WO2002077208A1 - Diminution de reactions immunitaires specifiques dependante de certains antigenes par un effet de co-stimulation - Google Patents

Diminution de reactions immunitaires specifiques dependante de certains antigenes par un effet de co-stimulation Download PDF

Info

Publication number
WO2002077208A1
WO2002077208A1 PCT/EP2002/003292 EP0203292W WO02077208A1 WO 2002077208 A1 WO2002077208 A1 WO 2002077208A1 EP 0203292 W EP0203292 W EP 0203292W WO 02077208 A1 WO02077208 A1 WO 02077208A1
Authority
WO
WIPO (PCT)
Prior art keywords
antigen
presenting cell
monoantigenic
receptor
monoantigenic antigen
Prior art date
Application number
PCT/EP2002/003292
Other languages
German (de)
English (en)
Inventor
Ahmed Sheriff
Birgit Vogt
Original Assignee
Genethor Gmbh
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 Genethor Gmbh filed Critical Genethor Gmbh
Publication of WO2002077208A1 publication Critical patent/WO2002077208A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0008Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4614Monocytes; Macrophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4615Dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4621Cellular immunotherapy characterized by the effect or the function of the cells immunosuppressive or immunotolerising
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4622Antigen presenting cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4637Other peptides or polypeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/46433Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/46434Antigens related to induction of tolerance to non-self
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/464839Allergens
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0639Dendritic cells, e.g. Langherhans cells in the epidermis
    • C12N5/064Immunosuppressive dendritic cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0645Macrophages, e.g. Kuepfer cells in the liver; Monocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/122Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells for inducing tolerance or supression of immune responses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells

Definitions

  • the present invention relates to antigen-presenting cells (APC), processes for producing antigen-presenting cells, medicaments containing antigen-presenting cells and use of the antigen-presenting cells.
  • APC antigen-presenting cells
  • an allergen that gets into the blood in some way can trigger anaphylaxis: an allergic reaction in regions of the body that are far from its point of entry. Severe anaphylactic shocks can upset all normal bodily functions and can be fatal.
  • the allergic reactions manifest themselves differently, they are always started by the same mechanism: the sensitization.
  • an allergen typically a protein
  • the allergenic substance meets so-called phagocytes or macrophages. These gobble up the foreign substance, dismember it and present the fragments on the cell surface with MHC II molecules.
  • T-helper lymphocytes recognize the fragments presented and bind to them.
  • the T helper lymphocytes are activated by the macrophages and then in turn activate some B lymphocytes, which also recognize the allergen.
  • the B cells then mature into plasma cells that produce antibodies. First of all, these are antibodies of the so-called IgM type; from a certain point in time, however, the plasma cells switch to IgE antibodies.
  • IgE molecules With their Fc region, they attach themselves to IgE receptors of two different classes of immune system cells.
  • mast cells which are usually found in the body tissues near blood vessels and epithelial cells. There is contact with the outside world via the epithelium (this also includes the epithelium of the respiratory tract and gastrointestinal tract).
  • IgE antibodies also bind to basophilic granulocytes (basophils). However, these cells circulate in the bloodstream.
  • IgEs Once production of the IgEs begins, it apparently lasts for months, sometimes even years. As a result, they constantly occupy IgE receptors on mast cells and basophils - ready to take immediate action the next time they come into contact with allergens.
  • the second exposure initiates a stage of the hypersensitivity reaction, which also appears externally.
  • the allergy-causing substance binds to the IgEs of the mast cells within seconds of contact with human tissue. If he attaches himself to two or more IgE molecules at the same time, he bridges between them. Such cross-links bring the affected IgE receptors closer together, and this activates the cell so that it releases highly effective substances that directly produce allergic symptoms. (The release can also be caused in other ways; allergic reactions are only spoken if IgEs are involved). The most important of these substances is histamine.
  • the second group of mediators consists mainly of prostaglandins and leukotrienes. They are only released after the allergen molecules have attached to the IgEs on the cells. Like histamine, they narrow the bronchi and dilate the blood vessels. However, their effects last longer.
  • stimulated mast cells emit a variety of potentially toxic enzymes. They also appear to release cytokines that regulate the activities of other immune cells.
  • Antihistamines are usually effective and still serve as standard therapy. The latest variants can no longer easily cross the blood-brain barrier and no longer make the patient tired. If antihistamines are ineffective in severe inflammation, inhalable corticosteroids, which are usually prescribed to relieve chronic inflammation in asthma, often help. In severe cases, immunotherapy or hyposensitization (also known as allergy shots or desensitization), which was introduced in 1911, can provide relief in the long term. With this treatment, doctors inject patients with increasing doses of the allergen to which they are sensitive. The dose is decisive in all cases: too little allergen does not confer tolerance. In addition, protection is rarely complete.
  • Bronchodilators are the most widely used drugs for asthma. They relieve the symptoms caused by histamine and other bronchoconstrictors very quickly, but are unlikely to affect the underlying inflammation. In addition, their excessive use can cause a back reaction of the body, so that after their effects have subsided, the respiratory flow is more restricted than before. In addition, the methods listed under 1. are used.
  • Another new strategy is the use of humanized monoclonal anti-IgE antibodies against the Fc ⁇ RI binding region for IgE. This prevents the binding of IgE to the IgE receptor, so that no mediators of the allergic reaction from mast cells or basophils can be released. This strategy has shown in clinical studies in patients with allergic rhinitis and allergic asthma that these antibodies are well tolerated and reduce the allergic reactions.
  • Tissue transplantation to replace diseased organs is an important medical therapy today.
  • a response from the adaptive immune system to the graft poses the greatest threat to successful treatment.
  • Responses from the adaptable immune system are induced by antigen presenting cells by activating T helper lymphocytes.
  • the ABO and Rh blood group antigens When transfusing blood, which is the first and most commonly used graft, the ABO and Rh blood group antigens must be matched to avoid the rapid destruction of inappropriate erythrocytes.
  • the very polymorphic main histocompatibility complexes (MHC) have to be matched to one another, since these almost always trigger the immune response. Unfortunately, the perfect matching of the MHCs is almost impossible, except for relatives.
  • the product of the mouse PD-1 gene (ACCESSION NM_021893), a member of the IgG superfamily, is a receptor that is common in many tissues. Analyzes carried out by flow cytometry and immunoprecipitation with the monoclonal antibody J43mAk showed that the PD-1 gene product is a 50 to 55 kDa membrane protein. The PD-1 protein appears to be highly glycosylated since the calculated molecular weight of the amino acid sequence is 29310 Da. Normal mouse lymphoid tissue such as thymus, spleen, lymph nodes and bone marrow contain only a small number of PD-1 positive cells.
  • PD-1 positive population appears in thymocytes as well as on T cells in spleen and lymph nodes due to the in vivo treatment with anti-CD3 monoclonal antibodies.
  • PD-1 antigen expression was strongly induced by in vitro stimulation in various subgroups of thymocytes and spleen T cells, either with anti-CD3 mAb or Concavalin A, which can cause T cells to activate as well as to die , PD-1 expression on spleen B cells was similarly stimulated with anti-IgM Ab.
  • PD-1 was not significantly expressed after treatments such as growth factor deprivation, dexamethasone or lipopolysaccharide.
  • PD-1 is structurally similar to CTL-associated antigen 4 (CTLA-4), which binds B7-1 and B7-2 and plays a crucial role in the maintenance of T cell homeostasis (for reviews, see (Sperling & Bluestone 1996; Thompson & Allison 1997)).
  • CTLA-4 CTL-associated antigen 4
  • PD-1 does not contain the MYPPPY motif, which is critical for B7-1 and B7-2 binding.
  • the extracellular region of PD-1 and CTLA-4 each consist of an IgV domain with 23% identity to each other.
  • a ligand (PD-Ll) exists for PD-1. The binding of PD-1 by PD-Ll leads to the inhibition of TCR-mediated T cell proliferation and cytokine secretion (Freeman et al 2000).
  • the human and mouse PD-Ll molecule (EMBL / GenBank / DDBS under accession nos. AF233516 and AF233517, (Freeman et al 2000)) are members of the B7 gene family and have a similar structural organization, which consists of an IgV and an IgC domain in the extracellular region (Boussiotis et al 1996), a hydrophobic transmembrane domain, followed by a short, charged intracellular region.
  • Human PD-Ll is identical to B7-H1 at 290 amino acids, which is reported to activate T cell stimulation (Dong et al 1999).
  • the mouse PD-Ll cDNA encodes a polypeptide with 70% amino acid identity to the human PD-Ll.
  • PD-Ll has amino acid identities of 21, 20, and 23% to B7-1, B7-2, and ICOS (ligand of inducible co-stimulator) (Boussiotis et al 1996; Ling et al 2000; Swallow et al 1999; Yoshinaga et al 1999).
  • the expression patterns of B7-1, B7-2, and PD-Ll are different.
  • B7-2, one of the ligands of CD28 and CTLA-4, is constitutively expressed on monocytes. However, the constitutive expression of B7-1 and B7-2 cannot be observed in any organ.
  • B7-1 and B7-2 expression can be induced in dendritic cells, macrophages and B cells (Boussiotis et al 1996), as well as some types of fibroblasts and epithelial cells.
  • PD-Ll is constitutively expressed by non-lymphoid, parenchymal organs such as the heart, placenta, skeletal muscles and lungs, but not the small intestine (Dong et al 1999).
  • PD-Ll is also expressed in some cancers. These tumors may be using PD-Ll to inhibit anti-tumor immune responses.
  • PD-1 Ligand 2 (PD-L2) is a second ligand for PD-1. Binding of PD-1 to PD-L2 dramatically inhibits T cell receptor (TCR) mediated proliferation and cytokine production by CD4 T cells. At low antigen concentrations, PD-L2-PD-1 interactions inhibit strong B7-CD28 signals.
  • PD-L-PD-1 interactions lead to cell cycle residue in G 0 / Gx (cell cycle phases) but do not increase the number of dead cells.
  • PD-L2 expression on APCs is increased by treatment with interferon ⁇ and could also be detected in some other tissues and tumor cell lines.
  • PD-Ll and PD-L2 thus have overlapping functions (Latchman et al 2001).
  • mPD ⁇ L2 (previous name: Protein AF142780) codes for a polypeptide with 38% amino acid identity to mPD-Ll.
  • Murines and human PD-L2 have 70% amino acid identity.
  • the five members of the B7 family - B7-1, B7-2, ICOS-L, PD-Ll and PD-L2 - have 21-27% amino acid identity and a structural organization consisting of a signal sequence, an IgV-like one IgC-like and a transmembrane domain and a short cytoplasmic tail.
  • the cytoplasmic tail of PD-Ll is conserved between mice and humans, which is in contrast to the poor conservation of the cytoplasmic tail of PD-L2 of humans and mice (Latchman et al 2001).
  • the PD-L2 tissue distribution is similar to that of PD-Ll. There is expression in the placenta, heart, pancreas, lungs and liver, low expression in the spleen, lymph nodes and thymus, no expression in unstimulated monocytes, but this could be induced by interferon ⁇ . However, the kinetics of this induction is slower than that of PD-Ll.
  • Autoimmune diseases are chronic diseases. These include rheumatism in various clinical forms, diabetes, multiple sclerosis, certain forms of cardiac muscle inflammation and Thyroid disease. The series of autoimmune diseases could be continued as desired, although approximately 90% epidemiologically only make up a small percentage.
  • autoimmune diseases are antibody-mediated.
  • immunology this means that the organism produces antibodies for mostly unknown reasons, which are directed against the body's own cellular structures (autoantigens). Once these antibodies have been formed, their interaction and binding to the respective organ or cell-specific structures trigger a cell and tissue-destroying reaction of the immune system. Ultimately, this leads to the clinical picture of the disease (Steinman 1994).
  • PCT / EP 00/03984 discloses a method for reducing specific immune reactions by suppressing the B7 expression of cells presenting antigen while simultaneously expressing predetermined (in particular pathological) antigens.
  • WO-A-01/14557 relates to PD-1 as a receptor for B7-4.
  • B7-4 can inhibit immune cell activation due to binding to an inhibitory receptor or to an immune cell.
  • Active substances are described for the modulation of PD-1, B7-4 and their interaction in order to modulate co-stimulating or inhibiting signals in an immune cell, which results in a modulation of the immune response.
  • a problem on which the invention is based is, inter alia, to provide genetic engineering, therapeutically usable products for the reduction of specific immune reactions in which targets (antigens, autoantigens) and antibodies, autoantibodies are known.
  • the antigen-presenting cell which predominantly presents certain antigens beforehand (monoantigenic antigen-presenting cell) and is characterized in that molecules which bind PD-1 are preferably produced in the monoantigenic antigen-presenting cell and if appropriate, CTLA4-binding molecules are produced and, if appropriate, one of the functions of stimulatory receptors, such as a B7 and / or CD40 receptor, is suppressed.
  • CTLA4 (CD152) is an analogue of CD28, also binds B7, but induces apoptosis of the T cell.
  • CTLA4 occurs mainly in intracellular vesicles and is only released after activation of the cells.
  • PD-1 is a CD28 and CTLA4 analogue and develops CTLA4-like effects.
  • Fig. 3 shows schematically the function of gene manipulation at the molecular level: PD-1 is bound by a ligand (PD-Ll), which is located on the plasma membrane of the monocyte (antigen-presenting cell), and thus inactivates the T cell.
  • PD-Ll ligand
  • the effect can be intensified if B7 is additionally suppressed in the antigen-presenting cell and, if appropriate, a CTLA4-binding molecule is also introduced.
  • Fig. 4 shows the function of gene manipulation at the molecular level: the autoantigen is synthesized by the PD-Ll producing antigen presenting cells.
  • FIG. 5 and 6 show a comparison of the antigen presentation of "normal" and genetically modified monocytes.
  • Figure 5 shows autoantigen levels presented on the MHC II. Normal monocytes rarely present the autoantigen and if so, only a few MHC II present it. 6 shows that the genetically manipulated monocytes almost exclusively present the autoantigen.
  • FIG. 7a and 7b demonstrate the function of the genetically modified monocytes in the body.
  • natural autoantigen-presenting monocytes induce the production of (pathological) autoantibodies if they are B7-positive.
  • genetically manipulated monocytes compete with the natural autoantigen-presenting monocytes and reduce autoantibody production.
  • the monoantigenic antigen-presenting cell according to the invention is preferably a monocyte, a dendritic cell and / or a macrophage.
  • the APCs are the switching points of the adaptable immune system. Only they can trigger a T cell-mediated immune response. This is shown in the following figures using the example of monocytes and the antibody production triggered by T helper cells: Within the immune system, the antibodies normally play an important role as specific defense molecules ("humoral immune response"). They are produced by mature B lymphocytes. However, the induction and production of soluble antibodies is not independent of the remaining cells in the immune system; rather, this humoral immune response is controlled by other cells in the immune system.
  • the antigen is e.g. a bacterial protein
  • the immune response is useful for the organism.
  • the antigen is an endogenous structure, one speaks of a (pathological) autoimmune reaction.
  • the monoantigenic antigen-presenting cell according to the invention has an increased expression of an antigen, preferably by transfection of an antigen-presenting cell with nucleic acid-containing material, the antigen-presenting cell essentially presenting only predefined antigens.
  • the gene therapy method according to the invention is based on parallel genetic engineering interventions on the patient's own blood monocytes.
  • the interventions are carried out using suitable probes and cause the production of PD 1 binding molecules, preferably PD-L1 and possibly CTLA4 binding molecules, preferably antibodies (not shown), and possibly the reduction of B7 molecules by hindering or preventing the formation of the molecule on the surface of the monocytes (not shown) (Fig. 3) and at the same time a strong presentation of autoantigen (Fig. 4 - 6).
  • pathological autoantibodies is specifically ended by the manipulation of monocytes (or DCs) and the resulting shutdown of antigen-specific T cells. This takes advantage of the fact that T cells produce the receptor PD-1. When bound to PD-Ll, this ensures that T cells no longer proliferate. Binding of PD-1 can thus shut down T cell responses. Binding molecules other than PD-Ll can also be used for this.
  • T cells produce two receptors that recognize B7 (1 or 2).
  • One receptor is CD28
  • the other is CTLA4.
  • CD28 has a stimulating effect, while CTLA4 does not.
  • the contact of CTLA4 with B7 switches off the respective T cell. This then either goes into apoptosis via programmed cell death, is switched off, or is converted into a tolerance-generating T cell. In this case, the tolerance is generated against the specific antigen recognized by this T cell.
  • CTLA4 For binding to CTLA4 instead of CD28, molecules such as antibodies are used which bind to CTLA4 but not to CD28. These can then induce the T cell response promoted by CTLA4. These CTLA4 binding molecules can preferably be antibodies.
  • the co-receptor B7 without which the antigen presentation or the induction cascade for antibody production does not start, can also be suppressed in order to suppress preferential binding of CD28.
  • the co-receptor is two different co-receptors called CD80 (B7-1) and CD86 (B7-2). Their structures are known.
  • the cells After the in vitro manipulation of the monocytes, the cells are returned to the patient's bloodstream.
  • the genetically modified monocytes now switch off the pathological T helper lymphocytes in the organism.
  • the genetically modified cells compete directly with the autoantigen-presenting monocytes already present in the organism (preferably the bloodstream and lymphatic system), which however carry B7 on the cell surface and normally activate the T helper lymphocytes and thus the antibody production (Fig 7a and 7b).
  • the cDNA of a protein that causes the production of pathological autoantibodies as an autoantigen is integrated into the monocyte genome. This genetic information then serves to overproduce the autoantigen. Peptides of this autoantigen are then preferably presented on MHC II and / or MHC I [see also FIGS. 5 and 6]. MHC II presents the peptides to the T helper cells and tries to find those that specifically recognize these presented peptides. If a PD-1 binding molecule activates PD-1 instead of CD28 and a CTLA4-binding molecule also activates CTLA4 instead of CD28 and the co-receptor B7 does not appear on the cell surface at the same time, the T helper cells are shut down and may experience premature cell death.
  • Monocytes present the autoantigen and then only at a few MHC II complexes.
  • the aim of the treatment is to displace the "normal" monocytes which present the autoantigen and activate the T helper cells in vivo by the genetically modified monocytes which have been programmed to switch off the antibody production or T cell response.
  • the monoantigenic antigen-presenting cell according to the invention can in particular show an increased number of homing receptors, such as CD44. Overexpression of homing receptors will show the monoantigenic antigen-presenting cell the way into the lymph nodes, as a result of which the genetically modified APCs increase and accumulate faster in lymph nodes.
  • the lymph nodes are where most of the responses of the adaptive immune system are triggered. There, the genetically modified APCs have a much greater effect than outside the lymph nodes.
  • transfection causes an increase in the number of homing receptors and or an increase in the number of PD-1 binding molecules and / or a suppression of functions of the B7, CD40 receptors and / or an increase in the number of CTLA4 binding molecules.
  • the PD-1 binding molecules can preferably be PD-Ll, PD-L2, antibodies, monoclonal antibodies. These can be such that they remain in the plasma membrane of the cell.
  • the CTLA4 binding molecules can preferably be antibodies, monoclonal antibodies. These can be such that they remain in the plasma membrane of the cell.
  • the B7 and / or CD40 receptor expression in the monoantigenic antigen-presenting cell according to the invention can be prevented or reduced.
  • the expression of the B7 and / or CD40 receptors can be suppressed by nucleic acids by co-suppression in the monoantigenic antigen-presenting cell according to the invention.
  • the monoantigenic antigen-presenting cell according to the invention can contain or be transfected with nucleic acids which bring about an expression of proteins or peptides which have structures affine with B7 and / or CD40 receptors. In this way, proteins are formed which, by forming complexes with B7 and / or CD40 receptors, practically neutralize these receptors.
  • CTLA4, CD28, antibodies, F (ab) 2 , scFv and / or F ab fragments can be considered as proteins.
  • the monoantigenic antigen-presenting cell according to the invention contains nucleic acids which code for a signal sequence or expression products of a signal sequence which determine whether the expression products remain in the endoplasmic reticulum, the Golgi apparatus, the Trans-Golgi network or intracellular vesicles causes.
  • the monoantigenic antigen-presenting cell according to the invention is transfected for the expression of antigens with nucleic acids which enable the transport of the expressed antigens in MHC II compartments of the cells.
  • All genetically modified monocytes present the autoantigen on most of their MHC complexes, while very few "normal” monocytes present the autoantigen at all and then only on a few MHC complexes.
  • the aim of the treatment is to displace the "normal" monocytes that present the autoantigen and activate the T helper cells by means of the genetically manipulated monocytes programmed to switch off the antibody products. It is very important to also administer the antigen (s) in a form that allows them to be transported into the MHC II endosomes. This is the only way to reliably achieve a presentation at MHC II. In the case of a genetic engineering intervention, this is usually done by manipulating the open reading frame, so that the reading frame is preceded by a signal sequence for this compartment. Genetic manipulation has the additional advantage that it has a much longer half-life than, for example, oligo- nucleotides or peptides. A stable integration of genes even leads to a permanent change in the properties of the target cells.
  • the corresponding nucleic acids can be DNA, RNA, oligonucleotides, polynucleotides, ribozymes, peptide nucleic acids (PNA).
  • PNA peptide nucleic acids
  • the DNA preferably has regulatory elements such as enhancers, promoters, polyA-coding 3 "ends for transcribing the DNA into RNA, the RNA regulatory elements for translating the RNA into protein.
  • the regulatory elements ensure efficient expression of the genes.
  • the monoantigenic antigen-presenting cell according to the invention is produced, for example, by ex vivo or in vivo methods.
  • An antigen-presenting cell is preferably ex vivo or in vivo by treatment with viruses, viral vectors, bacterial vectors, plasmids by electroporation techniques, iontophoresis, ballistic methods and / or other techniques for introducing molecules into a monoantigenic antigen-presenting cell transfected.
  • an antigen-presenting cell or a monoantigenic antigen-presenting cell can be obtained by treatment with viruses, viral vectors, bacterial vectors, plasmids, by electroporation techniques, iontophoresis, ballistic methods and / or other techniques for introducing molecules into a cell with an increased amount of PD-1 binding molecules and optionally with an increased amount of CTLA4 binding molecules and possibly suppressed function of co-stimulatory receptors or the expression of co-stimulatory receptors by preventing their expression or the co-stimulatory receptors are reacted with affine structures prevented from stimulating T cells bound to the monoantigenic antigen presenting cell.
  • nucleic acids In this case, antisense nucleic acids must come into contact with the mRNA of the co-stimulatory molecule. You will then likely bind the molecule and prevent translation.
  • RNAs A wide range of molecules, such as RNAs, come as nucleic acids
  • DNAs, PNAs, ribozymes in question.
  • a genetic engineering intervention would ensure the longest half-life of the effect.
  • Re 3. The binding of the co-stimulatory molecule e.g. by specific antibodies prevents this molecule from contacting the notified
  • Receptor takes up on a T cell and thus prevents activation of the T cell.
  • the external addition of such binding molecules has the disadvantage that they act on all antigen-presenting cells and thus prevent any immune reaction.
  • An extension of this model is intended to ensure that the binding molecule is retained in the intracellular compartment so that the co-stimulatory receptor does not reach the plasma membrane in the first place.
  • Signal sequences are known for this, which must be added to the open reading frame of the binding molecule.
  • This intracellular approach can be carried out well on isolated cells.
  • genetic engineering intervention is preferable.
  • the desired effects can also be achieved if only one of the two goals is carried out with a genetic engineering intervention.
  • z. B. the following other molecules are used:
  • Nucleic acids (mostly complementary to the target sequence), which are e.g. B. can be oligonucleotides, polynucleotides, ribozymes, peptide nucleic acids (PNAs),
  • PNAs peptide nucleic acids
  • Antibodies or other molecules that bind the co-stimulatory molecules are included in the immunoglobulin analogs.
  • Proteins, peptides, peptidomimetics Proteins, peptides, peptidomimetics.
  • molecules such as antibodies, proteins, peptides, peptidomimetics, PD-L1, PD-L2, CTLA4, CD28, CD40L and / or constituents and / or combinations of these molecules, which e.g. B7-1, B7-2, CD40 bind, which prevents co-stimulation of the T cell taking place in the presence of an antigen presentation, brought into contact with the monoantigenic antigen presenting cell or the antigen presenting cell.
  • molecules such as liposomes, hydrogels, cyclodextrins, nanocapsules, nanoparticles, in particular biodegradable nanocapsules or particles, bio-adhesive microspheres and / or by electroporation techniques, iontophoresis, ballistic methods and / or other techniques to transfer molecules into the monoantigenic antigen-presenting cell or the antigen-presenting cell.
  • Nucleic acids can in particular by viruses, viral vectors, bacterial vectors, plasmids by electroporation techniques, iontophoresis, ballistic methods and / or other techniques for the introduction of molecules are transferred into the monoantigenic antigen-presenting cell or the antigen-presenting cell.
  • a medicament containing at least one monoantigenic antigen-presenting cell according to the invention is further claimed.
  • the medicament according to the invention is preferably formulated as an infusion solution for intravenous or intraperitoneal administration.
  • the formulation is chosen such that when the medicament is administered there is no significant impairment of the effectiveness of the monoantigenic antigen-presenting cell according to the invention.
  • physiological saline is preferred as the infusion solution.
  • other solutions with a pH of 5.5 to 8.5 are also suitable.
  • Serum for example human serum, autologous serum or serum of other species, solutions with plasma substitutes, such as polyvinylpyrrolidone, are also suitable.
  • plasma substitutes such as polyvinylpyrrolidone
  • 0.5 ml to 500 ml should be applied.
  • the monoantigenic antigen-presenting cell according to the invention can be used in particular for the production of a medicament for the treatment of unwanted immune reactions such as autoimmune diseases and allergies or deliberately induced immune reactions such as in immunizations.
  • the monoantigenic antigen-presenting cell according to the invention can be used for the production of a medicament for the treatment of immune reactions against allologic and / or xenologic tissue characteristics.
  • the immune reactions to be treated are related to antigens or their gene sequences and / or parts thereof and are selected from the group consisting of Enzymes, their gene sequences and / or partial sequences, in particular glutamic acid decarboxylase (GAD), receptor type protein tyrosine phosphatase IA-2Beta, antigen: H + K + ATPase, U1RNP, transglutaminase, argininosuccinate lyase (ASL), tyrosinase related protein-2, thyroid peroxidase, factor VIII, factor IX;
  • GAD glutamic acid decarboxylase
  • IA-2Beta receptor type protein tyrosine phosphatase IA-2Beta
  • antigen H + K + ATPase
  • U1RNP transglutaminase
  • ASL argininosuccinate lyase
  • tyrosinase related protein-2 thyroid peroxidase, factor
  • Receptors their gene sequences and / or partial sequences, in particular acetylcholine receptor of the nicotine type, ⁇ 1-adrenergic receptor, ⁇ l-adrenergic receptor, angiotensin-2-ATl receptor, glutamate receptor, thyrotropin-stimulating hormone (TSH) receptor, LFA receptor 1,
  • HLA-B27 Epididymal Protein DE, Zona Pellucida (ZP) -3 Glycoprotein, Zona Pellucida (ZP) -4 Glycoprotein, Follicle-Stimulating Hormone (FSH) receptor, Sperm Immunogen SP-10 or Sperm Protein SP-10;
  • Hormones or messenger substances their gene sequences and / or partial sequences, in particular insulin, thyroglobulin, follicle-stimulating hormones (FSH), prostaglandin F2 alpha, gonadotropin-releasing hormones (GnRH), oestradiol-17beta, estrogen, luteinizing hormone (LH) receptor, inhibin , Testosterone, androgen, chorionic gonodrophin (CG), interleukins, interferons, cytokines, chemokines, bone morphogenetic factors, ß-interferon, estradiol;
  • Structural proteins their gene sequences and / or partial sequences, in particular myelin basic protein (MBP), proteolipid protein (PLP), myelin oligodendrocyte glycoprotein (MOG), ⁇ -fodrin, non-erythroid ⁇ -
  • MBP myelin basic protein
  • PGP proteolipid protein
  • MOG myelin oligodendrocyte glycoprotein
  • ⁇ -fodrin non-erythroid ⁇ -
  • beta-amyloid precursor protein beta-APP
  • type 2 collagen type 2 collagen
  • sperm plasma membrane protein PH-20 type 2 collagen
  • Antigens their gene sequences and / or partial sequences, in particular CENP-A autoantigens, Beta2GP-I, ribosomal P protein, Ro / SSA,
  • the invention claims a use in which the immune reactions to be treated are associated with allologic and / or xenological tissue characteristics, their gene sequences and / or partial sequences, in particular MHC I, MHC II, rhesus factor.
  • the method presented here for reducing immune responses is based on the genetic manipulation of certain blood cells, preferably outside the body, which are then subsequently returned to the organism.
  • the aim of this treatment is, among other things, the specific switching off of a chronic, immune system-driven production of autoantibodies that trigger the disease.
  • DCM Dilated cardiomyopathy
  • the antibodies play an important role as defense molecules ("humoral immune response"). They are produced by mature B lymphocytes. However, the induction and production of the antibodies is not detached from the remaining cells of the immune system; rather, this humoral immune response is controlled by other cells in the immune system. The immune response cannot be maintained without the help and mediation of antigen presenting cells and T helper lymphocytes.
  • the molecular mechanisms of the interaction - of cell-cell contact - of antigen-presenting cells with the T-helper lymphocytes is known in detail at the receptor level.
  • auxiliary receptors are involved in cell-cell contact and cell activation on the cell surface.
  • An essential molecule of the intercellular interaction in antigen presentation contact of antigen-presenting cells with the T-helper lymphocytes
  • costimulatory receptors with the names PD-Ll, PD-L2, B7h (LICOS), B7-1 and B7-2 , CD40 also plays a role in this signal transduction.
  • the designed procedure is based on at least two parallel interventions on the patient's own antigen-presenting cells.
  • the interventions are carried out using suitable probes and have an effect
  • a PD-1 binding molecule e.g. PD-Ll
  • PD-1 a PD-1 binding molecule which via the action of PD-1, which occurs in T cells, prevents the T cells from activation and proliferation
  • CTLA4 binding molecule e.g. Antibodies that prevent the T cells from activating through the action of CTLA4, which occurs in T cells
  • the cells are returned to the donor's bloodstream.
  • the genetically manipulated monocytes now switch off the corresponding T helper lymphocytes in the organism.
  • the genetically manipulated cells compete directly with the autoantigen-presenting monocytes already present in the organism (preferably the bloodstream and lymphatic system), which however carry B7 and no additional PD-1 binding molecule, CTLA4 binding molecule on the cell surface and usually the T-helper - Activate lymphocytes and, among other things, antibody production.
  • autoimmune diseases and allergies do not exist. With a few exceptions (extracorporeal elimination of antibodies from the patient's blood or plasmapheresis), autoimmune diseases and allergies are treated with medication by inhibiting immune system reactions.
  • Immunosuppressive preparations such as cortisone and its derivatives, as well as cyclosporin, beta interferon or cytostatics (methotrexate)
  • cytostatics metalhotrexate
  • antibodies, antagonists and oligonucleotides against various signal components of the immune system are tested with the purpose of preventing the activation of T cells.
  • Such drugs are at best selective, but never specifically directed against the wrongly programmed autoimmune reactions.
  • Immunosuppressants therefore have a negative effect on important, necessary and useful parts of the immune system; for this reason and their side effects, their use is limited.
  • Boussiotis VA, Freeman, GJ, Gribben, JG, Nadler, LM 1996.
  • ICOS is an inducible T-cell co-stimulator structurally and functionally related to CD28. Nature 397: 263-6.
  • CD28 co-stimulation stabilizes the expression of the CD40 ligand on T cells.
  • PD-L2 is a second ligand for PD-1 and inhibits T cell activation. 2: 261-268.
  • CTLA4 Cytotoxic T lymphocyte antigen 4
  • CTLA-4 ligation suppresses CD28-induced NF-kappaB and AP-1 activity in mouse T cell blasts [published erratum appears in J Biol Chem 1999 Jul 23; 274 (30): 21490]. J Biol Chem. 274: 14400-14405.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Cell Biology (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Mycology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • Hematology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Rheumatology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

L'invention concerne un procédé de diminution de réactions immunitaires spécifiques. Selon ce procédé, des cellules produisant des antigènes sont activées pour présenter des antigènes définis et produire simultanément une molécule de liaison PD-1.
PCT/EP2002/003292 2001-03-27 2002-03-23 Diminution de reactions immunitaires specifiques dependante de certains antigenes par un effet de co-stimulation WO2002077208A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP01107578.5 2001-03-27
EP01107578 2001-03-27

Publications (1)

Publication Number Publication Date
WO2002077208A1 true WO2002077208A1 (fr) 2002-10-03

Family

ID=8176950

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2002/003292 WO2002077208A1 (fr) 2001-03-27 2002-03-23 Diminution de reactions immunitaires specifiques dependante de certains antigenes par un effet de co-stimulation

Country Status (1)

Country Link
WO (1) WO2002077208A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003006636A1 (fr) * 2001-07-12 2003-01-23 Genethor Gmbh Reduction du pouvoir de stimulation de cellules presentant des antigenes
US8217149B2 (en) 2008-12-09 2012-07-10 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
US8652465B2 (en) 2005-06-08 2014-02-18 Emory University Methods and compositions for the treatment of persistent infections
US9598491B2 (en) 2008-11-28 2017-03-21 Emory University Methods for the treatment of infections and tumors
CN114805591A (zh) * 2022-03-31 2022-07-29 浙江大学 靶向ctla-4及配体cd80或cd86的双特异性抗体、筛选方法、组合物及应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000066715A1 (fr) * 1999-05-04 2000-11-09 Genethor Gmbh Procede pour reduire des immunoreactions specifiques
WO2001014557A1 (fr) * 1999-08-23 2001-03-01 Dana-Farber Cancer Institute, Inc. Pd-1, recepteur de b7-4, et son utilisation

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000066715A1 (fr) * 1999-05-04 2000-11-09 Genethor Gmbh Procede pour reduire des immunoreactions specifiques
WO2001014557A1 (fr) * 1999-08-23 2001-03-01 Dana-Farber Cancer Institute, Inc. Pd-1, recepteur de b7-4, et son utilisation

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
AGATA Y ET AL: "EXPRESSION OF THE PD-1 ANTIGEN ON THE SURFACE OF STIMULATED MOUSE T AND B LYMPHOCYTES", INTERNATIONAL IMMUNOLOGY, OXFORD UNIVERSITY PRESS, GB, vol. 8, no. 5, May 1996 (1996-05-01), pages 765 - 772, XP000971773, ISSN: 0953-8178 *
FREEMAN G ET AL: "ENGAGEMENT OF THE PD-1 IMMUNOINHIBITORY RECEPTOR BY A NOVEL B7 FAMILY MEMBER LEADS TO NEGATIVE REGULATION OF LYMPHOCYTE ACTIVATION", JOURNAL OF EXPERIMENTAL MEDICINE, TOKYO, JP, vol. 192, no. 7, 2 October 2000 (2000-10-02), pages 1027 - 1034, XP000971650, ISSN: 0022-1007 *
NISHIMURA H ET AL: "PD-1: an inhibitory immunoreceptor involved in peripheral tolerance", TRENDS IN IMMUNOLOGY, ELSEVIER, CAMBRIDGE, GB, vol. 22, no. 5, 1 May 2001 (2001-05-01), pages 265 - 268, XP004235272, ISSN: 1471-4906 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003006636A1 (fr) * 2001-07-12 2003-01-23 Genethor Gmbh Reduction du pouvoir de stimulation de cellules presentant des antigenes
US8652465B2 (en) 2005-06-08 2014-02-18 Emory University Methods and compositions for the treatment of persistent infections
US9457080B2 (en) 2005-06-08 2016-10-04 Emory University Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 (PD-1) pathway
US10370446B2 (en) 2005-06-08 2019-08-06 Emory University Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 (PD-1) pathway
US11359013B2 (en) 2005-06-08 2022-06-14 Emory University Methods and compositions for the treatment of persistent infections and cancer by inhibiting the programmed cell death 1 (PD-1) pathway
US9598491B2 (en) 2008-11-28 2017-03-21 Emory University Methods for the treatment of infections and tumors
US8217149B2 (en) 2008-12-09 2012-07-10 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
US9920123B2 (en) 2008-12-09 2018-03-20 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
CN114805591A (zh) * 2022-03-31 2022-07-29 浙江大学 靶向ctla-4及配体cd80或cd86的双特异性抗体、筛选方法、组合物及应用

Similar Documents

Publication Publication Date Title
DE69910362T2 (de) Anwendung der mesenchymalen stammzellen als immunsuppressiva
Greenfield et al. CD28/B7 costimulation: a review
Ding et al. IL-10 inhibits macrophage costimulatory activity by selectively inhibiting the up-regulation of B7 expression.
Bour-Jordan et al. CD28 function: a balance of costimulatory and regulatory signals
Lenschow et al. Differential effects of anti-B7-1 and anti-B7-2 monoclonal antibody treatment on the development of diabetes in the nonobese diabetic mouse.
McCoy et al. The role of CTLA‐4 in the regulation of T cell immune responses
DE69519513T2 (de) Verfahren zur modulation von "t-zell-anergy"
DE69533331T2 (de) Liganden zur induktion der antigen-spezifischen apoptose in t-zellen
Cobbold et al. Infectious tolerance
DE69332518T2 (de) Unterdrückung von autoimmunkrankheiten durch antigene in wartestellung
DE69935318T2 (de) Immununterdrückung durch blockierung des t-zellencostimulierungssignales 2 (b7/cd28 interaktion)
DE60313859T2 (de) Induktion der anti-tumor-ctl-immunität durch in-vivo-aktivierung von 4-1bb und/oder cd40
Goleva et al. Our current understanding of checkpoint inhibitor therapy in cancer immunotherapy
US20150232533A1 (en) B7-h1, a novel immunoregulatory molecule
DE69332921T2 (de) Hemmung des Tumorzellwachstums durch Verabreichung von B7-transfizierten Zellen
Gery et al. Autoimmunity in the eye and its regulation
Filippi et al. Transforming growth factor-β suppresses the activation of CD8+ T-cells when naive but promotes their survival and function once antigen experienced: a two-faced impact on autoimmunity
EP0885614A2 (fr) Procédé d'immunisation ex vivo utilisant des anticorps hétérologues intactes bispécifiques ou trispécifiques
DE102004063494A1 (de) Antikörper
DE69535713T2 (de) Verfahren zur modulierung von t-zellreaktionen durch manipulation einer gemeinsamen zytokinrezeptor-gammakette
WO2003006636A1 (fr) Reduction du pouvoir de stimulation de cellules presentant des antigenes
Chapoval et al. Immune checkpoints of the B7 family. Part 1. General characteristics and first representatives: B7-1, B7-2, B7-H1, B7-H2, and B7-DC
WO2002077208A1 (fr) Diminution de reactions immunitaires specifiques dependante de certains antigenes par un effet de co-stimulation
EP1173550B1 (fr) Procede pour reduire des immunoreactions specifiques
DE10038722C2 (de) Verfahren zur Reduzierung von spezifischen Immunreaktionen

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 BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP