WO2005033314A2 - Verfahren zur herstellung eines zell- und/oder gewebe- und/oder krankheitsphasen-spezifischen arzneimittels - Google Patents
Verfahren zur herstellung eines zell- und/oder gewebe- und/oder krankheitsphasen-spezifischen arzneimittels Download PDFInfo
- Publication number
- WO2005033314A2 WO2005033314A2 PCT/DE2004/002197 DE2004002197W WO2005033314A2 WO 2005033314 A2 WO2005033314 A2 WO 2005033314A2 DE 2004002197 W DE2004002197 W DE 2004002197W WO 2005033314 A2 WO2005033314 A2 WO 2005033314A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- mrna
- cell
- dnazyme
- acid molecules
- expression
- Prior art date
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P11/00—Drugs for disorders of the respiratory system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P11/00—Drugs for disorders of the respiratory system
- A61P11/06—Antiasthmatics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P17/00—Drugs for dermatological disorders
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P17/00—Drugs for dermatological disorders
- A61P17/06—Antipsoriatics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
- A61P37/06—Immunosuppressants, e.g. drugs for graft rejection
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/08—Antiallergic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
- A61P9/10—Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
- C12N15/1136—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against growth factors, growth regulators, cytokines, lymphokines or hormones
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/68—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
- C12Q1/6809—Methods for determination or identification of nucleic acids involving differential detection
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/12—Type of nucleic acid catalytic nucleic acids, e.g. ribozymes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/14—Type of nucleic acid interfering N.A.
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/68—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
- C12Q1/6876—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
- C12Q1/6883—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
Definitions
- the present invention relates to a method of producing a cell and / or tissue and / or disease phase specific drug useful in the treatment of chronic inflammation. Background of the invention
- Chronic inflammations represent an increasingly large medical problem area with high socio-economic impact.
- diseases and diseases of the rheumatic type manifestations of, inter alia, skin, lung, kidney, vascular system, nervous system, connective tissue, musculoskeletal system, endocrine system
- COPD Chronic obstructive pulmonary disease
- antigens may be environmental components (eg, allergens such as pollens, animal dander, foods, mites, chemical substances such as preservatives, dyes, detergents).
- allergens such as pollens, animal dander, foods, mites, chemical substances such as preservatives, dyes, detergents.
- COPD chronic obstructive pulmonary diseases
- the immune system can also react against components of one's own organism, recognize them as foreign and initiate an inflammatory reaction. In these cases, an autoimmune disease develops. In any case, harmless, non-toxic antigens are falsely recognized as foreign or dangerous, and an inappropriate inflammatory response is initiated.
- the diseases show patient-specific sub-phenotypic characteristics.
- Components of innate and acquired immunity are sustainably involved in initiation, maintenance, and destruction and remodeling processes. Under the influence of innate immunity (important components: antigen-presenting cells with their various populations and the complement system), activation and differentiation of the cells of the adaptive immune system occur (important components: T and B lymphocytes). The T cells take over central functions in the further course by differentiating into highly specialized effectors.
- effector mechanisms which include in particular the following functions: antibody production, control of the functionality of effector cells of the immune system (such as neutrophil, basophil, eosinophilic granulocytes), feedback on functions of the innate immune system, influence on functionality of non-hematopoietic cells such.
- effector cells of the immune system such as neutrophil, basophil, eosinophilic granulocytes
- feedback on functions of the innate immune system influence on functionality of non-hematopoietic cells such.
- non-hematopoietic cells such as epithelium, endothelium, connective tissue, bone and cartilage and especially neuronal cells.
- the diseases manifest themselves on different organs and compartments.
- the drugs must therefore have a compartment or organ specificity.
- DE 695 11 245 T2 discloses treatment with immunoglobulin A and DE 695 18 667 T2 the inhibition of phospholipase A (PLA 2 ) and / or coenzyme A-independent transacylase (CoA-IT).
- PKA 2 phospholipase A
- CoA-IT coenzyme A-independent transacylase
- Immunosuppressive drugs of the newer generation such as cyclosporin and tacrolimus show hepatotoxicity and nephrotoxicity. This situation has led to the search and clinical testing of a variety of newer molecules that are more specifically designed to interfere with immunological and cell biological dysregulation. These include cytokines, cytokine receptors and anti-cytokines. Problems associated with these newer therapeutic uses include lack of cell and organ specificity, development of unwanted immune responses to these molecules, and lack of potency in various phenotypes.
- DNAzyme Single-stranded molecules that can in principle bind to complementary regions of the RNA and inactivate them by cleavage.
- DNAzymes are single-stranded molecules that can in principle bind to complementary regions of the RNA and inactivate them by cleavage.
- the specific use of DNAzymes as therapeutic agents requires, however, that the disease-causing genes and their mRNA are known in detail.
- WO 01/11023A1 binds RelA (p65) mRNA and is thus directed against the transcription factor NF- ⁇ B, in WO 00/42173 an EGR-1 mRNA-binding DNAzyme is disclosed.
- WO99 / 50452 discloses a 10-23 DNAzyme which can be used in a diagnostic method for finding nucleic acid mutations.
- None of the currently known antisense molecules and DNAzymes can be used for the preparation of a medicament for the treatment of chronic inflammation in patients.
- the object of the present invention is to provide cell and / or tissue and / or disease phase-specific drugs which are involved in the functional inactivation of ribonucleic acid molecules of transcription factors and factors of the signal transduction pathways whose expression is involved in the development of chronic inflammatory reactions and autoimmune diseases . lead and are suitable for the treatment of chronic inflammatory reactions and Au ⁇ toimmunfixen, the disadvantages are eliminated in the prior art.
- the object is achieved by a method according to claim 1 and a medicament according to claims 16 to 18 using specific DNAzyme according to claims 10 to 15.
- the advantage of the invention is a functional inactivation of ribonucleic acid molecules of transcription factors and factors of the signal transduction pathways for the differentiation and / or expression of cytokines involved in the development of chronic inflammatory reactions and autoimmune diseases by means of specific DNAzymes and / or siRNA.
- This strategy is distinguished from conventional as well as gene therapy approaches by highest cell and / or tissue and / or disease phase specificity and selectivity, high stability of the molecules and a negligible antigenicity.
- Optimal conditions for tailor-made long-term therapy in patients with chronic inflammatory diseases are created.
- FIG. 2 Nucleotide sequence of the catalytic domain of the 10-23 DNAzyme and binding to a target RNA by means of Watson-Crick pairing.
- the arrow shows the cleavage site in the target mRNA.
- FIG. 4 Nucleotide sequences of human GATA-3 genes in alignment sequence 1: human GATA-3 from database no .: XM_043124. Sequence 2: Human GATA-3 from database no .: X58072. Sequence 3: Human GATA-3 (sequenced from plasmid pCR2.1). Divergent bases are highlighted in gray, primer localizations for the cloning of GATA-3 are underlined. The localization of the DNAzyme hgd40 is illustrated by bold letters, which are grayed out and underlined at the same time.
- FIG. 4 A nucleotide sequence 3 of the human GATA-3 gene from FIG. 4, inscribed therein (shaded gray) in each case the nucleotide pairs GT and AT, between see which other DNAzyme interfaces are.
- Fig. 5 Gel electrophoresis shows the cleavage of a target mRNA (here GATA-3 mRNA) with specific ribonucleic acid molecules after step b), here unmodified DNAzyme [hgdl 1 (lane 2), hgd13 (lane 4), hgd17 (lane 6 ), hgd40 (lane 8)] and modified DNAzymes [hgdl 1-M (lane 3), hgd13-M (lane 5), hgd17-M (lane 7), hgd40-M (lane 9)].
- M denotes the modified DNAzyme.
- Unmodified (0.25 ⁇ M) or modified DNAzymes (0.25 ⁇ M) are incubated for one hour at 37 ° C.
- Lane 1 contains mRNA without DNAzyme addition as a control. Included length standard (not shown) shows band sizes of 1000 bp, 2000 bp and 3000 bp. Arrows point to S, the band with the substrate (here GATA-3 mRNA) and the cleavage products P1 and P2.
- Fig. 6 Immunoblot with the reaction of specific ribonucleic acid molecules in cells.
- Jurkat E6.1 cells are isolated by lipofection with specific ribonucleic acid molecules, here DNAzyme [hgdl 1-M (lane 4), hgd13-M (lane 5), hgd17-M (lane 6), hgd40-M (lane 7). ] transfected.
- the controls used are untreated cells (lane 1), cells treated only with transfection medium (lane 2), or cells treated with DNAzymes (hgd11-M) without transfection medium (lane 3). After 48 h of incubation, the solubilized proteins are separated by SDS-PAGE and GATA-3 (A) detected by immunoblotting with specific antibodies.
- FIG. 8 Nucleotide sequences of human T-bet genes in alignment
- Sequence 1 Human T-bet from database no .: NM_013351.
- T-bet mRNA in vitro transcribed T-bet mRNA (0.025 ⁇ M) in a volume of 10 ⁇ l with the following reaction composition: 50 mM Tris pH 7.4, 150 mM NaCl, 10 mM MgCl 2, then the products are separated by gel electrophoresis, lane M has a length standard of 3000 bases and 2000 bases, lane 2 contains control mRNA without DNAzyme addition, arrow A points to the band with substrate (here T-bet the second cleavage product is smaller and no longer visible in this figure Fig.
- Fig. 11 Diagram of the relative quantification of T-bet mRNA in Jurkat E6.1 cells.
- Jurkat E6.1 cells are transfected twice with the T-bet-specific DNAzymes td54, td69 and td70 and RNA is isolated after 48 h. After reverse transcription, the amount of mRNA is determined by means of LightCycler. The control is nonsense DNAzyme.
- the relative quantification of T-bet and GAPDH mRNA is carried out according to instructions [described in User Bulletin # 2 (ABI Prism 7700 Sequence detection System User Bulletin # 2 (2001). Relative quantification of gene expression. Http://docs.appliedbiosystems.com/pebiodocs/04303859.pdf)].
- the amount of T-bet mRNA from the control experiment with nonsense DNAzyme is set to 100%.
- FIG. 1 shows, in a schematic representation modified according to Ho IC and Glimcher LH (Cell 2002; 109: S109-S120), the correlations of signal transduction in the differentiation of CD4 + cells into TH1 and TH2 cells, respectively.
- Stimulation via the T cell receptor by the corresponding peptide-MHC complex induces clonal expansion and programmed differentiation of CD4 + T lymphocytes into T helper (TH) 1 or TH2 cells.
- TH1 cells produce interferon- ⁇ (INF ⁇ ), interleukin 2 (IL-2) and tumor necrosis factor- ⁇ , whereas TH2 cells secrete IL-4, IL-5, IL-9 and IL-13.
- INF ⁇ interferon- ⁇
- IL-2 interleukin 2
- TH2 cells secrete IL-4, IL-5, IL-9 and IL-13.
- TH1 cells Bacterial and viral infections induce an immune response that is dominated by TH1 cells.
- TH2 cells regulate IgE production against parasites. There is an equilibrium between TH1 and TH2 cells. Disruption of this balance causes disease, an excess TH1 cell response is associated with autoimmune disease, while allergic disease is based on an enhanced TH2 cell response.
- TH1 cytokines are involved in the pathogenesis of autoimmune diseases such as autoimmune uveitis, experimental allergic encephalomyelitis, type 1 diabetes mellitus or Crohn's disease, while TH2 cytokines (IL-4, IL-5, IL-13 and IL -9) are involved in the development of chronic inflammatory respiratory diseases such as respiratory eosinophilia, mucus hypersecretion and respiratory hyperresponsiveness.
- IL-4, IL-5, IL-13 and IL -9 are involved in the development of chronic inflammatory respiratory diseases such as respiratory eosinophilia, mucus hypersecretion and respiratory hyperresponsiveness.
- transgenic mice that constitutively overexpress the TH2 cytokines IL-4, IL-5, IL-13 and IL-9 in the respiratory epithelium show typical allergic inflammatory reactions.
- TH2 cell subpopulations in the lung and respiratory tract cause the characteristic symptoms of bronchial asthma in TH2 cells in animal models.
- transcription factors and factors of signal transduction pathways for differentiation and / or expression of cytokines involved in the development of chronic inflammatory reactions and autoimmune diseases such as: the TH1 Cell-specific transcription factor T-bet and the TH2 cell-specific transcription factor GATA-3 are ideally suited.
- the TH1 cell-specific transcription factor T-bet is mainly responsible for the differentiation of na ⁇ ve CD + T cells into TH1 cells. Its expression is controlled via the signal transduction pathways of the T cell receptor (TCR) and via INF receptor / STAT1. T-bet transactivates the endogenous INF ⁇ gene and induces INF ⁇ production. In addition, it induces the up-regulation of protein expression of IL-12Rß2 chains and leads to chromatin remodeling of individual INF ⁇ alleles. The in vivo function of T-bet is confirmed in knockout mice (T-bet ";" ).
- T-bet deficient mice have normal lymphocyte development, CD4 + T cells from these mice do not produce INF ⁇ , either anti-CD3 / CD28 or PMA / ionomycin stimulation. T-bet deficient mice show no immune response to L major infection, the amount of TH2 cytokines is increased.
- T-bet in mucosal T cells in the development of inflammatory bowel disease.
- SCID severe Combined Immunodeficiency mice after retroviral transduction of T-bet in CD4 + CD26L + T cells, conversely, the transfer of T-bet deficient T cells does not lead to colitis -Induction.
- the transcription factor T-bet specifically induces the development of TH1 cells and controls INF ⁇ production in these cells. Inhibition of T-bet shifts the balance between TH1 and TH2 cells in favor of TH2 cells.
- the TH2 cell-specific transcription factor GATA-3 is mainly responsible for the differentiation of na ⁇ ve CD4 + T cells into TH2 cells. Th2 cell differentiation is mainly controlled by two signal transduction pathways, the T cell receptor (TCR) and the IL-4 receptor pathway. Signals forwarded by the TCR activate both the TH2 cell-specific transcription factors c-Maf and GATA-3 and the transcription factors NFAT and AP-1. Activation of the IL-4 receptor leads to the binding of STAT6 to the cytoplasmic domain of the IL-4 receptor, where it is phosphorylated by Jak1 and Jak3 kinases. In turn, phosphorylation leads to the dimerization and translocation of STAT6 into the nucleus, where STAT6 activates transcription of GATA-3 and other genes. GATA-3 is a zinc finger transcription factor, which according to "Representational
- RIBP RaBP
- PLCY phospholipase C ⁇ 1
- MAP kinase mitogen-activated protein kinase
- ERK JNK P38 • MKK (MAP kinase kinase) MKK1 MKK2 MKK3 MKK4 MKK6 MKK7 • Rac2 • GADD45 (Growth arrest and DNA damage gene 45)
- GADD45ß GADD45 ⁇ SOCS cytokine-induced SH2 protein
- SOCS1 SOCS2 SOCS3 JAK JAK1 JAK3 NIP45 (NF-AT interacting protein)
- a cell and / or tissue and / or disease phase-specific drug is provided which is suitable for the treatment of chronic inflammation.
- the drug preferably attacks at the intervention points of the complex cascade of immunological and cell biological dysregulations underlying the chronic inflammatory reactions and autoimmune diseases. These are particularly preferred intervention points for the regulation of the differentiation of the transcription factors involved, such as, for example, the TH2 cell-specific transcription factor GATA-3 or the TH1 cell-specific transcription factor T-bet.
- the achieved therapeutic effect consists in a functional inactivation of mRNA molecules by means of specific DNAzyme and / or siRNA.
- a method of producing a cell and / or tissue and / or disease phase specific drug comprising the steps of: a) identifying ribonucleic acid molecules whose expression differs in a target cell compared to expression of a control cell b ) Design of specific ribonucleic acid molecules which bind to ribonucleic acid molecules from step a) and functionally inactivate them c) introduction of the specific ribonucleic acid molecules from step b) into target cells d) formulation of the specific ribonucleic acid molecules from step b) and / or a target cell from step c) in a drug
- the term "cell and / or tissue and / or disease phase-specific” means that the medicament produced by means of the method according to the invention is essentially only present in a specific type of cell (target cell) and / or in is effective in certain tissues or organs and / or at certain stages of the disease, and has negligible influence on other cells (control cells), tissues or organs
- the drug is active in at least 2/3 of the target cells It is also preferred that the drug be effective at at most 10% of the control cells, more preferably at most 5%, and most preferably at ⁇ 1% of the control cells.
- Target cells are cells in tissues and organs known to cause disease contributing to, or reinforcing, contributing to, contributing to, or enhancing the processes that sustain the disease, or contributing to, or exacerbating, the long-term effects of a disease, including, for example, cells carrying certain transcriptional
- target cells may be isolated by technologies based on specific antibody binding.
- target cells may be isolated by technologies that are specific for the binding of specific antibodies.
- Magnetic beads available from Miltenyi (Macs system), Dynal (DynaBeads) or BD-Bioscience (iMAG), are used here. Alternatively, this is done by cell purification by means of fluorescence-labeled antibodies at cell sites, for example from the company Cy- tomation (MOFLO) or BD-Bioscience (FACS-Vantage). Len The purity of the Zielzel ⁇ is preferably at least 80%, more preferably at least 95% and most preferably at least 99%.
- mRNAdirect it is preferred to directly purify mRNA from the target cells by using commercial kits of, for example, the companies Qiagen (Oligotex mRNA Kit), Promega (PolyATract mRNA Isolation System) or Miltenyi (mRNAdirect).
- Qiagen Oligonucleic acid Kit
- Promega PolyATract mRNA Isolation System
- Miltenyi Miltenyi
- mRNAdirect Miltenyi
- Examples of these methods known to those skilled in the art include the SSH method (Clontech) or the RDA method.
- Another preferred application is the combination of chip technology and subtractive hybridization.
- the identification of the differentially expressed genes is carried out using the chip technology with the aid of commercially available programs, for example with the Vector Xpression program of the company InforMax.
- subtractive hybridization is used, the differentially expressed genes are isolated by conventional techniques known to those skilled in the art.
- Expression in the target cell differs from expression in a control cell.
- the expression in the target cell is increased in comparison to the expression in a control cell, preferably at least by a factor of 1.5.
- expression in the target cell is increased by at least a factor of 5 compared to expression in a control cell, and in a most preferred embodiment, expression is detectable only in the target cell but not in the control cell.
- the term "design of ribonucleic acid molecules which bind to ribonucleic acid molecules from step a) and functionally inactivate them” comprises the use of RNA-inactivating DNA enzymes (DNAzymes) and / or small-interfering RNA ( siRNA), which functionally inactivate ribonucleic acid molecules according to the invention
- a general DNAzyme model represents the "10-23" model.
- DNAzymes of the 10-23 model - also referred to as "10-23 DNAzymes" - possess a catalytic domain of 15 deoxyribonucleic acids flanked by two substrate binding domains.
- the length of the substrate binding domains is variable, they are either the same or different lengths. In a preferred embodiment, the length of the substrate binding domains is between 6 and 14 nucleotides. In a particularly preferred embodiment, the substrate binding domains are fully complementary to the region flanking the cleavage site. However, to bind and cleave the target RNA, the DNAzyme need not necessarily be completely complementary.
- a preferred embodiment is the introduction of a 3 ' -3 ' inversion at one or more ends of the DNA enzyme.
- the term 3 ' -3 ' inversion refers to a covalent phosphate bond between the 3 'carbons of the terminal nucleotide and the adjacent nucleotide.
- the nucleotide be inverse at the 3 ' end of the substrate binding domain adjacent the 3' end of the catalytic domain.
- the DNAzymes may contain modified nucleotides or nucleotide compounds. Modified nucleotides include, for example, N3 ' ⁇ P5' phosphoramidate compounds, 2 '-O-methyl substitutions and peptide nucleic acid compounds. Their preparation is familiar to the expert.
- siRNA according to the invention comprises double-stranded, 21-23 base long RNA molecules, which lead to a specific degradation of the complementary target mRNAs both in vitro and in vivo. It is known to the person skilled in the art from the literature (for example http://www.mpibpc.gwdg.de/ en/100/105/index.html) to produce siRNA molecules starting from the target mRNA sequence.
- siRNA molecules The likelihood of having at least one highly active (inhibiting target RNA of at least 80%) among three selected siRNA molecules is reported to be at least 70% in the literature. It will be from a pool selected on siRNA molecules those that lead to a specific Degenera ⁇ tion of the complementary target mRNA both in vitro and in vivo.
- the term "introduction of the specific ribonucleic acid molecules from step b) into target cells” comprises the transfection into the target cells of vectors, in particular plasmids, cosmids, viruses or bacteriophages, which contain the above-described specific ribonucleic acid molecules according to the invention
- the vectors are suitable for the transformation of animal and human cells and allow the integration of the ribonucleic acid molecules according to the invention.
- Methods for transfection such as lipofection by means of Invitrogen's DMRIE-C are known to the person skilled in the art from liposomal vectors are transcription factors, cells that secrete hormones, cytokines, and growth factors, as well as cells that carry on the surface of the expressed receptors.
- control cells As control cells according to the invention, healthy cells of the target tissue, cells of the same type from other compartments of the same patient or also from healthy individuals are used.
- the cultivation of the target cell is carried out in nutrient media, which are adapted to the needs of the target cell according to pH, temperature, salt concentration, antibiotics, vitamins, trace elements and ventilation.
- nutrient media which are adapted to the needs of the target cell according to pH, temperature, salt concentration, antibiotics, vitamins, trace elements and ventilation.
- patient refers equally to humans and vertebrates.
- the drug can be used in human and veterinary medicine.
- formulation of the specific ribonucleic acid molecules of step b) or a target cell of step c) in a pharmaceutical composition includes pharmaceutically acceptable compositions that include modifications and “prodrugs” provided that they do not show undue toxicity, irritation, or damage to a reliable medical judgment allergic reactions in the patient
- prodrug refers to compounds that are transformed to enhance uptake, such as by hydrolysis in the blood.
- the formulation that the specific Ribonuk ⁇ leincher molecules the patients in the form of a pharmaceutically acceptable composition either orally, rectally, parenterally, intravenously, intramuscularly or sub ⁇ cutaneous, intracistemal, intravaginal, intraperitoneal, intrathecal, intravascularly, locally (powders, Ointment or drops) or in spray form.
- Dosage forms for topical administration of the drug of this invention include ointments, powders, sprays or inhalants.
- the active component is mixed under sterile conditions with a physiologically acceptable carrier and possible preservatives, buffers or propellants, as needed.
- the type of dosage is determined by the attending physician according to the clinical factors. It is known to the person skilled in the art that the type of dosage depends on various factors, such as e.g. Body size, weight, body surface area, age, sex or general health of the patient, but also on the specific agent to be administered, the duration and route of administration and other medications that may be administered in parallel.
- the pharmaceutical composition produced by the method according to the invention has a high patient, disease, stage or phase specificity. It causes a cell-specific intervention and is specific to compartments and organs. There are no or very few reactions of the immune system to the drug, and the adverse event profile is in an acceptable proportion to the severity, prognosis and course of the disease.
- the drug can be used to treat all disease groups associated with chronic inflammation, such as autoimmune diseases, rheumatic diseases (manifestations of, inter alia, skin, lung, kidney, vascular system, nervous system, connective tissue, musculoskeletal system, endocrine system), allergic immediate type reactions and asthma.
- Chronic obstructive pulmonary disease (COPD) arteriosclerosis, psoriasis and contact dermatitis, as well as chronic organ rejection and bone marrow transplantation.
- CD4 + target cells are isolated by magnetic beads (Miltenyi (Macs system) Dynal (DynaBeads) or BD-Bioscience (iMAG), alternatively by fluorescence-labeled antibodies to cell sorters, for example from the companies CytoMation (MOFLO) or BD Bioscience (FACS-Vantage)
- MOFLO CytoMation
- BD Bioscience FACS-Vantage
- RNeasy kit from Qiagen is used, or there is direct isolation of the mRNA from CD4 + target cells with Oligotex mRNA kit from Qiagen companies according to the manufacturer's instructions.
- Oligotex mRNA kit from Qiagen companies according to the manufacturer's instructions.
- GATA-3 differs in the target cell (TH2 cell) compared to expression in a control cell (for example, ThO cell).
- FIG. 3 shows the inventive pool hgd 1 to hgd 70 at specific DNAzymes against GATA-3 mRNA.
- the DNAzymes have a total length of 33 nucleotides, the central catalytic domain corresponding to 15 nucleotides (in lower case letters) of the catalytic domain of the known 10-23 DNAzyme (FIG. 2).
- This catalytic domain is formed by two right and left substrate binding domains, each consisting of 9 nucleotides
- the nucleotide sequence of the right and left substrate binding domain is different and varies in the DNA enzymes hgd 1 to hgd 70, so that a different specific binding by means of Watson-Crick pairing to the GATA-3 mRNA takes place.
- Figure 2 shows the general model for binding the 10-23 DNAzyme to an N-labeled any target RNA, the arrow indicating the cleavage site in the target mRNA.
- DNAzymes can cleave the target mRNA on any purine-pyrimidine sequence, it is known from the literature that purine-uracil bonds are more efficiently cleaved than purine-cytosine bonds. Therefore, DNAzymes that cleave at purine-uracil bonds are preferably constructed.
- the model shown in FIG. 2 can be transferred in its mode of operation to the binding of DNAzyme hgd 1 to hgd 70 to GATA-3 mRNA.
- the DNAzymes hgd 1 to hgd 70 are used unmodified for in vitro experiments, modified for experiments in cell culture (purchased from Eurogentec). As modifications for stabilization and protection are used: 1) a stabilizing inverse thymidine at the 3 ' end 2) a FAM label at the 5 ' end to assess the transfection efficiency of the cells by FACS analysis. In order to test the DNAzyme in vitro, GATA-3 mRNA is required, which is produced by in vitro transcription. The individual steps are as follows: RNA isolation from human EDTA whole blood using QIAamp RNA Blood Mini Kit (Qiagen, Germany) according to the manufacturer's instructions reverse transcription with the primers: forward primer GGCGCCGTCTTGATACTTT
- Reverse primer CCGAAAATTGAGAGAAGGAA whereby a PCR product with a length of 2731 nucleotides is amplified (JumpStart Accu Taq DNA polymerase, Sigma).
- PCR conditions Initial denaturation (96 ° C, 30 sec) Amplification with 40 cycles (94 ° C, 15 sec, 48 ° C, 30 sec, 68 ° C, 3 min), Final Extention (68 ° C , 30 min).
- the PCR product is cloned into the plasmid pCR2.1 (Invitrogen) by standard methods and sequenced for verification.
- the preparation of GATA-3 mRNA is carried out after linearization of the plasmid pCR2.1 containing GATA-3 by cleavage with the restriction enzyme Spe I by in vitro transcription according to the manufacturer (Ambion). GATA-3 mRNA is present with a total length of 2876 nucleotides.
- Sequence 1 Human GATA-3 from database #: XM_043124
- Sequence 2 Human GATA-3 from database #: X58072
- Sequence 3 Human GATA-3 (isolated from plasmid pCR2.1).
- the GATA-3 mRNA sequences differ in length from the 3 ' untranslated or 5 ' untranslated ends.
- the mRNA sequences of the entries no .: XM_043124 and X58072 are used for primer selection.
- the primer localizations for the cloning of GATA-3 are highlighted in Figure 4 as underlining.
- FIG. 4 further shows an alignment of the nucleic acid sequence of GATA-3 from the database (sequence 1 and 2) with the nucleotide sequence sequenced from plasmid pCR2.1 (sequence 3). It turns out that the sequences not completely identical, but individual bases are different.
- the nucleic acid sequence 3 of GATA-3 from FIG. 4 forms the basis for the construction of DNAzymes against GATA-3 mRNA.
- FIG. 4 A shows the nucleotide sequence of the sequence 3 of the human GATA-3 gene from FIG. 4 and shows in each case two nucleotides GT or AT as gray background, between which there are further potential interfaces for DNAzymes.
- the in vitro cleavage experiments of GATA-3 mRNA with the DNAzymes are carried out in a volume of 10 ⁇ l of the following reaction composition: 50 mM Tris pH 7.4, 150 mM NaCl, 10 mM MgCl 2 , 0.25 ⁇ M DNAzyme and 0.025 ⁇ M in vitro transcribed GATA-3 mRNA (in a substrate to DNAzyme ratio of 1:10).
- the reactions are incubated at 37 ° C for the indicated times.
- formamide- and EDTA-containing RNA sample loading buffer stops the reaction.
- the denatured samples are separated in 1.3% TAE agarose gels and analyzed in the UV transilluminator.
- Lane 5 shows the cleavage of the GATA-3 target mRNA with unmodified DNAzyme [hgdl 1 (lane 2), hgdl 3 (lane 4), hgdl 7 (lane 6), hgd40 (lane 8)] and modified as a result of gel electrophoresis DNAzymes [hgdl 1-M (lane 3), hgd13-M (lane 5), hgd17-M (lane 7), hgd40-M (lane 9)].
- Lane 1 contains mRNA without DNAzyme addition as a control.
- the modified DNAzymes are labeled with an additional M.
- An entrained length standard shows band sizes of 1000 bp, 2000 bp and 3000 bp. Arrows point to S, the band with the substrate (here GATA-3 mRNA) and the cleavage products P1 and P2.
- the following table shows the classification of the DNAzyme hgd 1 to hgd 70 against GATA-3 mRNA into 4 groups. This grouping is done on the basis of led in vitro activity tests of the DNAzyme against GATA-3 mRNA.
- Group 1 high cleavage activity
- Group 2 medium cleavage activity
- Group 3 weak cleavage activity
- Group 4 no measurable cleavage activity.
- Jurkat E6.1 cells human acute cell leukemia Cells
- RPMI medium with 100 U / ml penicillin, 0.1 mg / ml streptomycin and 10% FCS at 37 ° C in humidified 5% C0 2 atmosphere cultured.
- the transfections are carried out in 6-well plates.
- 2x10 6 Jurkat E6.1 cells are transferred into Opti-MEM-I cell culture medium (Invitrogen) and transfected with the modified DNAzymes (0.3 ⁇ M) by means of DMRIE-C (Invitrogen) (according to the manufacturer's instructions from Invitrogen).
- DMRIE-C Invitrogen
- RPMI medium (with the additions indicated above) is added and incubation is continued for an additional 14 hours.
- the cells are washed with OptiMEM medium and then transfected again according to the protocol described above. After each transfection, the transfection efficiency is assessed by FACS analysis. Subsequently, the activity of the DNAzyme is checked by detecting the amount of GATA-3 protein in the Western blot (see FIG. 6).
- the cytoplasmic proteins and the nuclear proteins are processed separately by means of a protein extraction kit according to the manufacturer's instructions (Pierce).
- the protein concentration is determined with the BCA kit (Pierce).
- the separation of each 30 ug protein by denaturing gel electrophoresis in 10% SDS-polyacrylamide gels.
- the proteins are then blotted onto nitrocellulose membranes by standard techniques.
- the membranes are blocked with 5% skimmed milk powder in PBS (with 0.01% Tween 20) and then with mouse anti-GATA-3 antibodies (Santa Cruz) (1: 500) and subsequently with HRP-coupled mouse anti-GATA-3 antibodies.
- Rabbit antibodies (BD Biosciences) (1: 2000) for one hour at room temperature.
- the proteins are visualized by chemiluminescence.
- the parallel detection of beta-actin on the blots controls variations in the amount of protein applied.
- GA-TA-3 is first detected on the nitrocellulose membrane. Subsequently, the same membrane is left overnight in a humid chamber. After washing twice with PBS, ⁇ -actin is detected by immunostaining with specific antibodies (mouse anti-human beta-actin antibody (Sigma)).
- FIG. 6 shows the result of the immunoblot showing the activity of the DNAzyme in cells.
- the controls used are untreated cells (lane 1), transfection medium only (lane 2), or DNAzymes without transfection medium (lane 3). After 48 h of incubation, the solubilized proteins are separated by SDS-PAGE and GATA-3 (A) detected by immunoblotting with specific antibodies.
- DNAzyme hgd40 specifically inhibits GATA-3 expression in vivo and is useful as a specific ribonucleic acid for the production of a cell and / or tissue and / or disease phase specific drug .
- hgd40 5 ' ⁇ GTGGATGGAggctagctacaacgaGTCTTGGAG
- cells transfected with hgd40 are provided in a pharmaceutical composition with a pharmaceutically acceptable carrier, for example liposomes or biodegradable polymers.
- a pharmaceutically acceptable carrier for example liposomes or biodegradable polymers.
- siRNA is used for the specific inhibition of GATA-3 expression and for the production of a cell and / or tissue and / or disease phase-specific drug.
- siRNA is used to inhibit mouse and human GATA-3.
- the preparation of the siRNA is known to the person skilled in the art and described in the literature. Examples of siRNA sequences are listed below:
- Example 2 DNAzymes against T-bet a) Identification of ribonucleic acid molecules whose expression in a target cell differs compared to the expression of a control cell
- T-bet in the target cell differs in comparison to the expression in a control cell (ThO cell).
- step b) Design of specific ribonucleic acid molecules that bind to ribonucleic acid molecules from step a) and functionally inactivate them
- FIG. 7 shows the pool td1-td78 according to the invention of specific DNAzymes for T-bet mRNA.
- the DNAzymes have a total length of 33 nucleotides, with the central catalytic domain of 15 nucleotides (in lowercase letters) corresponding to the catalytic domain of the known 10-23 DNAzyme (FIG. 2).
- This catalytic domain is flanked by two 9-nucleotide right and left substrate binding domains (in capital letters).
- the nucleotide sequence of the right and left substrate binding domains is different and varies in DNAzymes td1 to td78, so that a different specific binding by means of Watson-Crick mating to the T-bet mRNA takes place.
- DNAzymes are preferably constructed which cleave to purine-uracil bonds.
- the model shown in FIG. 2 can be transferred in its mode of operation to the binding of the DNAzyme td1 to td78 to T-bet mRNA.
- the DNAzymes td1 to td78 are used unmodified for in vitro experiments, and for modifications in cell culture provided with modifications for GATA-3.
- the reverse transcription is carried out with the forward primer CGGCCCGCTGGAGAGGAAGC and reverse primer CACACACCCACACACAACC according to the standard procedure (ThermoScript from Invitrogen), whereby a PCR product with a length of 2450 nucleotides is amplified.
- This PCR product is cloned by standard methods into the plasmid pBluescript-SK (Stratagene) and sequenced for verification.
- Figure 8 shows a comparison of the nucleic acid sequence of T-bet No .: NM_013351 (Sequence 1) and Sequenced Sequence (Sequence 2). This shows both
- nucleic acid sequence 2 of T-bet of Figure 8 forms the basis for the construction of DNAzymes against T-bet mRNA in this invention.
- FIG. 8A shows the nucleotide sequence of the sequence 1 of the human T-bet gene
- FIG. 8 and therein depict, as a gray background, in each case two nucleotides GT or AT, between which there are further potential DNAzyme cleavage sites.
- the preparation of T-bet mRNA is carried out after linearization of the T-bet contained ⁇ the plasmid pBluescript-SK by cleavage with the restriction enzyme Xba I (Fermentas) and by in vitro transcription according to the manufacturer (Ambion). T-bet mRNA is present with a total length of 2550 nucleotides.
- FIG. 9 shows as a result of gel electrophoresis the cleavage of the T-bet target mRNA with modified DNAzymes [td54-M (lane 3), td69-M (lane 4), td70-M (lane 5)].
- Lane 2 contains as control T-bet target mRNA without DNAzyme addition.
- An entrained length standard (lane M) shows band sizes of 2000 bp and 3000 bp. Arrows point to A, the band with the substrate (here T-bet mRNA) and on B one of the two cleavage products (the other cleavage product is not visible in this figure).
- the comparison between all 78 DNAzymes shows that td54, td69 and td70 are particularly active, the modifications do not degrade the effectiveness of the DNAzyme.
- the following table shows the classification of the DNAzyme td 1 to td 78 against t-bet-3 mRNA into 4 groups. This group classification is carried out due to carried out in vitro activity tests of the DNAzyme against t-bet mRNA. Group 1: high cleavage activity, Group 2: medium cleavage activity, Group 3: weak cleavage activity, and Group 4: no measurable cleavage activity.
- step b) introducing the specific ribonucleic acid molecules from step b) into target cells
- the DNAzymes td54, td69 and td70 be with and without the described modes ⁇ fications used in target cells.
- the data for the transfection of Jurkat E6.1 cells correspond to those for the exemplary embodiment GATA-3.
- the T-bet mRNA quantity relative to GAPDH mRNA expression is quantitatively determined by real-time PCR (LightCycler, Roche) to obtain information about the in vitro effectiveness of the DNAzyme.
- LightCycler analyzes the RNA is purified from the Jurkat E6.1 cells using RNeasy Mini Kit (Qiagen, Germany) and subsequently normalized photometrically. After reverse transcription with SuperScript II (Gibco) according to the manufacturer's instructions, the quantitative analysis of the T-bet and GAPDH mRNA in the LightCycler follows.
- the total volume for the PCR is 20 ⁇ l, containing 1 ⁇ l DNA, 1 ⁇ l (0.5 ⁇ M) sense and antisense primer and 10 ⁇ l QuantiTect-SYBR-Green-PCR-Master-Mix (Qiagen, Germany).
- the PCR primers used for T-bet are: Sense 5 '-CCCACCATGTCCTACTACCG-3'; Antisense 5 ' - GCAATCTCAGTCCACACCAA-3 ' .
- the PCR primers for GAPDH are: sense 5 ' - TCTTCTTTTGCGTCGCCAG-3 ' and antisense 5 ' -AGCCCCAGCCTTCTCCA-3 ' .
- the PCR conditions are: denaturation (15min 95 ° C), amplification (15sec 95 ° C, 25sec 59 ° C, 25sec 72 ° C of 50 cycles) then final extension 2min 72 ° C.
- the subsequent melting curve is generated as follows: Osec 95 ° C, 15sec 60 ° C then the temperature is increased in 0.2 ° C steps to 97 ° C, at the same time the fluorescence is measured continuously.
- the melting curve is for internal control because all PCR products have a specific melting temperature.
- SYBR-Green is a fluorescent dye (included in the QuantiTect SYBR Green PCR Master Mix) that binds to double-stranded DNA. When the DNA is doubled during extension, SYBR-Green binds to it and generates a binding-dependent fluorescence signal, which is detected by the LightCycler at the end of each extension. The higher the amount of starting material, the sooner the significant increase in fluorescence will be detected.
- the LightCycler software plots collected fluorescence intensities versus cycles.
- FIG 10 shows T-bet and GAPDH mRNA LightCycler amplification curves after treatment of Jurkat E6.1 cells with DNAzymes td54m, td69m and td70m compared to nonsense DNAzyme treated.
- the respective Crossing Point (Ct) defined as the PCR cycle in which the fluorescence first differs significantly from the background fluorescence, is determined manually using the Fit-Point method of the LightCycler software T-bet and GAPDH mRNA in cells treated with DNAzyme compared to nonsense DNAzyme treated cells is determined according to the procedure described in User Bulletin # 2 (ABI Prism 7700 Sequence Detection System).
- DNAzymes td69 and td70 specifically inhibit T-bet expression in vivo and as a specific ribonucleic acid for producing a cell and / or tissue and / or disease phase specific Medicaments are suitable.
- td69 GGCAATGAAggctagctacaacgaTGGGTTTCT
- td70 TCACGGCAAggctagctacaacgaGAACTGGGT
- cells transfected with td69m or td70m are provided in a pharmaceutical composition with a pharmaceutically acceptable carrier, for example liposomes or biodegradable polymers.
- siRNA As an alternative to the DNAzymes, the use of siRNA is proposed for the specific inhibition of T-bet expression and for the production of a cell and / or tissue and / or disease phase-specific drug. Preferably, it is siRNA for the inhibition of human T-bet.
- the preparation of the siRNA is known to the person skilled in the art and described in the literature.
- DNAzymes or siRNAs can be produced as drugs in chronic inflammatory diseases and autoimmune diseases which are directed against further transcription factors that play a role in the differentiation to TH1 or TH2 cells
- STAT4 STAT ⁇ a, STAT1, c-Rel, CREB2, ATF-2, ATF-2, Hlx, IRF-1, c-Maf, NFAT, NIP45, AP1, Mel-18, SKAT-2, CTLA-4 or which are directed against further factors of the signal transduction pathways for differentiation and / or expression of cytokines, for example Src kinase, tea kinase, Rlk (Txk in humans), Itk, tea, RIBP, PLO ⁇ , MAP kinase, ERK, JNK, P38, MKK, MKK1, MKK2, MKK3, MKK4, MKK6, MKK7, Rac2, GADD45, GADD45 ⁇ , GADD
- These proteins have expression that is increased in a target cell compared to expression of a control cell.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Genetics & Genomics (AREA)
- General Health & Medical Sciences (AREA)
- Biomedical Technology (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Biotechnology (AREA)
- General Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Immunology (AREA)
- Biochemistry (AREA)
- Physics & Mathematics (AREA)
- Biophysics (AREA)
- Microbiology (AREA)
- Plant Pathology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Analytical Chemistry (AREA)
- Pulmonology (AREA)
- Endocrinology (AREA)
- Dermatology (AREA)
- Urology & Nephrology (AREA)
- Epidemiology (AREA)
- Vascular Medicine (AREA)
- Heart & Thoracic Surgery (AREA)
- Pain & Pain Management (AREA)
- Transplantation (AREA)
- Rheumatology (AREA)
Abstract
Description
Claims
Priority Applications (12)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
SI200430734T SI1670919T1 (sl) | 2003-10-02 | 2004-10-01 | Metoda za pripravo celic in/ali tkiva in/ali bolezni specifiäśno zdravilo |
DK04802618T DK1670919T3 (da) | 2003-10-02 | 2004-10-01 | Fremgangsmåde til fremstilling af et cellespecifikt og/eller vævsspecifikt og/eller sygdomsfasespecifikt lægemiddel |
PL04802618T PL1670919T3 (pl) | 2003-10-02 | 2004-10-01 | Sposób wytwarzania leku działającego swoiście na komórki i/lub tkanki i/lub wybrane fazy choroby |
KR1020067008072A KR101121818B1 (ko) | 2003-10-02 | 2004-10-01 | 세포 및/또는 조직 및/또는 질환 단계 특이적 의약품의제조 방법 |
CA2541240A CA2541240C (en) | 2003-10-02 | 2004-10-01 | Method for the production of a cell and/or tissue and/or disease phase specific medicament |
ES04802618T ES2304633T3 (es) | 2003-10-02 | 2004-10-01 | Procedimiento para la preparacion de un farmaco especifico para celulas y/o tejidos y/o fases de enfermedad. |
DE502004006791T DE502004006791D1 (de) | 2003-10-02 | 2004-10-01 | Verfahren zur herstellung eines zell- und/oder gewebe- und/oder krankheitsphasen-spezifischen arzneimittels |
JP2006529624A JP5013870B2 (ja) | 2003-10-02 | 2004-10-01 | 細胞および/または組織および/または疾患フェーズ特異的な薬剤の製造方法 |
DK07013576T DK1857555T3 (da) | 2003-10-02 | 2004-10-01 | Fremgangsmåde til fremstilling af et celle- og/eller vævs- og/eller sygdomsfase-specifikt lægemiddel |
US10/574,560 US8119789B2 (en) | 2003-10-02 | 2004-10-01 | Method for the production of a cell and/or tissue and/or disease phase specific medicament |
EP04802618A EP1670919B1 (de) | 2003-10-02 | 2004-10-01 | Verfahren zur herstellung eines zell- und/oder gewebe- und/oder krankheitsphasen-spezifischen arzneimittels |
US13/037,411 US8247544B2 (en) | 2003-10-02 | 2011-03-01 | Method for producing a cell and/or tissue and/or disease phase specific medicament |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
DE10346487.5 | 2003-10-02 | ||
DE10346487A DE10346487A1 (de) | 2003-10-02 | 2003-10-02 | Verfahren zur Herstellung eines Zell- und/oder Gewebe- und/oder Krankheitsphasen-spezifischen Arzneimittels |
Related Child Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US10/574,560 A-371-Of-International US8119789B2 (en) | 2003-10-02 | 2004-10-01 | Method for the production of a cell and/or tissue and/or disease phase specific medicament |
US13/037,411 Division US8247544B2 (en) | 2003-10-02 | 2011-03-01 | Method for producing a cell and/or tissue and/or disease phase specific medicament |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2005033314A2 true WO2005033314A2 (de) | 2005-04-14 |
WO2005033314A3 WO2005033314A3 (de) | 2005-09-01 |
Family
ID=34399325
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/DE2004/002197 WO2005033314A2 (de) | 2003-10-02 | 2004-10-01 | Verfahren zur herstellung eines zell- und/oder gewebe- und/oder krankheitsphasen-spezifischen arzneimittels |
Country Status (14)
Country | Link |
---|---|
US (2) | US8119789B2 (de) |
EP (2) | EP1857555B1 (de) |
JP (2) | JP5013870B2 (de) |
KR (1) | KR101121818B1 (de) |
AT (2) | ATE391788T1 (de) |
CA (2) | CA2541240C (de) |
CY (1) | CY1108161T1 (de) |
DE (3) | DE10346487A1 (de) |
DK (2) | DK1857555T3 (de) |
ES (2) | ES2320706T3 (de) |
PL (2) | PL1670919T3 (de) |
PT (2) | PT1670919E (de) |
SI (1) | SI1670919T1 (de) |
WO (1) | WO2005033314A2 (de) |
Cited By (13)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP2018395A1 (de) * | 2006-05-11 | 2009-01-28 | Wholesome Biopharma Pty Ltd | Kurze interferierende ribonukleinsäuren zur behandlung allergischer krankheiten |
WO2010107957A3 (en) * | 2009-03-19 | 2010-12-02 | Merck Sharp & Dohme Corp. | RNA INTERFERENCE MEDIATED INHIBITION OF GATA BINDING PROTEIN 3 (GATA3) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA) |
DE102011109868A1 (de) * | 2011-08-10 | 2013-02-14 | STERNA BIOLOGICALS GmbH & Co. KG. | Multiple Emulsion |
EP2708898A1 (de) | 2012-09-14 | 2014-03-19 | Sterna Biologicals GmbH & Co. Kg | Verfahren zur Diagnose eines molekularen Phänotyps eines an einer mit chronischen Entzündungen einhergehenden Erkrankung leidenden Patienten |
EP3093022A1 (de) | 2015-05-15 | 2016-11-16 | Sterna Biologicals GmbH & Co. KG | Gata-3-inhibitoren zur behandlung von th2-bewirktem asthma |
EP3211081A1 (de) | 2016-02-26 | 2017-08-30 | Secarna Pharmaceuticals GmbH & Co. KG | Neuartiger ansatz zur behandlung von entzündlichen erkrankungen |
WO2017144685A1 (en) | 2016-02-26 | 2017-08-31 | Secarna Pharmaceuticals Gmbh & Co. Kg | Novel approach for treating inflammatory disorders |
WO2017205506A1 (en) | 2016-05-24 | 2017-11-30 | Emory University | Particles with rna cleaving nucleobase polymers and uses for managing inflammatory disorders |
EP3501607A1 (de) | 2017-12-22 | 2019-06-26 | Sterna Biologicals GmbH & Co. KG | Zusammensetzung zur behandlung eines an einer mit chronischen entzündungen einhergehenden atemwegserkrankung leidenden patienten sowie herstellungsverfahren und verwendung der zusammensetzung |
EP3514235A1 (de) | 2018-01-18 | 2019-07-24 | Sterna Biologicals GmbH & Co. KG | Zusammensetzung zur behandlung eines an colitis ulcerosa leidenden patienten sowie verwendung der zusammensetzung als arzneimittel |
EP3603617A1 (de) | 2018-07-30 | 2020-02-05 | Sterna Biologicals GmbH & Co. KG | Aerosolerzeugungseinrichtung zur inhalativen verabreichung einer antisense-molekül-haltigen zusammensetzung |
EP3805242A1 (de) | 2019-10-07 | 2021-04-14 | Sterna Biologicals GmbH & Co. KG | Verfahren zur herstellung eines katalytisch aktiven dna-moleküls mit verbesserter aktivität und dessen verwendung in einem verfahren zur behandlung von asthma |
WO2023052422A1 (en) | 2021-09-30 | 2023-04-06 | Sterna Biologicals Gmbh | Dnazyme hydrogel formulations |
Families Citing this family (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JPWO2010110314A1 (ja) * | 2009-03-27 | 2012-10-04 | 協和発酵キリン株式会社 | 核酸を含有する肺高血圧症治療剤 |
DE102010007562A1 (de) | 2010-02-10 | 2011-08-11 | sterna biologicals GmbH & Co KG, 35043 | Dermatologische, pharmazeutische Zusammensetzung geeignet für Oligonukleotide |
DE102010056610A1 (de) * | 2010-12-31 | 2012-07-05 | Volker A. Erdmann | Pharmazeutische Zusammensetzung enthaltend L-DNA |
WO2019171191A1 (en) * | 2018-03-05 | 2019-09-12 | Dr. Reddy's Institute Of Life Sciences | Embryonic zebrafish models using dnazyme mediated knockdown |
US11976282B2 (en) | 2020-06-23 | 2024-05-07 | Qatar University | GATA3 inhibitors for the promotion of subcutaneous fat deposition |
AU2021416356A1 (en) | 2020-12-28 | 2023-08-10 | 1E Therapeutics, Ltd. | P21 mrna targeting dnazymes |
AU2021411103A1 (en) | 2020-12-28 | 2023-07-13 | 1E Therapeutics, Ltd. | P21 mrna target areas for silencing |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2000042173A1 (en) * | 1999-01-11 | 2000-07-20 | Unisearch Limited | Catalytic molecules |
WO2000051621A1 (en) * | 1999-03-05 | 2000-09-08 | Epigenesis Pharmaceuticals, Inc. | Method for validating/invalidating target(s) and pathways |
WO2001011023A1 (en) * | 1999-08-04 | 2001-02-15 | Johnson & Johnson Research Pty Ltd | Treatment of inflammatory or malignant disease using dnazymes |
Family Cites Families (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6361941B1 (en) * | 1998-03-27 | 2002-03-26 | Johnson & Johnson Research Pty Limited | Catalytic nucleic acid-based diagnostic methods |
CN1323344A (zh) * | 1998-08-13 | 2001-11-21 | 庄臣及庄臣研究股份有限公司 | 治疗再狭窄的DNAzyme和方法 |
WO2002068637A2 (en) * | 2000-10-20 | 2002-09-06 | Ribozyme Pharmaceuticals, Inc. | Nucleic acid-based treatment of diseases or conditions related to west nile virus infection |
US20030148971A1 (en) * | 2002-02-04 | 2003-08-07 | Handel Malcolm Lovell | Treatment of inflammatory and malignant diseases |
-
2003
- 2003-10-02 DE DE10346487A patent/DE10346487A1/de not_active Withdrawn
-
2004
- 2004-10-01 WO PCT/DE2004/002197 patent/WO2005033314A2/de active IP Right Grant
- 2004-10-01 DE DE502004008802T patent/DE502004008802D1/de not_active Expired - Lifetime
- 2004-10-01 US US10/574,560 patent/US8119789B2/en active Active
- 2004-10-01 AT AT04802618T patent/ATE391788T1/de active
- 2004-10-01 JP JP2006529624A patent/JP5013870B2/ja not_active Expired - Lifetime
- 2004-10-01 CA CA2541240A patent/CA2541240C/en not_active Expired - Lifetime
- 2004-10-01 KR KR1020067008072A patent/KR101121818B1/ko active IP Right Grant
- 2004-10-01 DK DK07013576T patent/DK1857555T3/da active
- 2004-10-01 CA CA2776997A patent/CA2776997C/en not_active Expired - Lifetime
- 2004-10-01 EP EP07013576A patent/EP1857555B1/de not_active Expired - Lifetime
- 2004-10-01 EP EP04802618A patent/EP1670919B1/de not_active Expired - Lifetime
- 2004-10-01 PL PL04802618T patent/PL1670919T3/pl unknown
- 2004-10-01 PL PL07013576T patent/PL1857555T3/pl unknown
- 2004-10-01 ES ES07013576T patent/ES2320706T3/es not_active Expired - Lifetime
- 2004-10-01 SI SI200430734T patent/SI1670919T1/sl unknown
- 2004-10-01 PT PT04802618T patent/PT1670919E/pt unknown
- 2004-10-01 ES ES04802618T patent/ES2304633T3/es not_active Expired - Lifetime
- 2004-10-01 PT PT07013576T patent/PT1857555E/pt unknown
- 2004-10-01 DE DE502004006791T patent/DE502004006791D1/de not_active Expired - Lifetime
- 2004-10-01 AT AT07013576T patent/ATE419364T1/de active
- 2004-10-01 DK DK04802618T patent/DK1670919T3/da active
-
2008
- 2008-06-27 CY CY20081100675T patent/CY1108161T1/el unknown
-
2011
- 2011-01-14 JP JP2011005485A patent/JP5291129B2/ja not_active Expired - Lifetime
- 2011-03-01 US US13/037,411 patent/US8247544B2/en not_active Expired - Lifetime
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2000042173A1 (en) * | 1999-01-11 | 2000-07-20 | Unisearch Limited | Catalytic molecules |
WO2000051621A1 (en) * | 1999-03-05 | 2000-09-08 | Epigenesis Pharmaceuticals, Inc. | Method for validating/invalidating target(s) and pathways |
WO2001011023A1 (en) * | 1999-08-04 | 2001-02-15 | Johnson & Johnson Research Pty Ltd | Treatment of inflammatory or malignant disease using dnazymes |
Non-Patent Citations (3)
Title |
---|
IMAGAWA S ET AL: "NEGATIVE REGULATION OF THE ERYTHROPOIETIN GENE EXPRESSION BY THE GATA TRANSCRIPTION FACTORS" 15. Februar 1997 (1997-02-15), BLOOD, W.B. SAUNDERS, PHILADELPHIA, VA, US, PAGE(S) 1430-1439 , XP000965159 ISSN: 0006-4971 Seite 1430, rechte Spalte, Absatz 2 * |
SANTORDO S W ET AL: "A GENERAL PURPOSE RNA-CLEAVING DNA ENZYME" PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE. WASHINGTON, US, Bd. 94, April 1997 (1997-04), Seiten 4262-4266, XP001009844 ISSN: 0027-8424 * |
SUN L Q ET AL: "Catalytic nucleic acids: From lab to applications" September 2000 (2000-09), PHARMACOLOGICAL REVIEWS, WILLIAMS AND WILKINS INC., BALTIMORE, MD,, US, PAGE(S) 325-347 , XP002272275 ISSN: 0031-6997 Seite 330 - Seite 337 * |
Cited By (29)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2009536518A (ja) * | 2006-05-11 | 2009-10-15 | ホールサム バイオファーム ピーティーワイ エルティーディー | アレルギー疾患を治療する短鎖干渉rna |
EP2018395A4 (de) * | 2006-05-11 | 2009-12-09 | Wholesome Biopharma Pty Ltd | Kurze interferierende ribonukleinsäuren zur behandlung allergischer krankheiten |
US8188262B2 (en) | 2006-05-11 | 2012-05-29 | Wholesome Biopharm Pty Ltd | Short interference ribonucleic acids for treating allergic diseases |
AU2007250524B2 (en) * | 2006-05-11 | 2012-11-01 | Wholesome Biopharm Pty Ltd | Short interference ribonucleic acids for treating allegic diseases |
EP2018395A1 (de) * | 2006-05-11 | 2009-01-28 | Wholesome Biopharma Pty Ltd | Kurze interferierende ribonukleinsäuren zur behandlung allergischer krankheiten |
WO2010107957A3 (en) * | 2009-03-19 | 2010-12-02 | Merck Sharp & Dohme Corp. | RNA INTERFERENCE MEDIATED INHIBITION OF GATA BINDING PROTEIN 3 (GATA3) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA) |
US20120029054A1 (en) * | 2009-03-19 | 2012-02-02 | Merck Sharp & Dohme Corp. | RNA Interference Mediated Inhibition of GATA Binding Protein 3 (GATA3) Gene Expression Using Short Intefering Nucleic Acid (siNA) |
US20140248377A1 (en) * | 2011-08-10 | 2014-09-04 | Sterna Biologicals Gmbh & Co. Kg Biomedizinisches Forschungszentrum | Multiple emulsion |
DE102011109868A1 (de) * | 2011-08-10 | 2013-02-14 | STERNA BIOLOGICALS GmbH & Co. KG. | Multiple Emulsion |
EP2708898A1 (de) | 2012-09-14 | 2014-03-19 | Sterna Biologicals GmbH & Co. Kg | Verfahren zur Diagnose eines molekularen Phänotyps eines an einer mit chronischen Entzündungen einhergehenden Erkrankung leidenden Patienten |
WO2014040891A3 (de) * | 2012-09-14 | 2014-05-30 | Sterna Biologicals Gmbh & Co. Kg | Verfahren zur diagnose eines molekularen phänotyps eines an einer mit chronischen entzündungen einhergehenden erkrankung leidenden patienten |
WO2014040891A2 (de) | 2012-09-14 | 2014-03-20 | Sterna Biologicals Gmbh & Co. Kg | Verfahren zur diagnose eines molekularen phänotyps eines an einer mit chronischen entzündungen einhergehenden erkrankung leidenden patienten |
US10591489B2 (en) | 2012-09-14 | 2020-03-17 | Sterna Biologicals Gmbh & Co. Kg | Method for diagnosing a molecular phenotype of a patient suffering from an illness accompanied by chronic inflammation |
RU2672595C2 (ru) * | 2012-09-14 | 2018-11-16 | ШТЕРНА БИОЛОГИКАЛС ГМБХ энд КО. КГ | Способ диагностики молекулярного фенотипа пациента, страдающего заболеванием, сопровождающимся хроническими воспалениями |
US9791454B2 (en) | 2012-09-14 | 2017-10-17 | Sterna Biologicals Gmbh & Co. Kg | Method for diagnosing a molecular phenotype of a patient suffering from an illness accompanied by chronic inflammation |
EP3093022A1 (de) | 2015-05-15 | 2016-11-16 | Sterna Biologicals GmbH & Co. KG | Gata-3-inhibitoren zur behandlung von th2-bewirktem asthma |
WO2017144685A1 (en) | 2016-02-26 | 2017-08-31 | Secarna Pharmaceuticals Gmbh & Co. Kg | Novel approach for treating inflammatory disorders |
EP3211081A1 (de) | 2016-02-26 | 2017-08-30 | Secarna Pharmaceuticals GmbH & Co. KG | Neuartiger ansatz zur behandlung von entzündlichen erkrankungen |
US10647987B2 (en) | 2016-02-26 | 2020-05-12 | Secama Pharmaceuticals GmbH & Co KG | Approach for treating inflammatory disorders |
WO2017205506A1 (en) | 2016-05-24 | 2017-11-30 | Emory University | Particles with rna cleaving nucleobase polymers and uses for managing inflammatory disorders |
EP3501607A1 (de) | 2017-12-22 | 2019-06-26 | Sterna Biologicals GmbH & Co. KG | Zusammensetzung zur behandlung eines an einer mit chronischen entzündungen einhergehenden atemwegserkrankung leidenden patienten sowie herstellungsverfahren und verwendung der zusammensetzung |
WO2019122267A1 (de) | 2017-12-22 | 2019-06-27 | Sterna Biologicals Gmbh & Co. Kg | Zusammensetzung zur behandlung eines an einer mit chronischen entzündungen einhergehenden atemwegserkrankung leidenden patienten sowie herstellungsverfahren und verwendung der zusammensetzung |
EP3514235A1 (de) | 2018-01-18 | 2019-07-24 | Sterna Biologicals GmbH & Co. KG | Zusammensetzung zur behandlung eines an colitis ulcerosa leidenden patienten sowie verwendung der zusammensetzung als arzneimittel |
WO2019141674A1 (de) | 2018-01-18 | 2019-07-25 | Sterna Biologicals Gmbh & Co.Kg | Zusammensetzung zur behandlung eines an colitis ulcerosa leidenden patienten sowie verwendung der zusammensetzung als arzneimittel |
EP3603617A1 (de) | 2018-07-30 | 2020-02-05 | Sterna Biologicals GmbH & Co. KG | Aerosolerzeugungseinrichtung zur inhalativen verabreichung einer antisense-molekül-haltigen zusammensetzung |
WO2020025559A1 (de) | 2018-07-30 | 2020-02-06 | Sterna Biologicals Gmbh & Co.Kg | Aerosolerzeugungseinrichtung zur inhalativen verabreichung einer antisense-molekül-haltigen zusammensetzung |
EP3805242A1 (de) | 2019-10-07 | 2021-04-14 | Sterna Biologicals GmbH & Co. KG | Verfahren zur herstellung eines katalytisch aktiven dna-moleküls mit verbesserter aktivität und dessen verwendung in einem verfahren zur behandlung von asthma |
WO2021069502A1 (en) | 2019-10-07 | 2021-04-15 | Sterna Biologicals Gmbh & Co. Kg | Method for the production of a catalytically active dna molecule having improved activity and its use in a method of treating asthma |
WO2023052422A1 (en) | 2021-09-30 | 2023-04-06 | Sterna Biologicals Gmbh | Dnazyme hydrogel formulations |
Also Published As
Similar Documents
Publication | Publication Date | Title |
---|---|---|
EP1670919B1 (de) | Verfahren zur herstellung eines zell- und/oder gewebe- und/oder krankheitsphasen-spezifischen arzneimittels | |
US9464292B2 (en) | Sense oligonucleotide capable of controlling the expression of INOS and composition comprising the same | |
CN102239260A (zh) | 通过抑制针对载脂蛋白-a1的天然反义转录物治疗载脂蛋白-a1相关疾病 | |
US20210267893A1 (en) | Artificial exosome composition and related methods | |
WO2022062440A1 (zh) | 靶向CTGF基因的gRNA及其应用 | |
Bhattarai et al. | Modulation of brain pathology by enhancer RNAs in cerebral ischemia | |
WO2014040891A2 (de) | Verfahren zur diagnose eines molekularen phänotyps eines an einer mit chronischen entzündungen einhergehenden erkrankung leidenden patienten | |
US8975023B2 (en) | Method for controlling the amount of gene product, and agent for controlling the amount of gene product | |
KR102544124B1 (ko) | eIF4E 저해제를 포함하는 eIF4E 활성증가와 관련된 상태의 진단 또는 치료용 조성물 | |
WO2013088853A1 (ja) | オリゴヌクレオチド、グルココルチコイド感受性増強剤、医薬組成物、及び発現ベクター | |
DE10148828A1 (de) | Modulation der Expression STAT-1-abhängiger Gene | |
JP2010529852A (ja) | 癌治療のためのNuMAのRNAi媒介ノックダウン | |
EP4355430A1 (de) | Behandlung von mst1r-bedingten erkrankungen und störungen |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
AK | Designated states |
Kind code of ref document: A2 Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW |
|
AL | Designated countries for regional patents |
Kind code of ref document: A2 Designated state(s): GM KE LS MW MZ NA 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 PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG |
|
DPEN | Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101) | ||
121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
WWE | Wipo information: entry into national phase |
Ref document number: 2004802618 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2006529624 Country of ref document: JP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2541240 Country of ref document: CA |
|
WWE | Wipo information: entry into national phase |
Ref document number: 1020067008072 Country of ref document: KR |
|
WWP | Wipo information: published in national office |
Ref document number: 2004802618 Country of ref document: EP |
|
WWP | Wipo information: published in national office |
Ref document number: 1020067008072 Country of ref document: KR |
|
WWE | Wipo information: entry into national phase |
Ref document number: 10574560 Country of ref document: US |
|
WWG | Wipo information: grant in national office |
Ref document number: 2004802618 Country of ref document: EP |