WO2004009801A1 - Cellules non presentatrices de l'antigene utilisees pour la selection de lymphocytes t - Google Patents

Cellules non presentatrices de l'antigene utilisees pour la selection de lymphocytes t Download PDF

Info

Publication number
WO2004009801A1
WO2004009801A1 PCT/EP2003/007894 EP0307894W WO2004009801A1 WO 2004009801 A1 WO2004009801 A1 WO 2004009801A1 EP 0307894 W EP0307894 W EP 0307894W WO 2004009801 A1 WO2004009801 A1 WO 2004009801A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
ciita
mhc
molecules
Prior art date
Application number
PCT/EP2003/007894
Other languages
German (de)
English (en)
Inventor
Ahmed Sheriff
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
Priority to AU2003250118A priority Critical patent/AU2003250118A1/en
Publication of WO2004009801A1 publication Critical patent/WO2004009801A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • 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
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • 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
    • A61K39/464Cellular 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
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/26Universal/off- the- shelf cellular immunotherapy; Allogenic cells or means to avoid rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • 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
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/99Coculture with; Conditioned medium produced by genetically modified cells

Definitions

  • the present invention relates to the use of non-antigen-presenting cells for the selection of T cells, methods for producing such cells, and preparation and medicament containing such cells, and use of the cells.
  • CIITA is a transcription factor that activates the MHC II gene locus.
  • APC antigen presenting cell
  • MHC class II Major histocompatibility complexes
  • T cell activation and antigen presentation depend on the amount of MHC-II on individual cells.
  • the regulation and expression of MHC-II genes is important for the control of immune responses.
  • the genes coding for the ⁇ and ⁇ chains of the HLA-DP, HLA-DQ and HLA-DR - class II molecules are grouped on chromosome 6 in the D region of the MHC locus.
  • the genes are subject to a sometimes complex regulatory control. Their expression is generally coordinated and mainly restricted to cells of the immune system, such as B lymphocytes, activated T lymphocytes, macrophages (MQ), dendritic cells (DC) and certain specialized cells such as Kupffer cells and Langerhans cells (Radka, SF, Charron, DJ.6k Brodsky, FM Class II molecules of the major histocompatibility complex considered as differentiation markers.Hum Immunol 16, 390-400. (1986)).
  • lymphokines such as interferon- ⁇ or interleukin 4.
  • Class II molecules are also expressed transiently during the developmental pathway of many haematopoietic cell types (Radka, SF, Charron, DJ. & Brodsky, FM Class II molecules of the major histocompatibility complex considered as differentiation markers. Hum Immunol 16, 390-400. ( 1986)).
  • MHC II genes represent a particularly complex type of regulated gene expression. This regulation affects not only the amount of expression of class II molecules but also the very restricted cell type specificity, since most cells in the body are normally MHC II negative.
  • the complexity of regulation includes two different types of control: constitutive expression in cells such as B cells and inducible expression in certain cell types such as monocytes and fibroblasts.
  • the family of genes includes the ⁇ and ⁇ chains from three different HLA class II isotypes. A number of protein factors can bind to the promoter of the MHC II genes in vitro and in vivo. CIITA is such a factor.
  • CIITA not only regulates the constitutive expression of MHC II genes in cells such as B lymphocytes, but also controls the inducible expression of the same genes in other cell types. It is therefore a factor that is involved in the general control of the MHC II genes.
  • CIITA is a protein with a sequence of 1130 amino acids in length, whose mRNA is only produced in small quantities. When CIITA cDNA is transfected into MHC class II negative cells, these cells begin producing MHC II molecules (Bradley, MB et al. Correction of defective expression in MHC class II deficiency (bare lymphocyte syndrome) cells by retroviral transduction of CIITA. J Immunol 159, 1086-1095. (1997)), (Mori-Aoki, A.
  • the human gene for the MHC class II transactivator is located on chromosome 16 (16P13).
  • the mouse gene for CIITA is located on mouse chromosome 16.
  • the overall intron and exon structure of the mouse MHC II TA gene was determined. It consists of 19 exons, which require a total of 42 kb of the genomic DNA (Reith, W. & Mach, B. The bare lymphocyte syndrome and the regulation of MHC expression. Annu Rev Immunol 19, 331-373 (2001)).
  • the transgenic expression of CIITA can reactivate the expression of all three MHC II isotypes. The same applies to the invariant chain, HLA-DMA and HLA-DMB.
  • the MHC II promoter contains the conserved S, X, X2 and Y sequences.
  • the key transcription factors that control promoter activity are RFX, X2BP and NF-Y. They cooperatively bind to the X, X2 and Y sequences of the promoter and form a very stable nucleoprotein complex. This is called the MHC II enhanceosome.
  • the proteins that bind to the S-Box have not yet been well characterized.
  • CIITA is a non-DNA-binding coactivator that is recruited to the promoter via protein-protein interactions with the DNA-binding components of the enhanceosome (Reith, W. & Mach, B. The bare lymphocyte syndrome and the regulation of MHC expression. Annu Rev Immunol 19, 331-373 (2001)).
  • CIITA activates transcription via an N-terminal transcription activation domain (AD).
  • AD N-terminal transcription activation domain
  • CIITA interacts directly with the RFX-ANK and RFX-5, subunits from RFX, as well as the B and C subunits from NF-Y and with CREB.
  • CIITA is a nuclear protein and contains three regions that appear important for transport to the cell nucleus. The first sits at the C-terminus of CIITA and contains five amino acids that resemble a nuclear localization signal (NLS).
  • the second region is a GTP-binding motif. Binding of GTP also appears to be necessary for the transport of CIITA into the cell nucleus.
  • the third region, called LRR, was also recognized by a mutation analysis as important for the direct transport into the cell nucleus. If CIITA does not get into the cell nucleus, the MHC II locus is not read either (Reith, W. & Mach, B. The bare lymphocyte syndrome and the regulation of MHC expression. Annu Rev Immunol 19, 331-373 (2001)) ,
  • CIITA In contrast to the ubiquitous DNA binding factors (also RFX) that form the MHC II enhososome, the expression of CIITA is very strictly regulated. The expression of CIITA dictates whether and to what extent MHC II genes are expressed. The MHC II TA gene is therefore the main regulator of MHC II expression and therefore also has an essential immunomodulatory role. Most cell types do not form CIITA and are consequently MHC II negative. Expression of CIITA and thus of MHC II can be activated by stimulating such cells with interferon- ⁇ (gamma). This has been demonstrated on many established cell lines.
  • interferon- ⁇ gamma
  • fibroblasts include: fibroblasts, melanoma cells, macrophages, but also some primary cell types, such as mouse embryonic fibroblasts, peritoneal macrophages, microglia and astrocytes.
  • Transfection of MHC II negative cells with CIITA vectors is usually sufficient to induce MHC II expression.
  • a large (> 12 kb) and complex regulatory region contains various independent promoters that control the transcription of CIITA.
  • 4 promoters pl to pIV were identified in the human genes. Three of these p1, pH, pIII and pIV are also highly conserved in the mouse gene. The use of these promoters leads to the synthesis of distinct CIITA mRNAs (types I, III and IV). These contain alternative first exons that are spliced to a conserved second exon.
  • the analysis of the different MHC II-TA promoters has shown that their different activity is crucial for the complex expression pattern of MHC II genes is. Each of them has a specific physiological relevance.
  • pl is very specific for dendritic cells, but pl is not the only promoter active in DCs. Significant transcriptions can also be detected by the plll promoter.
  • the type I transcripts contain an alternative first exon which contains a translation initiation codon which codes for a specific 94 amino acid N-terminal extension of CIITA.
  • pIII is mainly used in B cells.
  • Type III-CIITA mRNA contains an alternative first exon that contains a translation initiation codon and encodes a specific 17 amino acid N-terminal extension.
  • pIV is activated by interferon gamma. Unlike type I and type III, type IV mRNA does not contain a translation initiation codon and therefore does not code for an N-terminal extension.
  • the translation of CIITA is initiated by the first AUG of the second exon (Reith, W. & Mach, B. The bare lymphocyte syndrome and the regulation of MHC expression. Annu Rev Immunol 19, 331-373 (2001)).
  • a problem on which the invention is based is to provide therapeutically usable products for the reduction of allogeneic immune reactions in bone marrow transplants.
  • the problem is solved by the MHC II-producing non-antigen-producing cell (APC) according to the invention.
  • the invention relates to the use of non-antigen-presenting cells which are stimulated to produce MHC II for the ex vivo selection of T lymphocytes.
  • This can be achieved by interferon gamma or transfection with CIITA (CII transactivator).
  • CIITA CII transactivator
  • the induction can be brought about by interferon gamma or the transactivator CIITA, which in turn can be achieved by introducing the gene for CIITA into the target cell.
  • the MHC II-producing cell according to the invention has the advantage that it cannot stimulate potent immune reactions via CD4 + T cells.
  • the switching off of T cells is improved by small amounts of CD80 (B7-1) and CD86 (B7-2), so that these molecules can also be produced in the MHC II-producing cell according to the invention.
  • B7-1 / B7-2 have a 20-50-fold higher affinity for CTLA-4 than for CD28. B7 in small amounts therefore preferentially binds to CTLA-4. In contrast to CD28, CTLA-4 has a tolerogenic effect on T cells. This effect is used.
  • fibroblasts that are not professional APC express MHCII.
  • Fig. 1 Schematic representation of the planned treatment process
  • Fig. 2a The human MHC-II gene locus
  • Fig. 2b The murine MHC-II gene locus
  • Fig. 3 Transcriptional regulation of the murine MHC-II. Cis-acting elements W, X and Y boxes with the transcription factors and the CIITA. All factors together cause the transcription of the MHC-II genes.
  • Fig. 4 Cloned human CIITA in the pcD N A3.1 vector (see also the accompanying diagrams).
  • FIG. 5 shows FACS analysis that the primary human fibroblasts express MHCII after infection with hCIITA. The gene was transferred by lentiviral gene transfer.
  • FIG. 6 shows FACS analysis that the HEK293 cells express MHCII after transfection with hCIITA. It is a selected transgenic cell line.
  • Figure 7 relates to T cell proliferation on artificial APC
  • Figure 8 shows a T cell proliferation assay with artificial APC and control cells
  • FIG. 9 shows a scheme for carrying out an incubation of non-APC with allogeneic cells
  • Fig. 10 shows a plate layout
  • Fig. 13 Cloned murine CIITA in the pcDNA3.1 vector
  • Fig. 16 Proliferated CD4 / CD4CD25 (A) and CD8 T cells (B) after first contact with L929. The individual L929 are shown. T cells without L929 (T cells only) served as controls for background proliferation. The evaluation was carried out at the FACS.
  • Fig. 17 shows a scheme for experiments for the incubation of non-APC with allogeneic T cells.
  • Fig. 18 Proliferated CD4 and CD8 T cells on second contact with allogeneic L929.
  • the use according to the invention can lead, in particular, to the production of a medicament for suppressing immune reactions during transplantations, in particular bone marrow transplantations.
  • the non-AP cell can be selected from the group consisting of fibroblasts, epithelial cells, muscle cells, keratinocytes, hepatocytes, parenchymal cells, chondrocytes and / or melanocytes.
  • Interferon gamma and / or CIITA can cause any cell to produce MHC II. Those that are easy to obtain are the best target cells for the desired treatment.
  • the invention also relates to a method for producing a composition which contains T cells for a recipient, comprising the following steps:
  • the T cells preferably originate from a bone marrow donation or a bone marrow donor, in particular an allogeneic bone marrow donation.
  • the ex vivo selection offers the advantage of rendering alloreactive T cells harmless before contact with the recipient. In this way, only "useful" T cells are infused into the recipient, so that there is no fear of GvHD reactions.
  • a clinically relevant alternative is to remove T cells before infusing the haematopoietic stem cells.
  • this method has the disadvantage that the patients usually develop blood cancer again, since leukaemic cells that have survived radiation and chemotherapy proliferate again. The absence of allogeneic T cells also prevents the useful GvL reactions.
  • the non-AP cell is selected from the group consisting of fibroblasts, epithelial cells, muscle cells, keratinocytes, hepatocytes, parenchymal cells, chondrocytes, and / or melanocytes. It may be advantageous to separate the haematopoietic stem cells from T cells before using the non-AP cell to be used according to the invention.
  • the stem cells can be separated from T cells.
  • T lymphocytes from bone marrow donations are clinical practice because of the risk of GvHD.
  • the cultivation and storage of T lymphocytes is also routine.
  • composition with T cells obtainable by the method according to the invention is also the subject of the invention.
  • a medicament can be produced from the composition according to the invention. This medicament is also the subject of the present invention.
  • the drug is a selected T cell with reduced GvHD potential. These T cells with reduced risk potential can be infused into a recipient. In a leukemia patient, it also pays off that the GvL potential of the allogeneic T cells is still present.
  • the modified non-antigen presenting cells produce MHC II by incubation with interferon gamma (INF ⁇ ) or transfection with CIITA and they may also be transfected with B7-1 and / or B7-2. Then they are incubated with allogeneic bone marrow transplants. These grafts contain lymphocytes. In a preferred embodiment, the cells not presenting antigen can also be incubated alone with the lymphocytes or T cells (T lymphocytes). Among the lymphocytes, the T cells are of particular interest because they trigger important reactions for the recipient / patient. One of the reactions is wanted and useful, the other is harmful and often leads to the death of the recipient.
  • the useful response is the anti-leukemic GvL, the most harmful to the GvHD.
  • Incubation of the cells to be used according to the invention (modified non-AP) with the transplant and here in particular with the T cells occurring therein switches off the GvHD reaction and only maintains the GvL reaction.
  • the non-antigen presenting cells are selected or treated in such a way that they come into contact with T cells via MHC II, but cannot activate them, but still present the recipient's alloantigens.
  • such cells that do not belong to the blood system do not present tumor or cancer antigens because they are not affected by cancer. Therefore, they do not stop immune responses to these antigens.
  • an alloreactive T cell encounters such a cell that does not present an antigen, it is permanently anergic (stopped), or sent to programmed cell death (apoptosis), or differentiated into a regulatory T cell.
  • a regulatory T cell is understood to be a T cell that suppresses immune responses to specific antigens, here alloantigens.
  • a non-antigen-presenting cell that originates from the recipient of the transplant and is characterized in that this cell is provided with CIITA or is caused to produce CIITA is used.
  • CIITA induces the synthesis of MHC II molecules, which in turn can present CD4 + T cell antigens.
  • the T cell is not activated.
  • the T cells are switched off in this way and / or they are driven to programmed cell death (apoptosis). They can differentiate into regulatory T cells if the T cells recognize a presented antigen with their T cell receptor.
  • the antigens recognized by this method are predominantly transplantation antigens [main histocompatibility complexes (HLA, MHC I, MHC II), rhesus factor, Minor histocompatibility antigens], since these differ between donor and recipient of the transplant.
  • transplantation antigens main histocompatibility complexes (HLA, MHC I, MHC II), rhesus factor, Minor histocompatibility antigens
  • the cell is transfected with at least one gene for a CIITA.
  • the cells can be made to produce their MHC II molecules.
  • the cell to be used according to the invention can additionally or alternatively be transfected with CIITA as the protein.
  • CIITA protein the cells can be made to produce their MHC II molecules.
  • the cell to be used according to the invention can also be produced by incubation with INF ⁇ .
  • the cell according to A and / or D to be used according to the invention can additionally be transfected with B7-1 and / or B7-2 in order to force the negative selection of the T cells.
  • the cell according to the invention is incubated in vitro with allogeneic stem cells (eg including lymphocytes) or with allogeneic T cells or with allogeneic lymphocytes from a foreign stem cell transplant donor (eg bone marrow).
  • allogeneic stem cells eg including lymphocytes
  • allogeneic T cells eg including lymphocytes
  • allogeneic lymphocytes from a foreign stem cell transplant donor (eg bone marrow).
  • the named cells (not AP cells), which originate from the recipient of the transplant, cannot contain cancer cells, even in cancer patients with leukemia, since they do not come from the haematopoetic line.
  • they can be treated before incubation with the transplant in such a way that these cells will not proliferate. This is preferably achieved by irradiating the cells.
  • the non-alloreactive but allogeneic T cells positively selected by one of the aforementioned embodiments or any sequence of the same are infused to the patient together with the allogeneic stem cells (eg bone marrow).
  • the allogeneic stem cells eg bone marrow.
  • GvL graft-versus-leukemia effect
  • GvHD significantly reduced graft-versus-host disease effect
  • the methods that can be used make particular use of genetic engineering interventions on the patient's own cells.
  • the interventions are performed using suitable probes and produce a protein that induces MHC II expression, preferably CIITA.
  • Transfection of the non-antigen presenting cells may also result in the production of B7-1 and / or B7-2 CIITA.
  • nucleic acids which code for CIITA, B7-1 and B7-2.
  • the nucleic acids can be DNA, RNA, oligonucleotides, polynucleotides.
  • the DNA preferably contains regulatory elements such as enhancers, promoters, polyA-coding 3 'ends for the transcription of the DNA into RNA.
  • the RNA in turn should contain regulatory elements for translating the RNA into protein.
  • the cells mentioned can be ex vivo in a manner known per se by treatment with viruses, viral vectors, bacterial vectors, plasmids which are suitable for introducing molecules into eukaryotic cells by viral gene transfer, electroporation techniques, iontophoresis, ballistic methods and / or other techniques, be transfected.
  • Said cell can be treated with viruses, viral vectors, bacterial vectors, plasmids by viral gene transfer, electroporation techniques, iontophoresis, ballistic methods and / or other techniques for introducing molecules into a cell with increased production of CIITA and / or B7-1 and / or B7-2 are transfected, whereby T cells which bind to the cell according to the invention via allogeneic antigens, which are presented, for example, on MHC molecules, are switched off.
  • CIITA CIITA, B7-1, B7-2 and / or constituents (proteins, peptides, peptidomimetics) and / or combinations of these molecules can be used as molecules. These molecules hinder stimulation and / or co-stimulation of T cells which take place in the presence of an alloantigen presentation and are brought into contact with the cell according to the invention.
  • the molecules can be introduced by vehicles such as liposomes, hydrogels, cyclodextrins, nanocapsules, nanoparticles, bio-adhesive microspheres and / or by electroporation techniques, iontophoresis, ballistic methods and / or other techniques for introducing Molecules are transferred into the cell according to the invention.
  • vehicles such as liposomes, hydrogels, cyclodextrins, nanocapsules, nanoparticles, bio-adhesive microspheres and / or by electroporation techniques, iontophoresis, ballistic methods and / or other techniques for introducing Molecules are transferred into the cell according to the invention.
  • Nucleic acids can be transferred in particular by viruses, viral vectors, bacterial vectors, plasmids, which are transferred into the non-antigen-presenting cell by electroporation techniques, iontophoresis, ballistic methods and / or other techniques for introducing molecules.
  • cells can be infused into the patient.
  • a medicament containing the preparation according to the invention is therefore also claimed according to the invention.
  • the medicament according to the invention is preferably formulated as an infusion solution for intravenous or intraperitoneal administration.
  • the formulation is chosen so that when the drug is administered there is no significant impairment of the effectiveness of the cell presenting the antigen 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 or allologous 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 cell presenting the antigen to be used according to the invention can be used according to the invention in particular for the production of a medicament for the treatment of immune reactions against allologic tissue features or for the prevention of GvHD.
  • the immune reactions to be treated are related to allologic tissue features, their gene sequences and / or partial sequences, in particular major histocompatibility complexes, MHC I, MHC II, rhesus factor, minor histocompatibility antigens (minor histocompatibility antigens).
  • major histocompatibility complexes MHC I, MHC II, rhesus factor, minor histocompatibility antigens (minor histocompatibility antigens).
  • the ex vivo selected cells can be infused into the patient and then subjected to an in vivo treatment with IL-2.
  • the cells selected ex vivo can also be incubated ex vivo with IL-2 or other stimulants such as phytohemaglutinin (PHA) and then infused into the patient.
  • PHA phytohemaglutinin
  • FIG. 7 HEK293 cells (human embryo kidney) were transfected with plasmids which code for either CIITA or B7-2 or for both genes. After the selection, stable clones expressing MHCII and / or B7-2 were selected and used for a mixed leukocyte reaction (MLR). CFSE-labeled human T cells from different donors were cultured for six days together with irradiated 293 cells. This results in an allogeneic system, since the HEK293 cells carry different histocompatibility antigens (mainly MHC) than the T cells used. However, T cells need a primary stimulus above MHC II and B7 to proliferate. The Proliferation of the T cells was determined by FACS analysis of the decreasing CFSE staining during cell division; the total number of cells was calculated using Truecount Beads. Figure 8 shows a representative experiment.
  • MHC histocompatibility antigens
  • Figure 8 shows a T cell proliferation assay (human) with artificial APC (293 / MHCII-B7.2) and control cells (293, 293-B7-2 and 293-MHCII). 10 5 T cells were incubated with different amounts of artificial APC, control cells or alone for 6 days. The absolute number of proliferated CD4 and CD8 T cells was then determined.
  • CD4 T cells cultured on 293 cells or 293 cells expressing either MHCII or B7-2 show no increased proliferation.
  • the proliferation of CD8 T cells is dose-dependent for all 293 clones used. T cell proliferation was highest on the double positive 293 clones (MHCII and B7-2).
  • T cells which are brought into contact with such artificial APC subsequently increase or decrease allogeneic immune reactions.
  • the T cells were separated from the 293 after the first incubation described above and brought together a second time with different 293 clones.
  • FIG 11 shows CD4 T cell proliferation of T cells matured on double transgenic (MHC II / B7-2) or single positive (MHC II) 293. These T cells were incubated with various 293 clones. The ConA positive control was not shown for the 293-MHC II / B7-2, since this value (142962.9 +/- 11974.3) is far above the others.
  • FIG 12 shows CD8 T cell proliferation of T cells that have been matured to double transgenic (MHC II / B7-2) or single positive (MHC II) 293. These T cells were incubated with various 293 clones. The ConA positive control was not shown for the 293-MHC II / B7-2, since this value (64307.9 +/- 367.7) is far above the others.
  • This experiment shows that neither the CD4 and CDS T cells preincubated on 293 MHC II nor on 293-MHC II / B7-2 are able to carry out immune reactions beyond background proliferation. It can also be seen that T cells which were incubated on first contact with 293MHCII could no longer be stimulated even with ConA.
  • the experiment also showed that the T cells proliferated in all batches. Looking at the CD8 T cells, it was shown that these cells are in all batches proliferated most strongly but not in the presence of MHCII-B7-L929. In the CD4 T cells, on the other hand, proliferation is strongest in the presence of the MHCII-positive L929. Examination of another marker protein (CD25) showed that CD25 + cells were among the proliferated CD4 T cells when the L929 expressed MHCII. The percentage of CD4 + CD25 + T cells proliferated was highest in the MHCII-B7-L929. In the batches with L929 without MHCII, no "new" CD25 + T cells were produced.
  • T cells primed by non-APC should be tested for their proliferation / activation in a second MLR.
  • a reduced activation of L929-CIITA-B7.2, L929-CIITA primed T cells was expected upon contact with L929-CIITA and L929-CIITA-B7.2.
  • L929 and L929-B7.2 primed T cells were included as controls.
  • T cells were prepared from Balb / c spleens 3 days before the initial incubation (primary MLR). After 3 days of cultivation, the T cells were control stained for CD3-APC, CD4-PE, CD8-APC and PL
  • Irradiated non-APC (L929-CIITA, L929-CIITA-B7.2, as controls L929 and L929-B7.2) were treated with Balb / cT cells for three days in the ratio shown in Table 1 in 12-well plates incubated.
  • Tab. 1 Incubation ratios and cell counts / well of a 12-well plate.
  • T cells were carefully removed from the wells and transferred to 6-well plates. There they were cultivated for 2 days (rest phase).
  • non-APC primed T cells were stained with CFSE and again irradiated with non-APC (L929-CIITA, L929-CIITA-B7.2, as controls L929 and L929-B7.2) in the ratio shown in Table 2 in 48-well plates incubated.
  • Tab. 2 Incubation ratios and cell counts / well of a 48-well plate.
  • T cells and non-APC were removed from the wells by pipetting and transferred to Eppis. The cells were then washed with PBA and then taken up in 50 ⁇ l PBA / sample. 0.5 ⁇ l CD4-PE, CD8-APC were added to this approach. Samples were stained on ice for about 30 minutes and then washed with PBA.
  • TruCOUNT beads tubes (bead count: 50267) were filled with 2 ml PBA, vortexed and combined in a 50 ml falcon. The bead suspension was distributed to the samples to be measured with 500 ⁇ l per sample. Shortly before the FACS measurement, 2.5 ⁇ l PI (exclusion of dead cells) were added to the samples, vortexed and measured in the FACS.
  • the total number of cells could be determined on the basis of the known number of beads used and measured per batch.
  • MACS-sorted Balb / c spleen T cells were incubated with artificial, allogeneic APC (L929-CIITA-B7.2, L929-CIITA, L929-B7.2 and L929) in a ratio of 1: 1 for 3 days.
  • APC artificial, allogeneic APC
  • the now primed T cells were stained with CFSE and again combined with the various non-APC in a ratio of 1: 1 for 6 days.
  • the T cell proliferation could be measured by diluting the CFSE with each cell division.
  • Fibroblasts are easily removed from the patient (e.g. from the skin)

Landscapes

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

Abstract

La présente invention concerne l'utilisation de cellules non présentatrices de l'antigène qui sont transfectées avec CIITA (transactivateur CII), pour la sélection ex-vivo de lymphocytes T.
PCT/EP2003/007894 2002-07-19 2003-07-18 Cellules non presentatrices de l'antigene utilisees pour la selection de lymphocytes t WO2004009801A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003250118A AU2003250118A1 (en) 2002-07-19 2003-07-18 Non-antigen presenting cells for selecting t cells

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
DE10233070 2002-07-19
DE10233070.0 2002-07-19
EP02025239.1 2002-11-12
EP02025239 2002-11-12

Publications (1)

Publication Number Publication Date
WO2004009801A1 true WO2004009801A1 (fr) 2004-01-29

Family

ID=30771722

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2003/007894 WO2004009801A1 (fr) 2002-07-19 2003-07-18 Cellules non presentatrices de l'antigene utilisees pour la selection de lymphocytes t

Country Status (2)

Country Link
AU (1) AU2003250118A1 (fr)
WO (1) WO2004009801A1 (fr)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998015626A2 (fr) * 1996-10-08 1998-04-16 Institute Of Child Health Molecule mutante ciita et ses utilisations
EP0648836B1 (fr) * 1993-08-26 2002-01-09 NovImmune SA Transactivateur du complexe majeur d'histomcompatibilité de la classe II et ses utilisations

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0648836B1 (fr) * 1993-08-26 2002-01-09 NovImmune SA Transactivateur du complexe majeur d'histomcompatibilité de la classe II et ses utilisations
WO1998015626A2 (fr) * 1996-10-08 1998-04-16 Institute Of Child Health Molecule mutante ciita et ses utilisations

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
ARMSTRONG TODD D ET AL: "Major histocompatibility complex class II-transfected tumor cells present endogenous antigen and are potent inducers of tumor-specific immunity.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES, vol. 94, no. 13, 1997, 1997, pages 6886 - 6891, XP002224817, ISSN: 0027-8424 *
GUINAN EVA C ET AL: "Transplantation of anergic histoincompatible bone marrow allografts", NEW ENGLAND JOURNAL OF MEDICINE, THE, MASSACHUSETTS MEDICAL SOCIETY, WALTHAM, MA, US, vol. 340, no. 22, 9 June 1999 (1999-06-09), pages 1704 - 1714, XP002187717, ISSN: 0028-4793 *
REITH W ET AL: "MOLECULAR DEFECTS IN THE BARE LYMPHOCYSTE SYNDROM SNF REGULATION OFMHC CLASS II GENES", IMMUNOLOGY TODAY, ELSEVIER PUBLICATIONS, CAMBRIDGE, GB, vol. 16, no. 11, 1995, pages 539 - 545, XP002051564, ISSN: 0167-5699 *
STEIMLE V ET AL: "REGULATION OF MHC CLASS II EXPRESSION BY INTERFERON-GAMMA MEDIATED BY THE TRANSACTIVATOR GENE CIITA", SCIENCE, AMERICAN ASSOCIATION FOR THE ADVANCEMENT OF SCIENCE,, US, vol. 265, no. 5168, 1 July 1994 (1994-07-01), pages 106 - 109, XP002051563, ISSN: 0036-8075 *

Also Published As

Publication number Publication date
AU2003250118A1 (en) 2004-02-09

Similar Documents

Publication Publication Date Title
DE102017127984B4 (de) Verfahren für die Vermehrung und Aktivierung von γδ-T-Zellen
DE69910362T2 (de) Anwendung der mesenchymalen stammzellen als immunsuppressiva
DE69334062T2 (de) In vitro Aktivierung von cytotoxischen T Zellen
DE60218302T2 (de) Verfahren zur gewinnung antigenspezifischer tr1-regualtorischer lymphozyten
DE69737023T2 (de) Methode zur behandlung von krebs und pathogenen infektionen unter verwendung von antigen-präsentierenden zellen beladen mit rna
EP1017723B1 (fr) Polypeptide costimulant de lymphocytes t, anticorps monoclonaux, leur production et leur utilisation
EP0778349A2 (fr) Construction génétique et son utilisation
EP3363892A1 (fr) Procédé d'augmentation de la réactivité immunitaire
DE19722888A1 (de) Human-CD28 spezifische monoklonale Antikörper zur antigenunspezifischen Aktivierung von T-Lymphozyten
DE112019000348B4 (de) Umprogrammierte t-zellen ähnliche nk-zellen
DE69535713T2 (de) Verfahren zur modulierung von t-zellreaktionen durch manipulation einer gemeinsamen zytokinrezeptor-gammakette
EP1331944B1 (fr) Utilisation d'anticorps monoclonaux specifiques a la molecule cd28 en vue de stimuler des cellules sanguines qui ne portent aucune molecule cd28
DE69530793T2 (de) Verfahren des transfers von genen in aktivierten zellen ausgehend von einem zustand des teilungsstillstandes
DE69529473T2 (de) Gezielte t-lymfozyten
EP0693120B1 (fr) Lymphocites t tumoricides
EP1171587B1 (fr) Peptide derive de l'antigene muc-1 destine a declencher une reaction immunitaire contre des cellules tumorales
EP1308167A1 (fr) Vésicles présentatrices d'antigène
DE60130634T2 (de) Identifizierung von antigenen peptiden durch zytotoxische t-lymphozyten aktiviert von dendritischen zellhybriden
DE10313819A1 (de) An MHC-Moleküle bindende Tumor-assoziierte Peptide
WO2004009801A1 (fr) Cellules non presentatrices de l'antigene utilisees pour la selection de lymphocytes t
EP0607593A2 (fr) Procédé d'obtention de cultures cellulaires avec un taux amélioré de cytonines autogènes
EP1381387A2 (fr) Utilisation de cellules mononucleaires du sang peripherique stimulees pour traiter des affections cancereuses
WO2009021995A1 (fr) Procédé d'amplification in vitro de lymphocytes t régulateurs
DE19516673A1 (de) Vakzine gegen Tumorerkrankungen
WO2004047860A2 (fr) Cellules non presentatrices d'antigene pour supprimer des reactions immunitaires pathologiques

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 NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

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

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