EP1409668A2 - Antisense nucleic acid against alphav integrin - Google Patents

Antisense nucleic acid against alphav integrin

Info

Publication number
EP1409668A2
EP1409668A2 EP01978406A EP01978406A EP1409668A2 EP 1409668 A2 EP1409668 A2 EP 1409668A2 EP 01978406 A EP01978406 A EP 01978406A EP 01978406 A EP01978406 A EP 01978406A EP 1409668 A2 EP1409668 A2 EP 1409668A2
Authority
EP
European Patent Office
Prior art keywords
nucleic acid
acid according
integrin
cells
vector
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP01978406A
Other languages
German (de)
French (fr)
Inventor
Ralf Kronenwett
Thorsten Gräf
Rainer Haas
Wolfgang Nedbal
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
A3D INTERNATIONAL GMBH
Original Assignee
A3D International 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 A3D International GmbH filed Critical A3D International GmbH
Priority to EP01978406A priority Critical patent/EP1409668A2/en
Publication of EP1409668A2 publication Critical patent/EP1409668A2/en
Withdrawn legal-status Critical Current

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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-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
    • C12N15/1138Non-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 receptors or cell surface proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Genetics & Genomics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Pain & Pain Management (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Rheumatology (AREA)
  • Biochemistry (AREA)
  • Dermatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to nucleic acid constructs for inhibiting the expression of αV integrins in mammalian cells, as well as to pharmaceutical compositions containing said antisense constructs.

Description

"Antisense nucleic acid against alphaV integrin"
Description
The present invention relates to nucleic acid constructs for inhibiting the expression of αV integrins in mammalian cells, as well as to pharmaceutical compositions containing said antisense constructs.
Integrins are a family of cell surface proteins that facilitate cell adhesion and play a major role in a number of biological processes including platelet aggregation, immune fuctions, tissue repair, cell proliferation, tumor invasion, inflammation, and inherited diseases. They are composed of two subunits α and β. So far, more than 11 α subunits and more than 6 β subunits are known. The subunit isotype αV is part of a number of α-β heterodimers including β 1 , β3, β5, β6 and β8. In the following, the integrin αVβ3 will be described as an example in more detail.
The integrin Vβ3 (also known as vitronectin receptor) is a heterodimeric protein which is expressed on the surface of various cell types and which functions as an adhesion molecule. Integrin αVβ3 consists of two subunits: The β3 chain which is present in all integrins of the β3-type and the specific αV chain. Integrin αVβ3 serves as a receptor for several extracellular matrix proteins such as vitronectin, fibronectin, laminin and collagen. This adhesion molecule is found, e.g. on activated endothelial and vascular muscle cells. Furthermore, this receptor is expressed on activated leucocytes, macrophages, osteoclasts and on cells of several types of tumors such as metastatic malignant melanoma, glioblastoma, urothelial carcinoma, breast cancer, ovarian carcinoma, prostatic carcinoma, astrocytoma, lung cancer, colon carcinoma, pancreatic carcinoma and osteosarcoma. αV integrins, in particular integrin αVβ3, play a central role in angiogenesis since their upregulation on endothelial cells promotes migration and sustains survival of vascular cells in angiogenic blood vessels. Neoangiogenesis is, in turn, important for many physiological processes such as wound healing, but also for pathological disorders such as growth of solid tumors or hematological neoplasias, restenosis after angioplasty, stenosis of vein bypasses, chronic inflammatory diseases and diabetic retinopathy. Therefore, the inhibition of neoangiogenesis represents a promising approach for the treatment of the above-mentioned pathological conditions.
So far, cancer diseases were treated by surgery, radiation therapy or chemotherapy. Recently, treatments using growth factors or immunological therapeutic approaches have been developed. The success thereof is highly dependent on the type of disease. However, concerning restenosis after angioplasty there are no successful therapeutic regimens available. Recent approaches using gene therapy are based on the local production of vasodilators. However, the effectiveness of such approaches remains to be shown. Chronic inflammatory diseases are commonly treated with non-steroidal antiphlogistics and corticosteroids. As a molecular therapeutic approach, antisense oligonucleotides directed against the adhesion molecule ICAM-1 have been developed recently (Yacyshyn et al., 1998, Gastroenterology 114: 1133-1142).
Recently, several substances for the inhibition of neoangiogenesis have been evaluated in clinical trials. Among them are interferons, angiogenesis-inhibiting cytokins, various substances which bind proangiogenic factors, substances which interfere with the matrix-endothelial interaction and thalidomide (reviewed in Gradishar, 1997, Invest. New Drugs 15: 49-59; Kuiper et al., 1998, Pharmacolog. Res. 37: 1-16).
Furthermore, a first specific inhibitor of integrin αVβ3 has been developed recently. This inhibitor is based on a peptide which binds to the αV subunit, thereby blocking the receptor. The inhibitor is suggested to be used in the treatment of malignant, tumors (malignant melanoma, colon carcinoma, neuroblastoma); cf. Lode et al., 1999, Proc. Natl. Acad. Sci. USA 96: 1591-1596.
All of the above-mentioned therapeutic approaches show one or more disadvantages. For example, chemotherapy and radiation therapy are unspecific and consequently have severe side effects. Furthermore, most tumors become resistant against chemotherapeutics. Corticosteroids and non-steroidal antiphlogistics also show severe side-effects. The specific blockage of integrin αVβ3 by monoclonal antibodies or peptides may lead to immunological reactions against these substances. Furthermore, the preparation of antibodies with the required quality and quantity is difficult and expensive. Moreover, besides their possible immunogenicity, peptides are often unstable in serum.
Accordingly, the technical problem underlying the present invention is to provide a novel system for the specific inhibition of the expression of αV integrins, e.g. in order to inhibit neoangiogenesis.
The solution to the above technical problem is achieved by providing the embodiments of the present invention as characterized in the claims.
In particular, the present invention provides a nucleic acid, e.g. an antisense nucleic acid, comprising a nucleotide sequence which is capable of binding to a transcription product of the gene coding for the αV integrin chain thereby inhibiting the expression of αV integrins in mammalian cells by at least 50%.
Preferably, the nucleic acid according to the present invention is capable of inhibiting the expression of the αV integrins selected from a group including αVβl , αVβ3, αVβδ, αVβ6 and αVβ8. A more preferred example is the inhibition of expression of integrin αVβ3.
The terms "nucleic acid" and "nucleotide sequence" refer to endogenously expressed, semisynthetic or chemically modified nucleic acid molecules of deoxyribonucleotides and/or ribonucleotides.
The term "transcriptional product" comprises all nucleic acid molecules which are primarily produced through transcription of the corresponding gene by the trans- cription machinery of the cell, or which are processed to mature mRNA by the splice apparatus and the further enzymatic systems for the formation of the 5'-cap and the 3'-poly-A+tail.
The term "binding" means, that the nucleic acid according to the present invention and the transcription product of the gene coding the αV integrin chain are capable of interacting specifically which may comprise the formation of covalent chemical bonds, electrostatic interactions based on salt bridges, or bonds based on hydrogen bridges and/or hydrophobic interactions as well as binding due to base stacking. In particular, hydrogen bonds and hydrophobic interactions are the driving forces for binding through hybridization between the nucleic acid according to the present invention and the transcription product of the gene coding for the αV integrin chain. A hybridization between the above-defined nucleic acid and the αV integrin transcription product may, for example, occur in case at least a part of the nucleotide sequence of the nucleic acid according to the present invention is, at least in part, complementary to at least a part of the mRNA coding for the αV integrin chain.
In a preferred embodiment of the present invention, the mammalian cells are human cells which are preferably selected from endothelial cells, vascular muscle cells, activated leukocytes, macrophages, osteoclasts and tumor cells such as cells from the metastating malignant melanoma, glioblastoma, urothelial carcinoma, breast cancer, ovarian carcinoma, prostatic carcinoma, astrocytoma, lung cancer, colon carcinoma, pancreatic carcinoma and osteosarcoma.
The nucleic acid according to the present invention may contain deoxyribonucleotides and/or ribonucleotides. Preferably, it contains modified nucleotides such as phosphorothiate nucleotides, aminonucleotides, [α-S]- trisphosphate nucleotides, pseudouridine nucleotides and nucleotides having bases containing chemical modifications such as methyl, acetyl or isopentenyl groups. Furthermore, the nucleic acid may be modified by gap-mer technology or may be a locked nucleic acid (LNA).
According to a further preferred embodiment, the nucleic acid according to the present invention is an antisense nucleic acid, wherein the 3'-end thereof is directed against the loop and/or the 5'-stem part of a local structure of the αV integrin chain mRNA; i.e. the 3'-end is capable of binding within the region of nucleotides 3871 to 3992 of the αV integrin chain mRNA (cf. also Fig. 7A). It is further preferred that the nucleic acid of the present invention has a length of 16 to 24 nucleotides.
Preferably, the nucleic acid according to the present invention is an antisense nucleic acid, wherein the nucleotide sequence contains one of the following sequences:
-GCTTGAAGGATTTGACCTAA-3 ' (SEQ ID NO 1 ), -TAGGTTGGCTTGAAGGATTT-3 ' (SEQ ID NO 2), -TATAGGTTGGCTTGAAGGAT -3 ' (SEQ ID NO 3), -AGTATAGGTTGGCTTGAAGG-3 ' (SEQ ID NO 4), -TTTAGTATAGGTTGGCTTGA-3 1 (SEQ ID NO 5), -GAAAAGCCATCGCCGAAGTG-3 ' (SEQ ID NO 6), -TTGGAAATATAAGGAAAAGT-3 ' (SEQ ID NO 7), -GCGGAAAAGCCATCGCCGAA-3 ' (SEQ ID NO 8),
-AATAAAATGCTTTTTTTATA-3 ' (SEQ ID NO 9), -TATTTTAATAAAATGCTTTT-3 ' (SEQ ID NO 10), or -TGGAAATATAAGGAAAAGTA-3 ' (SEQ ID NO 11 ).
The term "antisense nucleic acid" means that at least a part of the nucleotide sequence of the nucleic acid is at least in part complementary to at least a part of a nucleotide sequence of the transcription product of the gene coding for the αV integrin chain.
A further embodiment of the present invention relates to a vector comprising the above-defined nucleic acid. The term "vector" refers to a DNA and/or RNA replicon that can be used for the amplification of one or more heterologuous or homologuous nucleotide sequences. The vector according to the present invention may contain regulatory elements known in the art. In particular useful in the context of the present invention are DNA or RNA vectors which upon transcription provide the nucleic acid according to the present invention. An example is a DNA vector encoding the nucleic acid according to the present invention downstream of the 3'-end of a suitable promoter.
A further embodiment of the present invention relates to a host organism contai-ning the nucleic acid or the vector as defined above. The term "host organism" according to the present invention relates to any suitable prokaryotic or eukaryotic cell or may be a virus.
A further embodiment of the present invention relates to a method for the production of the above-defined nucleic acid or the vector, comprising cultivating the above- defined host organism under suitable conditions such as culture medium temperature etc. Preferably, the method further comprises isolating the desired products from the culture (cells and/or culture medium) according to procedures known in the art.
According to a further embodiment, the present invention provides a pharmaceutical composition containing the above-defined nucleic acid or the vector, optionally in combination with a pharmaceutically acceptable carrier and/or diluent.
A preferred embodiment of the pharmaceutical composition according to the present invention relates to the treatment of pathological disorders by modulation of cell adhesion. More preferably, the pharmaceutical composition according to the present invention affects platelet aggregation, immune functions, tissue repair, cell proliferation, tumor invasion, inflammation and inherited diseases. In case of the inhibition of the expression of integrin αVβ3, the pharmaceutical composition according to the present invention is particularly useful for the treatment of pathological disorders by inhibition of neoangiogenesis.
Examples of disorders which can be treated with the pharmaceutical composition according to the present invention comprise the group of cancer diseases such as the growth of solid tumors such as metastating malignant melanoma, glioblastoma, urothelial carcinoma, breast cancer, ovarian carcinoma, prostatic carcinoma, astrocytoma, lung cancer, colon carcinoma, pancreatic carcinoma and osteosarcoma or hematologic neoplasias such as multiple myeloma, acute and chronic leukemias, myelodysplastic syndrome and malignant lymphomas, restenosis after angioplasty, stenosis of vein bypasses, chronic inflammatory diseases such as Crohn's disease, ulcerative colitis, rheumatiod arthritis, ocular neovascular diseases such as diabetic retinopathy, disorders associated with excessive bone resorption such as osteoporosis, the above-mentioned multiple myeloma and bone metastasis, disorders of mammalian oral cavity which are related to inflammatory processes, e.g. concerning the gingival mucosa, such as gingivitis and peritonitis, and inflammatory skin disorders such as psoriasis, allergies, neurodermitis and akne. Further examples are acute or chronic inflammatory disorders of the cavity of the mouth, particularly inflammatory disorders of the gums.
A further embodiment of the present invention relates to a cosmetological composition containing the above-defined nucleic acid or the vector, optionally in combination with a cosmetologically acceptable carrier and/or diluent.
An application of the nucleic acid, the vector, the host organism or the pharmaceutical composition according to the present invention relates, for example, to a method for the inhibition of the expression of αV integrins such as integrin αVβ3 in mammals comprising introducing the nucleic acid, the vector, the host organism or the pharmaceutical composition according to the present invention in the mammalian organism. The method may be used, e.g. for the treatment of pathological disorders by modulation of cell adhesion, e.g. in order to inhibit neoangiogenesis, preferably for the treatment of the diseases as mentioned above.
The term "mammalian organism" comprises any type of single cells derived from mammals such as culture cells, as well as the mammal perse. Preferred mammalian organisms include man, dog, canine, cat, cow, pig and horse.
The term "introducing" into a mammalian organism may, e.g. in the case of single cells such as culture cells of mammalian cell lines, comprise a chemical transfection such as the Ca-phoshphate procedure or lipofection, or an electroporation, in the case of the whole animal or the human a subcutaneous, percutaneous, intramuscular, oral, intratracheal, peritoneal or intravenous and intraarterial administration of the nucleic acid, the vector, the host organism or the pharmaceutical composition according to the present invention. An intravenous or intraarterial administration may comprise, for example, a bolus injection and/or a continuous infusion.
Another embodiment of the present invention relates to a diagnostic kit containing the above-defined nucleic acid and/or the vector and/or the host organism. The diagnostic kit according to the present invention may, e.g. be used for the detection of expression levels of αV integrins. For example, the above-defined nucleic acid contained in the diagnostic kit according to the present invention may be used for PCR for the detection of αV integrin mRNA.
The figures show:
Fig. 1 is a graphical representation showing the relative αV expression after stimulation with phorbol-12-myristate-13-acetate (PMA) in human umbilical vein endothelial cells (HUVEC) which have been transfected with antisense oligodeoxyribonucleotides (ODN) according to the present invention (filled bars) or with control ODN (unfilled bars). The basis-αV- expression of unstimulated cells was set to 0% and the expression following stimulation by PMA was set to 100%.
Fig. 2 is a graphical representation showing the relative proliferation rate of HUVEC cells transfected with antisense ODN according to the present invention (filled bars) or with control ODN (unfilled bar). The proliferation rate of untransfected stimulated cells was set to 100%.
Fig. 3 is a graphical representation showing the annexin binding as a measure of apoptosis rate (expressed as mean fluorescence intensity) of HUVEC cells transfected with an ODN according to the present invention (av32, filled bar) or with a control ODN (avSC32, unfilled bar). Fig. 4 is a graphical representation showing the concentration dependence of αV-directed asON on protein expression in HUVEC. asON were transfected in HUVEC at indicated concentrations (10-100 nM) following stimulation of αV expression with PMA. Effects of asON on αV protein expression were determined by FACS analysis. The basis αV-expression of unstimulated cells was set to 0% and the expression following stimulation by PMA was set to 100%.
Fig. 5 is a graphical representation showing the effect of αV-directed asON on mRNA levels in HUVEC after stimulation with PMA. αV mRNA levels are strongly decreased in the presence of 30 nM asON (left hand side), whereas levels of control mRNA 18S are not affected indicating specificity of asON-mediated inhibiton (right hand side).
Fig. 6 is a graphical representation showing a comparison of αV-directed asON according to the present invention (aV3871 , aV3880, aV3882) with control asON (aV3865, aV3867) and asON previously described in the literature (aV40=SeqlD6 in EP-A-0 950 709; aV40=ASODN3 in Treasurywala et al. (1998) GLIA 24, 236-243); aV136=AS4 in Nip et al. (1995) J. Clin. Invest. 95, 2096-2103). Effects of asON on αV protein expression were determined by FACS analysis. The basis αV-expression of unstimulated cells was set to 0% and the expression following stimulation by PMA was set to 100%. Oligonucleotides av3865 and av3867 are inactive control asON.
Fig. 7 is a graphical representation showing positions (A) and efficacy (B) of αV- directed asON against target region pos. 3865 - pos. 3910. Effects of asON (50 nM) on αV protein expression were determined by FACS analysis. The basis αV-expression of unstimulated cells was set to 0% and the expression following stimulation by PMA was set to 100%. The present invention is further illustrated by the following non-limiting example.
EXAMPLE
Computer-aided selection of target sequences
The secondary structures of the αV mRNA were calculated with the computer program mfold 2.0 of the Heidelberg Unix Sequence Analysis Resources (HUSAR). The secondary structures were systematically analysed by the method of Patzel et al. (Nucl. Acids Res. 27: 4823-4334, 1999). On the basis of these analyses, suitable local target sequences were identified.
Antisense oligonucleotides
Selected antisense ODN and control ODN were all phosphorothioate-modified and were synthesized by a commercial supplier.
The antisense oligonucleotides which showed the best performance in the inhibition of αV expression had the following sequences:
av3871: 5' -GCTTGAAGGATTTGACCTAA-3 (pos. 3871-3890, SEQ ID NO 1) av3878: 5' -TAGGTTGGCTTGAAGGATTT-3 (pos. 3878-3897, SEQ ID NO 2) av 3880: 5' - ATAGGTTGGCTTGAAGGAT-3 (pos. 3880-3899, SEQ ID NO 3) av3882: 5' -AGTATAGGTTGGCTTGAAGG-3 (pos. 3882-3901 , SEQ ID NO 4) av3884: 5' -TTTAGTATAGGTTGGCTTGA-3 (pos. 3884-3903, SEQ ID NO 5) av32: 5' -GAAAAGCCATCGCCGAAGTG-3 (pos: 32-51 , SEQ ID NO 6) av4175: 5 -TTGGAAATATAAGGAAAAGT-3 (pos.4175-4194, SEQ ID NO 7) av35: 5 -GCGGAAAAGCCATCGCCGAA-3 (pos. 35-54, SEQ ID NO 8) av 4044: 5' -AATAAAATGCTTTTTTTATA-3 (pos. 4044-4063, SEQ ID NO 9) av 4050: 5 -TATTTTAATAAAATGCTTTT-3 (pos. 4050-4069, SEQ ID NO 10 ) av4174: 5 -TGGAAATATAAGGAAAAGTA-3 (pos. 4174-4193, SEQ ID NO 11 ) The positions indicated above refer to the αV cDNA sequence as published by Suzuki et al. (J. Biol. Chem. 262:14080-14085, 1987).
Transfection
The activity of antisense ODN was tested on human umbilical vein endothelial cells (HUVEC). The oligonucleotides (0,05 μM final concentration) were transfected in HUVEC using lipofectin as carrier. The cells were stimulated with PMA after 8 hours. After further 48 hours, the expression of the αV integrin chain, the proliferation and the apoptosis rate were studied.
Functional studies
Expression of the αV integrin chain was determined by flowcytometry. Using the mean fluorescence intensity of the cells in each sample, the inhibition of the PMA- induced expression of the αV integrin chain by the ODN was calculated.
The proliferation after transfection with ODN was determined by photometrical measurement of the BrdU incorporation. The apoptosis rate was determined by flowcytometry by the binding of annexin.
As assessed by flowcytometry already after 48 hours following transfection, 3 out of 7 antisense ODN inhibited the upregulation of αV completely at an ODN concentration of 0,05 μM. Furthermore, all ODN according to the present invention surprisingly inhibit the upregulation of αV by more than 50% at this low ODN concentration (see Fig. 1). The inhibition of αV expression was furthermore associated with an inhibition of proliferation of HUVEC cells by 32% (Fig. 2) and led to a 5-fold increase in the apoptosis rate of antisense-transfected cells in comparison to control cells (Fig. 3).
In addition, pretreatment of HUVEC cells with an αV-directed antisense oligonucleotide inhibited the formation of tubular structures in an endothelial cell tube assay using Matrigel-coated cell culture plates.
In further experiments, asODN av3831 (SEQ ID NO 1 ), av3880 (SEQ ID NO 3) and av3882 (SEQ ID NO 4) as preferred examples of the nucleic acid according to the present invention were characterized more specifically. In particular, the dose- response relationship of these asODN was studied at concentrations in the range of 10 to 100 nM. Experiments were carried out as described above and the results are summarized in Fig. 4. The concentration dependency of the inhibition obtained with all three species examined indicates a specific inhibition of αV protein expression through an antisense mechanism.
Furthermore, the inhibitory power of the above three asODN was studied on the αV mRNA level. The presence of the αV-directed antisense species according to the present invention led to an extraordinarily strong decrease in the copy number of αV mRNA which is not observed in case of a control mRNA (18S RNA). This result further demonstrates the specificity of the antisense-Iinked inhibiton of αV expression by the nucleic acid of the present invention.
As shown in Fig. 6, antisense nucleic acids according to the present invention display a significantly stronger inhibiton of αV expression than prior art asODN (aV40: 5 ' - GCGGCGGAAAAGCCATCG-3 ' disclosed in EP-A-0 950 709; aV4: 5 ' - ACGCCAAGCCGGGAGCGGTA-3 ' disclosed in Treasurywala et al. (1998) GLIA 24, 236- 243); aV136: 5 ' -GACTGTCCACGTCTAGGT- 3 disclosed in Nip et al. (1995) J. Clin. Invest. 95, 2096-2103). Furthermore, the 3'-end of preferred asODN is directed against a common structure, i.e. the loop and 5'-stem part of the αV integrin mRNA (Fig. 7A). These preferred asODN all showed a surprisingly strong inhibition of αV integrin expression whereas control asODN having a 3'-end lying outside the region of nucleotides 3871 to 3992 of the αV mRNA were ineffective (Fig. 7B).
In conclusion, antisense sequences were identified which, after transfection in form of DNA oligonucleotides, lead to an inhibition of PMA-induced αV-expression in endothelial cells. The inhibition of αV-expression was associated with a decreased proliferation of the endothelial cells and an increased apoptosis rate. Therefore, the present invention provides for the first time a highly specific inhibitor of αV- expression, e.g. in endothelial cells, on the basis of antisense oligonucleotides which do not act by interfering with the extracellular protein-protein interaction but which act intracellularly by downregulation of the expression of the receptor. In comparison to other αV-antagonists the nucleic acid inhibitors of the present invention can be prepared easily in large amounts and in good manufacturing practice (GMP) quality by simple chemical synthesis.
The antisense-based inhibitors of the present invention can be used successfully in inhibition of neoangiogenesis, e.g. for the treatment or prevention of restenosis following angioplasty or for the treatment of malignant tumors or hematological malignancies such as multiple myeloma.

Claims

Claims
1. A nucleic acid comprising a nucleotide sequence which is capable of binding to a transcription product of the gene coding for the αV integrin chain thereby inhibiting the expression of αV integrins in mammalian cells by at least 50%.
2. The nucleic acid according to claim 1 , wherein the 3'-end is capable of binding within the region of nucleotides 3871 to 3992 of the mRNA coding for the αV integrin chain.
3. The nucleic acid according to claim 1 or 2, wherein the αV integrin is selected from the group consisting of integrin αVβl , αVβ3, αVβ5, αVβ6 and αVβδ.
4. The nucleic acid according to claim 3, wherein the αV integrin is integrin Vβ3.
5. The nucleic acid according to any one of claims 1 to 4, wherein the mammalian cells are human cells.
6. The nucleic acid according to claim 5, wherein the human cells are selected from endothelial cells, vascular muscle cells, activated leukocytes, macrophages, osteoclasts and tumor cells.
7. The nucleic acid according to any one of claims 1 to 6 containing modified nucleotides.
8. The nucleic acid according to claim 7, wherein the modified nucleotides are phosphorothioate nucleotides.
9. The nucleic acid according to any one of claims 1 to 8, wherein the nucleotide sequence has a length of 16 to 24 nucleotides.
10. The nucleic acid according to any one of claims 1 to 9, wherein the nucleotide sequence contains one of the sequences of SEQ ID NO 1 to 5.
11. A vector comprising the nucleic acid according to any one of claims 1 to 10.
12. A host organism containing the nucleic acid according to any one of claims 1 to 10 or the vector according to claim 11.
13. A method for the production of the nucleic acid according to any one of claims 1 to 10 or the vector according to claim 11 , comprising cultivating the host organism according to claim 12.
14. A pharmaceutical composition containing the nucleic acid according to any one of claims 1 to 10 or the vector according to claim 11 , optionally in combination with a pharmaceutically acceptable carrier and/or diluent.
15. The pharmaceutical composition according to claim 14 for the treatment of pathological disorders by modulation of cell adhesion.
16. The pharmaceutical composition according to claim 15, which affects platelet aggregation, immune functions, tissue repair, cell proliferation, tumor invasion, inflammation and inherited diseases.
17. The pharmaceutical composition according to claim 15 or 16, wherein the disorder is selected from the group of cancer diseases, restenosis after angioplasty, stenosis of vein bypass, chronic inflammatory diseases, ocular neovascular diseases, disorders associated with excessive bone resorption, disorders of mammalian oral cavity and inflammatory skin disorders.
18. The pharmaceutical composition according to claim 14 for the treatment of acute or chronic inflammatory disorders of the cavity of mouth, particularly inflammatory disorders of the gums.
19. A cosmetological composition containing the nucleic acid according to any one of claims 1 to 10 or the vector according to claim 11 , optionally in combination with a cosmetologically acceptable carrier and/or diluent.
20. A diagnostic kit containing the nucleic acid according to any one of claims 1 to 10 and/or the vector according to claim 11 and/or the host organism according to claim 12.
EP01978406A 2000-10-12 2001-10-11 Antisense nucleic acid against alphav integrin Withdrawn EP1409668A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP01978406A EP1409668A2 (en) 2000-10-12 2001-10-11 Antisense nucleic acid against alphav integrin

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP00121394A EP1197553A1 (en) 2000-10-12 2000-10-12 Antisense nucleic acid against alphaV integrin
EP00121394 2000-10-12
PCT/EP2001/011790 WO2002031142A2 (en) 2000-10-12 2001-10-11 Antisense nucleic acid against alphav integrin
EP01978406A EP1409668A2 (en) 2000-10-12 2001-10-11 Antisense nucleic acid against alphav integrin

Publications (1)

Publication Number Publication Date
EP1409668A2 true EP1409668A2 (en) 2004-04-21

Family

ID=8169980

Family Applications (2)

Application Number Title Priority Date Filing Date
EP00121394A Withdrawn EP1197553A1 (en) 2000-10-12 2000-10-12 Antisense nucleic acid against alphaV integrin
EP01978406A Withdrawn EP1409668A2 (en) 2000-10-12 2001-10-11 Antisense nucleic acid against alphav integrin

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP00121394A Withdrawn EP1197553A1 (en) 2000-10-12 2000-10-12 Antisense nucleic acid against alphaV integrin

Country Status (3)

Country Link
EP (2) EP1197553A1 (en)
AU (1) AU2002210530A1 (en)
WO (1) WO2002031142A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9345739B2 (en) 2007-11-08 2016-05-24 The General Hospital Corporation Methods and compositions for the treatment of proteinuric diseases

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002000877A2 (en) * 2000-06-29 2002-01-03 Mcgill University Antisense oligonucleotides to inhibit angiogenesis and/or metastasis
CN100537761C (en) * 2003-11-07 2009-09-09 中国人民解放军军事医学科学院放射医学研究所 Antisense oligonucleotide contains their drug regimen and is used for the treatment of the purposes of cancer

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0950097A2 (en) * 1996-12-27 1999-10-20 Instituto de Investigaciones Bioquimicas Fundacion Campomar Compositions and methods for tumour therapy
EP0950709A1 (en) * 1998-04-17 1999-10-20 Hoechst Marion Roussel Deutschland GmbH Antisense oligonucleotides for the inhibition of integrin alphaV-subunit expression
WO2000044409A1 (en) * 1999-01-27 2000-08-03 University College London Formulations comprising antisense nucleotides to connexins

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0231142A2 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9345739B2 (en) 2007-11-08 2016-05-24 The General Hospital Corporation Methods and compositions for the treatment of proteinuric diseases

Also Published As

Publication number Publication date
EP1197553A1 (en) 2002-04-17
WO2002031142A3 (en) 2003-01-09
AU2002210530A1 (en) 2002-04-22
WO2002031142A2 (en) 2002-04-18

Similar Documents

Publication Publication Date Title
US6566127B1 (en) Method and reagent for the treatment of diseases or conditions related to levels of vascular endothelial growth factor receptor
JP6991966B2 (en) Compositions and Methods for Inhibition of LPA Gene Expression
JP3054745B2 (en) Antisense oligonucleotide regulation of raf gene expression
JP3231779B2 (en) Antisense oligonucleotide regulation of raf gene expression
US5585479A (en) Antisense oligonucleotides directed against human ELAM-I RNA
JP2019014733A (en) Splice switching oligomers for tnf superfamily receptors and use of oligomers in treatment of disease
EP0690726B1 (en) Antisense inhibition of c-myc to modulate the proliferation of smooth muscle cells
US20070042029A1 (en) Enzymatic nucleic acid-mediated treatment of ocular diseases or conditions related to levels of vascular endothelial growth factor receptor (VEGF-R)
JPH10500309A (en) Methods and compositions for treating restenosis and cancer using ribozymes
JP2000506725A (en) Inhibit cell growth
JPH10512446A (en) Compositions and methods for treating tumor cells
JP2004507263A (en) Antisense oligonucleotides to VR1
US5580969A (en) Antisense oligonucleotides directed against human ICAM-I RNA
JP2014148515A (en) Administration of the reg1 anticoagulation system
Tang et al. Intravenous angiotensinogen antisense in AAV-based vector decreases hypertension
US5596090A (en) Antisense oligonucleotides directed against human VCAM-1 RNA
KR20120048613A (en) Sparc antisense compositions and uses thereof
JPH06209778A (en) Nf-kappa. b anti sense polynucleotide
WO1998024797A1 (en) Antisense inhibition of human adhesion molecules
Bennett et al. Pharmacology of antisense therapeutic agents: cancer and inflamination
WO2002031142A2 (en) Antisense nucleic acid against alphav integrin
KR100211178B1 (en) Antisense oligonucleotide modulation of raf gene expression
US20040220131A1 (en) Method for treatment of cancerous angiogenic disorders
RU2112766C1 (en) Oligonucletides, pharmaceutical composition
Sioud et al. Therapeutic RNA and DNA enzymes

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20031016

AK Designated contracting states

Kind code of ref document: A2

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

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

17Q First examination report despatched

Effective date: 20050524

RAP3 Party data changed (applicant data changed or rights of an application transferred)

Owner name: A3D INTERNATIONAL GMBH

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

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

18D Application deemed to be withdrawn

Effective date: 20061213