン、その使用、及びがんワクチン混合物

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0001704D0 (en) * 2000-01-25 2000-03-15 Glaxo Group Ltd Protein
SK287689B6 (sk) * 2000-06-08 2011-06-06 Intercell Ag Použitie molekuly oligodeoxynukleovej kyseliny na výrobu imunostimulačnej farmaceutickej kompozície a farmaceutický prípravok
JP2002034565A (ja) * 2000-07-19 2002-02-05 Japan Science & Technology Corp 細菌dnaを特異的に認識する受容体タンパク質

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO03035695A3 *

Also Published As

Publication number Publication date
WO2003035695A2 (fr) 2003-05-01
CN1642982A (zh) 2005-07-20
WO2003035695A3 (fr) 2003-11-13
AU2002360243A1 (en) 2003-05-06

Similar Documents

Publication Publication Date Title
EP1412390A2 (fr) Agents pouvant activer ou inhiber le recepteur 9 de type toll
EP1470159B1 (fr) Anticorps dirige contre des proteines membranaires latentes (lmp) et utilisations de ceux-ci
AU765822B2 (en) CD40 binding molecules and CTL peptides for treating tumors
JP4764585B2 (ja) タンパク質抗原およびペプチド抗原の免疫原性を増強するためのfc融合タンパク質
US6294654B1 (en) Modified immunoglobulin molecule incorporating an antigen in a non-CDR loop region
JP2023524098A (ja) 感染症を治療する方法
JP2011502163A (ja) 持続性ウイルス感染を治療するための併用療法
US11471522B2 (en) Methods and compositions for stimulating immune response
ZA200604053B (en) Uses of specific histones for the treatment of parasitic disease
CZ302801B6 (cs) Použití protilátky namírené proti antigenu bunecné membrány Ep-CAM pro výrobu farmaceutického prostredku
US20210030783A1 (en) Compositions and methods for tumor immunotherapy
EP3292139B1 (fr) Peptides ctl h3.3 et leurs utilisations
US20110064745A1 (en) Therapeutic Antibodies, Antibody Fragments And Antibody Conjugates
US6787524B2 (en) CpG oligonucleotides and related compounds for enhancing ADCC induced by anti-IgE antibodies
KR20220131895A (ko) 항-pd-1/il-15 면역사이토카인을 이용하여 pd-1을 표적으로 하는 신규한 면역요법
JP7086000B2 (ja) 体液性親和性の加速に関する方法及び組成物
JP2006514921A (ja) 抗−tsg101抗体およびウイルス感染の処置に対するそれらの用法
CA3214934A1 (fr) Polymersomes comprenant un polynucleotide encapsule soluble et un lipide ionisable, ainsi que leurs procedes de fabrication et d'utilisation
KR100372958B1 (ko) 면역반응을개시시키기위해다중에피토프항원의입체형태를변화시키기위한방법및조성물
Stevenson et al. DNA vaccines against B-cell tumors
Miyagi et al. The Timing of GM-CSF Expression Plasmid
MXPA98009586A (en) Method and composition for the reconformation of multi-peptide antigens to start an animal response

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20040219

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LI LU MC NL PT SE SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

17Q First examination report despatched

Effective date: 20071030

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20080710