US20030114410A1 - Pharmaceutical compositions and methods useful for modulating angiogenesis and inhibiting metastasis and tumor fibrosis - Google Patents

Pharmaceutical compositions and methods useful for modulating angiogenesis and inhibiting metastasis and tumor fibrosis Download PDF

Info

Publication number
US20030114410A1
US20030114410A1 US10/305,348 US30534802A US2003114410A1 US 20030114410 A1 US20030114410 A1 US 20030114410A1 US 30534802 A US30534802 A US 30534802A US 2003114410 A1 US2003114410 A1 US 2003114410A1
Authority
US
United States
Prior art keywords
gly
lysyl
ala
arg
oxidase
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.)
Abandoned
Application number
US10/305,348
Inventor
Gera Neufeld
Gal Akiri
Zahava Vadasz
Stela Gengrinovitch
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.)
Technion Research and Development Foundation Ltd
Original Assignee
Technion Research and Development Foundation Ltd
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
Priority claimed from PCT/IL2001/000728 external-priority patent/WO2002011667A2/en
Application filed by Technion Research and Development Foundation Ltd filed Critical Technion Research and Development Foundation Ltd
Priority to US10/305,348 priority Critical patent/US20030114410A1/en
Assigned to TECHNION RESEARCH AND DEVELOPMENT FOUNDATION LTD. reassignment TECHNION RESEARCH AND DEVELOPMENT FOUNDATION LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GENGRINOVITCH, STELA, VADASZ, ZAHAVA, AKIRI, GAL, NEUFELD, GERA
Publication of US20030114410A1 publication Critical patent/US20030114410A1/en
Priority to DK08020753.3T priority patent/DK2062919T3/en
Priority to EP08020752A priority patent/EP2062918A3/en
Priority to US10/536,440 priority patent/US8163494B2/en
Priority to PCT/IL2003/001008 priority patent/WO2004047720A2/en
Priority to AU2003286391A priority patent/AU2003286391A1/en
Priority to EP08020753.3A priority patent/EP2062919B1/en
Priority to PT08020753T priority patent/PT2062919E/en
Priority to ES08020753.3T priority patent/ES2584847T3/en
Priority to EP03777136A priority patent/EP1572100A4/en
Priority to US12/571,167 priority patent/US8168180B2/en
Priority to HK09110803.7A priority patent/HK1131165A1/en
Priority to US13/412,544 priority patent/US20120202206A1/en
Priority to US13/416,976 priority patent/US8815823B2/en
Priority to US14/311,110 priority patent/US20140302499A1/en
Priority to CY20161100760T priority patent/CY1117943T1/en
Priority to US15/881,339 priority patent/US20190040470A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING 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
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/0012Oxidoreductases (1.) acting on nitrogen containing compounds as donors (1.4, 1.5, 1.6, 1.7)
    • C12N9/0014Oxidoreductases (1.) acting on nitrogen containing compounds as donors (1.4, 1.5, 1.6, 1.7) acting on the CH-NH2 group of donors (1.4)
    • C12N9/0022Oxidoreductases (1.) acting on nitrogen containing compounds as donors (1.4, 1.5, 1.6, 1.7) acting on the CH-NH2 group of donors (1.4) with oxygen as acceptor (1.4.3)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING 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
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/26Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving oxidoreductase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y104/00Oxidoreductases acting on the CH-NH2 group of donors (1.4)
    • C12Y104/03Oxidoreductases acting on the CH-NH2 group of donors (1.4) with oxygen as acceptor (1.4.3)
    • C12Y104/03013Protein-lysine 6-oxidase (1.4.3.13), i.e. lysyl-oxidase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57419Specifically defined cancers of colon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Definitions

  • the present invention relates to pharmaceutical compositions and methods useful for modulating angiogenesis and for inhibiting metastasis and fibrosis in a mammalian tissue.
  • vasculogenesis the transformation of pre-existing arterioles into small muscular arteries
  • angiogenesis the sprouting of existing blood vessels (which occurs both in the embryo and in the adult).
  • vascular endothelial growth factor VEGF
  • bFGF basic fibroblast growth factor
  • VEGF vascular endothelial growth factor
  • bFGF basic fibroblast growth factor
  • Platelet factor-4 is an anti-angiogenic protein normally sequestered in platelets (Tanaka et al., 1997; Maione et al., 1990; Neufeld et al., 2000). PF4 inhibits angiogenesis using poorly defined mechanisms (Gengrinovitch et al., 1995; Brown, and Parish, 1994; Gupta, and Singh, 1994; Watson et al., 1994). It was previously speculated that PF4 binds to cell surface heparan-sulfate proteoglycans and in this manner inhibits the activity of angiogenic growth factors such as basic fibroblast growth factor (Watson et al., 1994).
  • the present inventors While reducing the present invention to practice, the present inventors have also discovered that the lysyl oxidase protein, LOR-1, participates in metastasis and as such can be used as a target for inhibiting metastasis and reducing tumor invasiveness.
  • LOR-1 lysyl oxidase protein
  • a pharmaceutical composition useful for modulating angiogenesis in mammalian tissue comprising, as an active ingredient, a molecule capable of modifying a level and/or activity of at least one type of lysyl-oxidase of the mammalian tissue and a pharmaceutically effective carrier.
  • the antibody or the antibody fragment is directed against at least a portion of the polypeptide set forth in SEQ ID NO: 2, 3, 6, 8 or 9.
  • the molecule is a polynucleotide capable of down regulating expression of the at least one type of lysyl-oxidase.
  • polynucleotide is at least partially complementary with the polynucleotide set forth in SEQ ID NO: 1, 4, 5 or 7.
  • polypeptide is as set forth in SEQ ID NO: 2, 3, 6, 8 or 9.
  • a method of modulating angiogenesis in a mammalian tissue comprising administering into the mammalian tissue a nucleic acid construct being capable of expressing a polypeptide having lysyl-oxidase activity to thereby modulate angiogenesis within the mammalian tissue.
  • polypeptide is at least 75% homologous to the polypeptide set forth in SEQ ID NO: 2, 3, 6, 8 or 9.
  • step (a) is effected by binding assays and/or lysyl-oxidase activity assays.
  • the method comprising (a) determining a lysyl-oxidase expression level and/or activity of the cancerous tissue; and (b) comparing the lysyl-oxidase expression level and/or activity with that determined for control tissue to thereby determine the malignancy of the cancerous tissue.
  • a mammalian tissue comprising administering into the mammalian tissue a molecule capable of downregulating a tissue level and/or activity of at least one type of lysyl-oxidase to thereby inhibit metastasis in the mammalian tissue
  • a pharmaceutical composition useful for inhibiting metastasis and fibrosis in mammalian tissue comprising, as an active ingredient, a molecule capable of downregulating a level and/or activity of at least one type of lysyl-oxidase of the mammalian tissue and a pharmaceutically effective carrier.
  • the molecule is an antibody or an antibody fragment capable of binding with, and at least partially inhibiting the activity of, the at least one polypeptide.
  • the molecule is a polynucleotide capable of downregulating expression of the at least one type of lysyl-oxidase.
  • step (a) is effected by binding assays and/or lysyl-oxidase activity assays.
  • FIG. 1 illustrates SDS-PAGE analysis of extracts of Pporcine aortic endothelial cells (PAE Cells) which were transfected with vector along (lane 1) or with vector containing the LOR-1 cDNA (lane 3) and metabolically labeled with 35 S-methionine. Extracts from the vector transfected cells (lane 2), from vector containing LOR-1 cDNA transfected cells (lane 4) or from 35 S-methionine labeled human umbilical vein endothelial cells (HUVEC) (lane 5) were purified on a PF4 affinity column. A Band corresponding in size to the original band observed in the HUVEC is evident (compare lanes 4 and 5); this band is absent in extracts of vector transfected cells.
  • PEP Cells Pporcine aortic endothelial cells
  • FIG. 3 illustrates expression of recombinant LOR-1 in MCF-7 breast cancer cells (lane 1).
  • Vector transfected MCF-7 cells (lane 2) and two clones of MCF-7 expressing recombinant LOR-1 (lane 3, clone 12, lane 4, clone 22) were grown for two days in serum free medium.
  • the medium from an equal number of cells was collected, concentrated 30 fold using CentriconTM, and 10 ⁇ l aliquots were electrophoresed using an SDS/PAGE gel. Proteins were blotted onto nitrocellulose, and LOR-1 protein was identified using an antibody directed against the C-terminal of LOR-1.
  • a secondary alkaline-phosphatase coupled antibody and NBT-BICP staining were used to detect the primary bound antibody.
  • FIG. 4 illustrates tumor size as correlated to LOR-1 expression.
  • Parental MCF-7 cells (par), MCF-7 cells transfected with pCDNA3 vector alone (vec) and two recombinant LOR-1 expressing MCF-7 cells (clones 12 and 24) were deposited under the skin of immune deficient mice (10 7 /injection site) along with an estrogen slow release pellet. Six animals were used for each cell type implanted. The area of the tumors was measured every few days. Bars represent standard deviation from the mean.
  • FIGS. 5 a - b illustrate anti-factor-8 immunostaining of tumors generated by MCF-7 cells transfected with the expression vector alone (FIG. 5 a ) or with an expression vector containing the LOR-1 cDNA (FIG. 5 b ). Counter staining was performed with hematoxylin-eosin (blue). Invasion of blood vessels into the tumor mass is more abundant in LOR-1 expressing tumors (FIG. 5 b ) as compared to tumors generated by control cells which do not express LOR-1 (FIG. 5 a ).
  • FIGS. 9 a - b illustrate the expression of LOR-1 and LOR-2 in human breast cancer derived cells:
  • Total RNA was prepared from confluent MCF-7 cells (MCF-7), MDA-MB-231 cells (MDA-231) and MDA-MB-435 cells (MDA-435) and was subjected to Northern blot analysis using a LOR-1 (FIG. 9 a ) and LOR-2 (FIG. 9 b ) specific cDNA probes.
  • LOR-1 specific hybridization signal is seen in tumors derived from both MDA-231 and MDA-435 cells but not in tumors derived from MCF-7 cells.
  • LOR-2 specific hybridization signal is seen only in tumors derived from MDA-435 cells.
  • LOR-1 and LOR-2 non-specific hybridization signals are seen in all tumors' RNA in a band that corresponds to the 28S rRNA.
  • FIGS. 9 c - f illustrate the expression pattern of LOR-1 in normal human breast (FIG. 9 c ), in in-situ non-invasive breast carcinoma (FIG. 9 d ), in grade-1 invasive ductal carcinoma (FIG. 9 e ) and in grade-3 invasive ductal carcinoma (FIG. 9 f ).
  • a polyclonal affinity purified rabbit antibody directed against the C-terminal of LOR-1 was used to detect the expression of LOR-1.
  • High level expression of LOR-1 protein is seen in the epithelium of the normal duct (FIG. 9 c , open arrow); magnification ⁇ 100.
  • in-situ non-invasive breast carcinoma FIG.
  • the cancer cells have filled the duct but are still confined to it. Many of the cells located at periphery of the tumor have lost their ability to express LOR-1, while at the center, the cells still express high levels of LOR-1 (FIG. 9 d , empty arrow); magnification ⁇ 200.
  • LOR-1 high levels of LOR-1
  • the tumorigenic cells In grade-1 invasive breast carcinoma the tumorigenic cells have formed pseudo-ducts (FIG. 9 e , black arrows) but they do not express LOR-1 anymore. However, cells found in a nearby carcinoma in-situ express LOR-1 (FIG. 9 e , empty arrow); magnification ⁇ 200.
  • grade-3 invasive breast carcinoma the tumorigenic cells express large amounts of LOR-1 and the morphology is completely disorderly (FIG. 9f); magnification ⁇ 200.
  • FIG. 10 a illustrates the expression of recombinant LOR-1 in MCF-7 cells transfected with the expression vector alone (vec) or with an expression vector containing the LOR-1 cDNA clone 12 or 24 (#12 or #24, respectively).
  • LOR-1 proteins were deteceted by Western blot with an antibody directed against the C-terminal of human LOR-1.
  • FIG. 10 c illustrates the growth rate of tumors derived from control or LOR-1 expressing MCF-7 cells.
  • MCF-7 cells transfected with the expression vector alone (vec) or with an expression vector containing the LOR-1 cDNA clone 12 or 24 were injected into the mammary fat pads of female athymic nude mice. Each cell type was implanted in 8 animals. Tumor area was measured at the indicated times. Error bars represent the standard error of the mean. The experiment was terminated and the mice sacrificed when the tumor reached a diameter of about 1 cm.
  • FIG. 10 d illustrates the relative size of tumor in mice 25 days following injection of MCF-7 cells. Shown are mice harboring tumors that developed from cells transfected with the expression vector alone (left) or with the expression vector containing LOR-1 cDNA clone 24 (center) or 12 (right).
  • FIGS. 11 a - h illustrate fibrotic foci and collagen deposits in tumors derived from MCF-7 cells expressing recombinant LOR-1.
  • Hematoxilin-eosin staining demonstrate a few necrotic foci in a tumor derived from MCF-7 cells transfected with expression vector alone (FIG. 11 a , arrow, magnification ⁇ 20) and numerous necrotic foci in a tumor derived from MCF-7 cells transfected with expression vector containing LOR-1 cDNA clone 12 (FIG. 11 b , arrows, magnification ⁇ 20).
  • FIG. 11 c illustrates human keratin-7 immunostaining of tumors generated by MCF-7 cells transfected with expression vector containing clone 12. Counter staining was performed with hematoxylin-eosin (blue). Arrow points to nuclei of host cells concentrated in the fibrotic foci. No necrosis can be seen; magnification ⁇ 40.
  • FIGS. 11 d - f illustrate collagen deposits in tumor cells as viewed by Masson's Trichrome stain. A few collagen deposits are seen in tumors generated from MCF-7 cells transfected with the expression vector alone (FIG. 11 d , arrows, magnification ⁇ 200).
  • Thick collagen bundles are seen between tumor cells generated from MCF-7 cells transfected with expression vector containing clone 12 (FIG. 11 e , magnification ⁇ 200).
  • the fibrotic area is full with collagen fibers and interspaced with host derived cells in tumors generated from MCF-7 cells transfected with expression vector containing clone 24 (FIG. 11 f , magnification ⁇ 200).
  • FIGS. 11 g - h illustrate blood vessels stained with Masson's Trichrome in tumors generated from MCF-7 cells transfected with the expression vector alone (FIG. 11 g ) or expression vector containing clone 12 (FIG. 11 h ); magnification ⁇ 400.
  • FIGS. 12 a - d illustrate the deposition of collagen type-3 by reticulum stain in tumors generated from MCF-7 and C6 glioma cells transfected with expression vector alone (FIG. 12 a, c ) or vector expressing LOR-1 cDNA clone 12 (FIG. 12 b,d ); magnification ⁇ 200.
  • FIGS. 13 a - i illustrate the invasiveness of tumors derived from MCF-7 cells expressing recombinant LOR-1. Shown are histological sections of tumors generated from MCF-7 cells transfected with an expression vector alone (FIGS. 13 a, b ) or a vector expressing LOR-1 (FIGS. 13 c - h ). Sections were labeled with either a monoclonal antibody specific to human keratin-7 (FIGS. 13 a - d and f - h , blue purple stain) or with an antibody directed against LOR-1 (FIG. 13 e , red stain). Counterstain was with hematoxylin (light blue) in all sections.
  • the present invention is of pharmaceutical compositions and methods that can be used to modulate angiogenesis and to inhibit tumor invasiveness and tumor fibrosis. Specifically, the present invention can be used to suppress tumor growth and metastasis as well as to treat disorders such as, for example, arthritis, diabetic retinopathy, psoriasis and vasculitis.
  • PF4-LOR-1 binding assays Further support to the angiogenic activity of lysyl-oxidases is provided by the PF4-LOR-1 binding assays presented herein.
  • PF4 is an inhibitor of angiogenesis.
  • the anti-angiogenic activity exhibited by PF4 may be effected through LOR-1 inhibition, which, as demonstrated in the Examples section which follows, is highly expressed in the endothelial cells lining blood vessels.
  • the method is effected by administering into the mammalian tissue a molecule capable of modifying a tissue level and/or activity of at least one type of lysyl-oxidase to thereby modulate angiogenesis in the mammalian tissue.
  • tissue level refers to the level of lysyl-oxidase protein present in active form in the tissue at a given time point. Protein levels are determined by factors such as, transcription and/or translation rates, RNA or protein turnover and/or protein localization within the cell. As such any molecule which effects any of these factors can modify the tissue level of the lysyl-oxidase.
  • activity refers to an enzymatic activity of the lysyl-oxidase.
  • a molecule which can modify the enzymatic activity may directly or indirectly alter substrate specificity of the enzyme or activity of the catalytic site thereof.
  • lysyl-oxidase There are numerous examples of molecules which can specifically modify the tissue level and/or activity of a lysyl-oxidase. Such molecules can be categorized into lysyl-oxidase “upregulators” or “downregulators”.
  • An antibody polyclonal, monoclonal or monospecific
  • an antibody portion e.g., Fab fragment
  • an antibody or an antibody fragment directed at a lysyl-oxidase can be used to suppress or arrest the formation of blood vessels, and to inhibit tumor fibrosis and metastasis.
  • antibody inhibitors include inhibitors of angiogenesis which target angiogenic factors (Brooks et al., 1994; Brooks et al., 1998).
  • An antisense molecule which can be used with the present invention includes a polynucleotide or a polynucleotide analog of at least 10 bases, preferably between 10 and 15, more preferably between 50 and 20 bases, most preferably, at least 17, at least 18, at least 19, at least 20, at least 22, at least 25, at least 30 or at least 40 bases which is hybridizable in vivo, under physiological conditions, with a portion of a polynucleotide strand encoding a polypeptide at least 50% homologous to SEQ ID NO: 1, 4, 5 or 7 or at least 75% homologous to an N-terminal portion thereof as determined using the BestFit software of the Wisconsin sequence analysis package, utilizing the Smith and Waterman algorithm, where gap creation penalty equals 8 and gap extension penalty equals 2.
  • the antisense oligonucleotides used by the present invention can be expressed from nucleic acid construct administered into the tissue, in which case inducible promoters are preferably used such that antisense expression can be switched on and off, or alternatively such oligonucleotides can be chemically synthesized and administered directly into the tissue, as part of, for example, a pharmaceutical composition.
  • antisense oligonucleotides or analogs that bind target mRNA molecules prevent, by steric hindrance, binding of essential translation factors (ribosomes), to the target mRNA, a phenomenon known in the art as hybridization arrest, disabling the translation of such mRNAs.
  • the oligonucleotides or analogs must fulfill the following requirements (i) sufficient specificity in binding to the target sequence; (ii) solubility in water; (iii) stability against intra- and extracellular nucleases; (iv) capability of penetration through the cell membrane; and (v) when used to treat an organism, low toxicity.
  • oligonucleotide binding affinity can be predicted (Walton et al., 1999, Biotechnol Bioeng 65: 1-9; Matveeva et al., 1998, Nature Biotechnology 16, 1374-1375.) Unmodified oligonucleotides are typically impractical for use as antisense sequences since they have short in vivo half-lives, during which they are degraded rapidly by nucleases. Furthermore, they are difficult to prepare in more than milligram quantities. In addition, such oligonucleotides are poor cell membrane penetrants.
  • International patent application WO 89/12060 discloses various building blocks for synthesizing oligonucleotide analogs, as well as oligonucleotide analogs formed by joining such building blocks in a defined sequence.
  • the building blocks may be either “rigid” (i.e., containing a ring structure) or “flexible” (i.e., lacking a ring structure). In both cases, the building blocks contain a hydroxy group and a mercapto group, through which the building blocks are said to join to form oligonucleotide analogs.
  • the linking moiety in the oligonucleotide analogs is selected from the group consisting of sulfide (—S—), sulfoxide (—SO—), and sulfone (—SO2-).
  • Antisense oligonucleotides are typically synthesized in lengths of 13-30 nucleotides. The life span of oligonucleotide molecules in blood is rather short. Thus, they have to be chemically modified to prevent destruction by ubiquitous nucleases present in the body. Phosphorothioates are very widely used modification in antisense oligonucleotide ongoing clinical trials. A new generation of antisense molecules consist of hybrid antisense oligonucleotide with a central portion of synthetic DNA while four bases on each end have been modified with 2′O-methyl ribose to resemble RNA.
  • RNA oligonucleotides may also be used for antisense inhibition as they form a stable RNA-RNA duplex with the target, suggesting efficient inhibition.
  • RNA oligonucleotides are typically expressed inside the cells using vectors designed for this purpose. This approach is favored when attempting to target a mRNA that encodes an abundant and long-lived protein.
  • Ribozymes may also be used as down regulators. Ribozymes are being increasingly used for the sequence-specific inhibition of gene expression by the cleavage of mRNAs encoding proteins of interest. The possibility of designing ribozymes to cleave any specific target RNA has rendered them valuable tools in both basic research and therapeutic applications. In the therapeutics area, ribozymes have been exploited to target viral RNAs in infectious diseases, dominant oncogenes in cancers and specific somatic mutations in genetic disorders. Most notably, several ribozyme gene therapy protocols for HIV patients are already in Phase 1 trials (Welch et al., 1998).
  • ribozymes have been used for transgenic animal research, gene target validation and pathway elucidation.
  • Several ribozymes are in various stages of clinical trials.
  • ANGIOZYME was the first chemically synthesized ribozyme to be studied in human clinical trials.
  • ANGIOZYME specifically inhibits formation of the VEGF-r (Vascular Endothelial Growth Factor receptor), a key component in the angiogenesis pathway.
  • Ribozyme Pharmaceuticals, Inc. as well as other firms have demonstrated the importance of anti-angiogenesis therapeutics in animal models.
  • HEPTAZYME a ribozyme designed to selectively destroy Hepatitis C Virus (HCV) RNA, was found effective in decreasing Hepatitis C viral RNA in cell culture assays (Ribozyme Pharmaceuticals, Incorporated).
  • RNA interference an approach which utilizes small interfering dsRNA (siRNA) molecules that are homologous to the target mRNA and lead to its degradation (Carthew, 2001).
  • siRNA small interfering dsRNA
  • RNAi is an evolutionarily conserved surveillance mechanism that responds to double-stranded RNA by sequence-specific silencing of homologous genes (Fire et al., 1998; Zamore et al., 2000).
  • RNAi is initiated by the dsRNA-specific endonuclease Dicer, which promotes cleavage of long dsRNA into double-stranded fragments between 21 and 25 nucleotides long, termed small interfering RNA (siRNAs) (Zamore et al., 2000; Elbashir et al., 2001; Hammond et al., 2000; Bernstein et al., 2001). siRNA are incorporated into a protein complex that recognizes and cleaves target mRNAs (Nykanen et al., 2001).
  • siRNAs small interfering RNA
  • RNAi has been increasingly used for the sequence-specific inhibition of gene expression. The possibility of interfering with any specific target RNA has rendered RNAi a valuable tool in both basic research and therapeutic applications. RNAi was first used for gene silencing in nematodes (Fire et al., 1991;Guo & Kemphues, 1995).
  • RNAi has been utilized to inhibit expression of hepatitis C (McCaffrey et al., 2002), HIV-1 (Jacque et al., 2002), cervical cancer cells (Jiang and Milner 2002) and leukemic cells (Wilda et al., 2002).
  • siRNA molecules can be chemically synthesized as 21 to 25-nucleotide siRNA duplexes (Elbashir et al., 2001; McCaffrey et al., 2002).
  • Synthetic siRNA oligonucleotide duplexes can be prepared with either ribonucleotide 3′ overhangs or with deoxyribonucleotide 3′ overhangs (Hohjoh 2002). They can also be prepared as a sense-stranded DNA/antisense-stranded RNA hybrids or vise versa.
  • siRNA used by the present invention can be transcribed in vitro from plasmids and administered into the tissue. Transcripts that include two self-complementary siRNAs annealed to form a loop region can be further processed by single-stranded ribonucleases and/or other proteins into a functional duplex siRNA molecule (Leirdal and Sioud, 2002). siRNA can also be prepared from dsRNA by Escherichia coli RNase III cleavage into endoribonuclease-prepared siRNA (esiRNA).
  • siRNA molecules utilized by the present invention are preferably delivered into cell using retroviruses. Delivery of siRNA using retroviruses provides several advantages over methods, such as lipofection, since retroviral delivery is more efficient, uniform and immediately selects for stable “knock-down” cells (Devroe and Silver, 2002).
  • siRNA molecules of the present invention are preferably transcribed from expression vectors which can facilitate stable expression of the siRNA transcripts once introduced into a host cell.
  • These vectors are engineered to express small hairpin RNAs (shRNAs), which are processed in-vivo into siRNA molecules capable of carrying out gene-specific silencing (Brummelkamp, 2002; Paddison et al. 2002; Paul et al., 2002; Yu et al., 2002.
  • An example of a suitable expression vector is the pSUPERTM, which includes the polymerase-III H1-RNA gene promoter with a well defined start of transcription and a termination signal consisting of five thymidines in a row (T5) (Brummelkamp, 2002). Most importantly, the cleavage of the transcript at the termination site is at a site following the second uridine, thus yielding a transcript which resembles the ends of synthetic siRNAs, which also contain nucleotide overhangs.
  • siRNA is cloned such that it includes the sequence of interest, i.e., LOR-1 separated by a short spacer from the reverse complement of the same sequence. The resulting transcript folds back on itself to form a stem-loop structure, which mediates LOR-1 RNAi.
  • siRNA expression vector encodes the sense and antisense siRNA under the regulation of separate polIII promoters (Miyagishi and Taira (2002) Nature Biotech. 20:497-500).
  • the siRNA, generated by this vector also includes a 5 thymidine termination signal.
  • Oligoribonucleotide sequences of the present invention can also be placed under bi-directional promoters to produce both the sense and antisense transcripts from the same promoter construct, thus simplifying the construction of expression vectors and achieving an equal molar ratio of cellular sense and antisense sequences.
  • bi-directional promoters are disclosed in U.S. pat. application No. 20020108142.
  • VEGF vascular endothelial growth factor
  • VPF potent blood vessel permeabilizing factor
  • a nucleic acid construct including a constitutive, inducible or tissue specific promoter positioned upstream of a polynucleotide encoding a polypeptide having lysyl oxidase activity such as the polypeptide set forth in SEQ ID NO: 2, 3, 6, 8 or 9 can be administered into a mammalian tissue.
  • the lysyl oxidase expressed from this construct would substantially increase the levels of lysyl oxidase within the cells of the tissue and as such enhance angiogenesis.
  • Suitable mammalian expression vectors include, but are not limited to, pcDNA3, pcDNA3.1(+/ ⁇ ), pZeoSV2(+/ ⁇ ), pSecTag2, pDisplay, pEF/myc/cyto, pCMV/myc/cyto, pCR3.1, which are available from Invitrogen, pCI which is available from Promega, pBK-RSV and pBK-CMV which are available from Stratagene, pTRES which is available from Clontech, and their derivatives.
  • Gene “knock-in” techniques can also be used to increase the expression levels of the lysyl oxidase.
  • affinity binding assays and/or activity assays can be used to screen for novel compounds (e.g., substrate analogs) which can specifically regulate activity of lysyl oxidase and as such can be used with the present invention.
  • the present invention also provides a method of inhibiting metastasis and/or fibrosis in a mammalian tissue.
  • the method is effected by administering to the mammalian tissue a molecule capable of downregulating a tissue level and/or activity of at least one type of lysyl-oxidase.
  • the method of the present invention can be used to treat human patients that have been diagnosed with cancerous tumors, by administering any of the downregulating molecules described herein above, in order to reduce the tissue level and/or activity of at least one type of a lysyl oxidase.
  • administering refers to all modes of administration described hereinbelow with respect to the pharmaceutical compositions of the present invention.
  • Administration can also be effected in a systemic manner in order to treat the affected tissue, i.e., the tissue where the cancerous tumor was formed and where metastases are present or likely to be formed with tumor progression.
  • the present invention also provide a method of identifying molecules capable of inhibiting metastasis and/or fibrosis.
  • This method is effected by screening and identifying molecules which exhibit specific reactivity with at least one type of lysyl-oxidase and testing a metastasis and/or fibrosis inhibitory potential of these molecules.
  • lysyl-oxidase Numerous types of molecules can be screened for reactivity with at least one type of lysyl-oxidase, examples include, but are not limited to, molecules such as antisense oligonucleotides, siRNA and ribozymes that interact with a polynucleotide expressing a lysyl oxidase activity or molecules such as antibodies that interact with polypeptides having a lysyl oxidase activity. In addition, short peptides and other small molecules can also be screened by this method of the present invention.
  • Screening for cross reactivity can be effected by lysyl-oxidase enzymatic activity assays, by binding assays and the like.
  • suitable assays are provided in Rodriguez et al., 2002, Arterioscler Thromb Vasc Biol 22: 1409-14; Wilson and Nock, 2002, Curr Opin Chem Biol 6: 81-5; Uetz, 2002, Curr Opin Chem Biol 6: 57-62; Stoll et al., 2002, Front Biosci 2002 7: c13-32).
  • Testing a metastatic phenotype of transformed tumor cells can be performed in-vitro since nearly all steps of the metastatic process, including attachment, matrix degradation and migration, can be modeled experimentally in-vitro by measuring invasion of a reconstituted basement membrane (RBM).
  • RBM reconstituted basement membrane
  • Metastatic invasiveness of tumor cell can be modeled by migration of tumor cells into reconstituted basement membrane (RBM) in the presence and absence of a chemoattractant, such as fibroblast conditioned medium (FCM).
  • FCM fibroblast conditioned medium
  • in-vitro metastasis events correspond to steps observed in the metastatic spread of tumor cells through the basement membrane in-vivo
  • in-vitro invasiveness of cells can be assayed by the methods described in Albini et al., 1987 Cancer Research 47: 3239-3245, which is incorporated herein by reference in its entirety.
  • Invasiveness assays and other methods for assessing metastatic affects are described in Leyton et al., 1994 Cancer Research 54:3696-3699, which is incorporated by reference herein in its entirety.
  • Reconstituted basement membrane preparations for use in accordance with the hereinabove described assays are readily available from numerous commercial suppliers.
  • One suitable example membrane in this regard is “MATRIGEL” available from Collaborative Biomedical Products of Bedford, Mass.
  • In-vitro evaluation of tumor cell metastatic phenotype can also be effected by determining level and pattern of expression of one or more metastasis associated markers such protease markers, which are considered to be an integral part of tumor metastasis (see U.S. Pat. No.: 6,303,318).
  • One example is the arachidonic acid, the release of which in cells can serve to indicate metastatic potential of a tumor (U.S. Pat. No: 6,316,416).
  • determining phospholipase A-2 (PLA 2 ) activity, and the activity or abundance of factors that affect the activity of PLA 2 , such as uteroglobin protein (U.S. Pat. No.: 6,316,416) can serve as an indication of metastatic potential.
  • Determining pattern and level of expression of metastasis-associated markers can be effected by one of several methods known in the art.
  • the presence or level of proteins indicative of metastatic potential of tumors can be determined in cells by conventional methods well known to those of skill in the art. For instance, the techniques for making and using antibody and other immunological reagents and for detecting particular proteins in samples using such reagents are described in current protocols in immunology, Coligan et al., Eds., John Wiley & Sons, New York (1995), which is incorporated by reference herein in parts pertinent to making and using reagents useful for determining specific proteins in samples. As another example, immunohistochemical methods for determining proteins in cells in tissues are described in Volume 2, Chapter 14 of current protocols in molecular biology, Ausubel et al., Eds., John Wiley & Sons, Inc.
  • Metastatic potential can also be determined in vivo at the mRNA level.
  • the presence and/or level of mRNA transcripts can be determined by a variety of methods known to those of skill in the art.
  • a given mRNA may be detected in cells by hybridization to a specific probe.
  • Such probes may be cloned DNAs or fragments thereof, RNA, typically made by in-vitro transcription, or oligonucleotide probes, usually generated by solid phase synthesis. Methods for generating and using probes suitable for specific hybridization are well known and used in the art.
  • a “pharmaceutical composition” refers to a preparation of one or more of the active ingredients described herein with other chemical components such as physiologically suitable carriers and excipients.
  • the purpose of a pharmaceutical composition is to facilitate administration/targeting of a compound to a mammal.
  • active ingredients refers to the preparation accountable for the biological effect, i.e. the upregulator/downregulator molecules used by the present invention to modulate angiogenesis and the downregulators molecules used by the present invention to inhibit metastasis and tumor fibrosis.
  • physiologically acceptable carrier and “pharmaceutically acceptable carrier” are interchangeably used to refer to a carrier, such as, for example, a liposome, a virus, a micelle, or a protein, or a diluent which do not cause significant irritation to the mammal and do not abrogate the biological activity and properties of the active ingredient.
  • a carrier such as, for example, a liposome, a virus, a micelle, or a protein, or a diluent which do not cause significant irritation to the mammal and do not abrogate the biological activity and properties of the active ingredient.
  • An adjuvant is included under these phrases.
  • excipient refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient.
  • excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
  • compositions may be found in “Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, Pa., latest edition, which is incorporated herein by reference.
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, transnasal, intestinal or parenteral delivery, including intramuscular, subcutaneous and intramedullary injections as well as intrathecal, direct intraventricular, intravenous, inrtaperitoneal, intranasal, or intraocular injections.
  • the active ingredients of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological salt buffer.
  • physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological salt buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • the compounds can be formulated readily by combining the active ingredient with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the active ingredient of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient.
  • Pharmacological preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores.
  • compositions which can be used orally, include push-fit capsules made of gelatin as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules may contain the active ingredients in admixture with filler such as lactose, binders such as starches, lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active ingredients may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for the chosen route of administration.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • compositions described herein may be formulated for parenteral administration, e.g., by bolus injection or continuos infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers with optionally, an added preservative.
  • the compositions may be suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions for parenteral administration include aqueous solutions of the active preparation in water-soluble form. Additionally, suspensions of the active ingredients may be prepared as appropriate oily or water based injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acids esters such as ethyl oleate, triglycerides or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the active ingredients to allow for the preparation of highly concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water based solution, before use.
  • a suitable vehicle e.g., sterile, pyrogen-free water based solution
  • the pharmaceutical composition may form a part of an article of manufacturing which also includes a packaging material for containing the pharmaceutical composition and a leaflet which provides indications of use for the pharmaceutical composition.
  • the present invention provides a method and pharmaceutical compositions useful modulating angiogenesis.
  • Such modulation activity can be used to treat arthritis (Koch, 1998; Paleolog and Fava, 1998), diabetic retinopathy (Miller et al., 1997), psoriasis (Detmar et al., 1994; Creamer et al., 1997) and vasculitis (Lie, 1992; Klipple and Riordan, 1989).
  • the present invention can also be used to treat disease characterized by fragile blood vessels, including Marfans syndrom, Kawasaki, Ehlers-Danlos, cutis-laxa, and takysu (Lie, 1992; Klipple and Riordan, 1989; Brahn et al., 1999; Cid et al., 1993; Hoffman et al., 1991).
  • the present invention can also be used to treat diseases which are characterized by changes in the wall of blood vessels. For example, restenosis which is a common complication following balloon therapy, Fibromuscular dysplasia (Begelman, and Olin, 2000) and aortic stenosis (Palta et al., 2000) are all potentially treatable by the method of the present invention.
  • diseases which are characterized by changes in the wall of blood vessels. For example, restenosis which is a common complication following balloon therapy, Fibromuscular dysplasia (Begelman, and Olin, 2000) and aortic stenosis (Palta et al., 2000) are all potentially treatable by the method of the present invention.
  • LOR-1 is more highly expressed in metastatic tumors and cell lines than in non-metastatic tumors and cell lines (FIGS. 3, 9, and 13 ). This suggests that levels of LOR-1 expression can be used as a diagnostic tool to determine the malignancy of cancer cells, as well as, to determine and implement suitable treatment regimens.
  • LOR-1 expression vector transfection into MCF-7 cells, and expression:
  • the LOR-1 cDNA (SEQ ID NO: 1) was cloned into a pCDNA3.1-hygro expression vector (Invitrogen Inc., USA) under the control of a CMV promoter. Clones of cells expressing LOR-1 were selected using hygromycine and assayed for LOR-1 expression using the polyclonal antisera described below.
  • PF4 was coupled to sepharose using a modification of the method of Miron and Wilchek (Wilchek and Miron, 1982) as previously described for vascular endothelial growth factor (Soker et al., 1998).
  • Serum free conditioned medium was collected from 35 S-methionine labeled MCF-7 cells which over-expressed LOR-1. The conditioned medium was passed through the column twice. The column was washed with phosphate buffered saline (300 mM NaCl, pH-7.2) and eluted with PBS (containing 2M NaCl).
  • In-situ hybridization Fragments encompassing nucleotides 922-1564 of LOR-1, nucleotides 976-1391 of LOL, nucleotides 400-950 of LO and nucleotides 1061-1590 of LOR-2 (as numbered from the ATG codon of these sequences) where each independently subcloned into the Bluescript SK and KS (Stratagene) vectors.
  • a DIG cRNA labeling kit of Boehringer-Mannheim was used to transcribe sense (s) and antisense (as) digoxigenin-labeled cRNA probes from the T7 promoter of the Bluescript constructs. Hybridization and subsequent detection of hybridized probes was carried out essentially as previously described (Cohen et al., 2001).
  • Anti-LOR-1 polyclonal antisera Antisera was generated by injecting a recombinant peptide containing the C-terminal 200 amino-acids of LOR-1 (amino acids 540-744 of SEQ ID NO: 2) into female rabbits. Serum was collected 10 days following each injection and an immunoglobulin fraction was purified using a protein A Sepharose affinity column (Pharmacia).
  • LOR-1 purification A PF4 affinity column was used to detect endothelial cell proteins which specifically interact with PF4.
  • PF4 binding proteins were detected in conditioned medium of human umbilical vein endothelial cells (HUVEC), whereas PF4 binding proteins were not detected in detergent extracts of endothelial cells.
  • LOR-1 The full length amino acid sequence of LOR-1 (as deduced from the isolated cDNA sequence) displayed a high degree of identity to WS9-14, a protein over expressed in senescent fibroblasts, in several types of adherent cells (but not in non-adherent cells) and in fibroblasts, in which it was correlated to pro-collagen I-a1 expression levels, as well as being induced by TGF- ⁇ and inhibited by phorbol esters and retinoic acid (Saito et al., 1997).
  • LOR-1 Recombinant LOR-1 expressed in PAE cells specifically bound with the PF4 affinity column (FIG. 1). Since LOR-1 is a member of the LO family it was hypothesized that it participates in ECM formation during angiogenesis. Furthermore it was also hypothesized that PF4 suppresses or inhibits the pro-angiogenic activity of LOR-1 thus inhibiting the later stages of blood vessel formation and as a result limiting tumor growth.
  • LOR-1 and cancer as shown in FIG. 2, a direct correlation between the expression levels of LOR-1 and the metastatic properties of breast cancer derived cell lines was demonstrated herein.
  • LOR-1 cDNA was expressed in non-metastatic breast cancer derived MCF-7 cell lines which do not normally express LOR-1.
  • the expression of LOR-1 was examined using rabbit polyclonal antibodies generated as described above (FIG. 3).
  • a control cell line which was transfected with an empty expression vector, and an MCF-7 cell line expressing LOR-1 were implanted under the skin of immune deficient mice along with an estrogen slow release pellet as described above. Estrogen was added since the development of tumors from this non-metastatic cell line is estrogen dependent (Mcleskey et al., 1993).
  • LOR-1 cDNA was placed under the control of a tetracycline induced promoter (The TET-off system). Such a construct will enable to determine conclusively whether the reduced rate of tumor growth observed in LOR-1 expressing cells is indeed caused by LOR-1.
  • LOR-1 The role of LOR-1 in Wilson's disease and in other chronic liver diseases: Normal and diseased liver tissue were probed with LOR-1 sense (FIGS. 6 a and 6 c ) and antisense probes (FIGS. 6 b and 6 d ). Normal liver tissues expresses very low levels of LOR-1 (FIG. 6 b ). However, fibrotic liver tissues such as those observed in Wilson's disease, exhibit a strong increase in hepatocyte expression of LOR-1 (FIG. 6 d ).
  • the Lysyl oxidase family A homology comparison between five members of the lysyl oxidase family which includes the LO and LOL subfamily and the LOR-1 and LOR-2 subfamily revealed a strong homology at the C-terminal portion which includes the conserved lysyl oxidase motif.
  • LOR-1 and LOR-2 are characterized by long N-terminal stretches which are not found in LO and LOL.
  • Estrogen pellets (17 ⁇ -estradiol, 0.72 mg/pellet, 60-days release) were from Alternative Research of America, FL, USA. Masson Trichrome stain kit was purchased from Bio-Optica (Milano, Italy), Reverse Transcriptase, G418 were from GIBCO BRL (U.K.), Hygromycin B, Tetracycline hydrochloride, Reticulum stain kit fast green and Sirius red (direct red 80) were from Sigma (USA) , Restriction enzymes, T4 ligase were from New England Biolabs (USA).
  • MCF-7 breast cancer cells were kindly provided by Dr. Hadasa Degani (Weizmann Institute, Israel).
  • the MDA-MB-435 breast cancer cell line was kindly provided by Dr. Israel Vlodavsky (Technion, Israel).
  • the MDA-MB-23 1 cells were kindly provided by Dr. Michael Klagsbrun (Harvard University, USA).
  • FCS Fetal Calf Serum
  • Tissue culture media, sera, and cell culture supplements were from Beth-Haemek Biological Industries, Israel, or from Gibco-BRL.
  • MCF-7 TetOff cells (Clontech, USA) containing the tetracycline trans-activator (tTA) were grown in DMEM medium containing 10% Tet system approved fetal calf serum (Clontech), in the presence of 100 ⁇ g/ml G418, 150 ⁇ g/ml Hygromycin B and 1 ⁇ g/ml Tetracyclin.
  • LOR1 and LOR2 cDNAs Total RNA (4 ⁇ g) from HUVEC cells (for LOR1) or melanoma cells (for LOR2) was reversed transcribed using MMLV reverse transcriptase (GIBCO BRL) as described (Chomczynski and Sacchi, 1987).
  • the LOR-1 and LOR-2 cDNAs were amplified using the Expand Long High Fidelity PCR system (ROCHE) and the following pairs of amplification primers: For LOR-1 SEQ ID NOs: 10 and 11, and for LOR-2 SEQ ID NOs: 12 and 13.
  • ROCHE Expand Long High Fidelity PCR system
  • LOR-1 SEQ ID NO: 1
  • SEQ ID NO: 4 The 2.3 kb cDNA of LOR-1 (SEQ ID NO: 1) and the 2.26 KB of LOR-2 (SEQ ID NO: 4) were subcloned into the pGEM-T Easy vector (Promega) by T-A cloning.
  • a 613 bp LOR1 cDNA fragment (SEQ ID NO: 14) was digested with the Sph-I and Hind III restriction enzymes and ligated into the bacterial expression vector pQE-30, which added an in-frame sequence encoding a six histidine (6 ⁇ His) tag to the 5′ end of the insert.
  • the resulting plasmid was used to produce a recombinant, 6 ⁇ His tagged 23 kDa peptide (SEQ ID NO: 17).
  • the peptide was purified from bacterial cell extracts using nickel affinity chromatography, and further purified using SDS-PAGE.
  • the gel was electroblotted onto nitrocellulose and the band containing the peptide was cut out from the blot, solubilized in DMSO, and used to immunize rabbits.
  • Antiserum was affinity purified on protein-A sepharose followed by affinity purification on a column to which the recombinant peptide was coupled using a previously described method (Wilchek and Miron, 1982). The antibody was eluted from the column using 0.1 M glycine at pH 3.
  • Hygro plasmid or pcDNA-LOR1 plasmid (10-20 ⁇ g) were stably transfected into MCF-7 cells using electroporation with a BioRad gene pulser (960 ⁇ F, 0.28 V). Stable transfectants were selected using 300 ⁇ g/ml hygromycin B. Clones expressing recombinant LOR1 were obtained in two consecutive stable transfections and screened for LOR1 expression using our anti-LOR-1 polyclonal antibodies. Conditioned medium was collected after 48 hours from transfected cells and LOR-1 expression was monitored using western blot is analysis (FIG. 10A).
  • LOR1 cDNA SEQ ID NO: 1
  • pTET-Splice vector Clontech
  • Tet off system tetracycline
  • the pTET-Splice plasmid DNA was digested with Hind III and SpeI and ligated to the 2.3 kb HLOR1 cDNA fragment which was rescued out of the pCDNA-LOR1 plasmid using Hind III and XbaI.
  • the resultant plasmid which was designated as pTET-LOR1, was co-transfected into MCF-7 TetOff cells along with pTK-Hygro at a ratio of 20:1 respectively.
  • LOR-1 expressing cells were selected in medium containing 100 ⁇ g/ml G418, 150 ⁇ g/ml hygromycin B and 1 ⁇ g/ml tetracycline.
  • Stable transfectants were screened for inducible expression of LOR1 using western blot analysis 48 hours following removal of the tetracycline from the growth media.
  • the clone having the highest induction levels in the absence of tetracycline and lowest basal expression levels in the presence of tetracycline was selected and designated MCF-7/Tet-LOR1.
  • C6 glioma cells were transfected and screened for LOR1 expression as described above.
  • Protein blot analysis Serum free conditioned media (40 ⁇ l) was separated on a 8% SDS-PAGE gel and the proteins were electroblotted onto a nitrocellulose filter using semi-dry electroblotting. The filter was blocked for 1 hour at room temperature with TBST buffer containing 10 mM Tris-HCl (pH-7.0), 0.15 M NaCl, and 0.3% Tween-20 supplemented with 10% low fat milk. The filter was incubated over night at 4° C. with affinity purified rabbit anti-LOR1 polyclonal antibody in TBST (1:2500).
  • the blot was subsequently washed 3 times in TBST and incubated with goat anti-rabbit IgG peroxidase-conjugated secondary antibodies for 1 hour at room temperature. Bound antibody was visualized using the ECL detection system (Biological Industries, Israel).
  • the sections were then washed 3 times with TBST, and secondary detection was applied using anti FITC alkaline phosphates-conjugate (Roche) at a 1:200 dilution (for anti cytokeratin) or DAKO Envision detection system (anti rabbit or anti mouse—HRP).
  • Sections were developed in 3-amino-9-ethylcarbazole (AEC, DAKO) solution, counterstained by Hematoxylin, and photographed under a microscope. In control experiments the primary antibodies were omitted.
  • Masson Trichrome and reticulum stains were according to manufacture's protocol. For Sirius red stain sections were incubated for 5 min. with 0.03% (w/v) fast green solution, rinsed twice with 1% acetic acid, incubated for 15 min. in 0.1% Sirius red solution, rinsed as above, dehydrated and examined under polarizing light microscope.
  • LOR-1 is expressed in highly metastatic breast cancer derived cell types but not in non-metastatic MCF-7 cells: Desmoplasia and formation of fibrotic foci in breast cancer tumors is associated with the transition from a localized, relatively benign tumor to an invasive/metastatic tumor (Colpaert et al., 2001; Hasebe et al., 2000). Lysyl-oxidases contribute to the deposition of collagen by covalently cross-linking collagen monomers (Smith-Mungo and Kagan, 1998). To find out whether expression of lysyl-oxidases is associated with the invasive/metastatic phenotype several human breast cancer derived cell types have been screened for the expression of lysyl-oxidases.
  • both LOR-1 expressing cells displayed extra protein bands of about 70 kDa (FIGS. 10 a, b ) suggesting that LOR-1 may undergo proteolytic processing like other members of the lysyl-oxidase family (Borel et al., 2001; Panchenko et al., 1996).
  • LOR-1 cDNA has been expressed in MCF-7 cells under the control of a Tetracycline inducible promoter (Shockett and Schatz, 1996).
  • LOR-1 producing cells and cells transfected with expression vector alone were pre-implanted subcutaneously in nude mice (described above). Interestingly, the growth rate of tumors containing LOR-1 expressing cells was retarded as compared with that of tumors developed from parental cells (not shown) or cells transfected with the expression vector alone (FIGS. 10 c, d ).
  • Tumors that develop from LOR-1 producing MCF-7 cells contain many necrotic and fibrotic foci rich in collagen deposits: Hematoxilin-eosin staining of tumor sections revealed major necrosis in tumors generated from LOR-1 expressing MCF-7 cells (FIG. 11 b ) and only a few necrotic areas in tumors generated from parental (not shown) or MCF-7 cells transfected with expression vector alone (FIG. 11 a ).
  • LOR-1 expressing tumors also contained extensive fibrotic areas mainly composed of host derived cells such as mouse fibroblasts rather than MCF-7 cells. These cells are easily distinguishable from the host cells since they do not react with an antibody against human keratin 7 (FIG. 11 c ).
  • Tumors that developed from parental or MCF-7 cells transfected by an expression vector alone contained limited amounts of collagen that was scattered between the tumor cells (FIG. 11 d , arrows).
  • tumors that developed from clone 12 or clone 24 LOR-1 expressing MCF-7 cells contained denser deposits of collagen between the tumor cells (FIG. 11 e ).
  • the fibrotic and necrotic areas in these tumors appeared choke full with collagen fibers (FIG. 11 f ).
  • the blood vessels within tumors that developed from vector-transfected MCF-7 cells remained almost unstained (FIG. 11 g , arrows)
  • the blood vessels in tumors derived from LOR-1 expressing MCF-7 cells were sheathed by a thick layer of collagen (FIG. 11 h , arrows).
  • VEGF Vascular endothelial growth factor
  • Integrin alpha(v)beta(3) antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels. Cell 79, 1157-1164.
  • Neuropilin-1 is expressed by endothelial and tumor cells as an isoform specific receptor for vascular endothelial growth factor. Cell 92, 735-745.
  • RNAi double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals.
  • RNA interference is mediated by 21- and 22-nucleotide RNAs. Genes Dev. 15, 188-200.
  • RNA interference RNA interference

Abstract

Methods and compositions suitable for modulating angiogenesis in a mammalian tissue are provided. Further provided are methods suitable for inhibiting metastasis and fibrosis in a mammalian tissue.

Description

  • This is a Continuation-In-Part of PCT/IL01/00728, filed Aug. 7, 2001, which claims the benefit of priority from U.S. Provisional Patent Application No. 60/223,739, filed Aug. 8, 2000.[0001]
  • FIELD AND BACKGROUND OF THE INVENTION
  • The present invention relates to pharmaceutical compositions and methods useful for modulating angiogenesis and for inhibiting metastasis and fibrosis in a mammalian tissue. [0002]
  • Angiogenesis [0003]
  • In an adult, formation of new blood vessels in normal or diseased tissues is regulated by two processes, recapitulated vasculogenesis (the transformation of pre-existing arterioles into small muscular arteries) and angiogenesis, the sprouting of existing blood vessels (which occurs both in the embryo and in the adult). [0004]
  • The process of angiogenesis is regulated by biomechanical and biochemical stimuli. Angiogenic factors such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) are released by vascular cells, macrophages, and cell surrounding blood vessels. These angiogenic factors activate specific proteases that are involved in degradation of the basement membrane. As a result of this degradation, vascular cells migrate and proliferate thus leading to new blood vessel formation. Peri-endothelial cells, such as pericytes in the capillaries, smooth muscle cells in larger vessels and cardiac myocytes in the heart are recruited to provide maintenance and modulatory functions to the forming vessel. [0005]
  • The establishment and remodeling of blood vessels is controlled by paracrine signals, many of which are mediated by protein ligands which modulate the activity of transmembrane tyrosine kinase receptors. Among these molecules are vascular endothelial growth factor (VEGF) and its receptor families (VEGFR-1, VEGFR-2, neuropilin-1 and neuropilin-2), Angiopoietins 1-4 (Ang-1, Ang-2 etc.) and their respective receptors (Tie-1 and Tie-2), basic fibroblast growth factor (bFGF), platelet derived growth factor (PDGF), and transforming growth factor β (TGF-β). [0006]
  • The growth of solid tumors is limited by the availability of nutrients and oxygen. When cells within solid tumors start to produce angiogenic factors or when the levels of angiogenesis inhibitors decline, the balance between anti-angiogenic and angiogenic influences is perturbed, initiating the growth of new blood vessels from the existing vascular bed into the tumor. This event in tumor progression is known as the angiogenic switch (Folkman, 1990; Hanahan and Folkman 1996). It had been demonstrated that inhibitors of tumor angiogenesis are able to completely inhibit tumor growth in mice (Boehm et al., 1997; Bergers et al., 1999) and also inhibit tumor metastasis, a process that relies upon close contact between the vasculature and tumor cells (Zetter, 1998). It has also been demonstrated that angiogenesis plays an important role in the progression of breast cancer (Weidner, N. (1998; Degani et al., 1997; Guidi et al., 1997; Balsari et al., 1999). [0007]
  • Such findings have prompted the use of known anti-angiogenic factors in breast cancer therapy (Klauber et al., 1997; Harris et al., 1996; Weinstatsaslow et al., 1994) and a search for novel angiogenesis inhibitors. [0008]
  • During the past decade several novel inhibitors of angiogenesis have been isolated including inhibitors of VEGF signaling (Neufeld et al., 1999) and inhibitors of processes which lead to the maturation and stabilization of new blood vessels. Anti-integrin antibodies have been used as inhibitors of blood vessel maturation (Brooks et al., 1994; Brooks et al., 1998). [0009]
  • Although several anti-angiogenic drugs are now available commercially, the anti-angiogenic mechanisms of most of these drugs (e.g., angiostatin and endostatin) remain unclear (O'Reilly et al., 1997; Oreilly et al., 1996). [0010]
  • Since angiogenesis can be initiated by many (possibly compensatory) angiogenic factors it stands to reason that anti-angiogenic factors which target later processes in the angiogenic response such as vessel maturation or a combination of anti-angiogenic factors would be most effective in arresting vessel formation. [0011]
  • Platelet factor-4 (PF4) is an anti-angiogenic protein normally sequestered in platelets (Tanaka et al., 1997; Maione et al., 1990; Neufeld et al., 2000). PF4 inhibits angiogenesis using poorly defined mechanisms (Gengrinovitch et al., 1995; Brown, and Parish, 1994; Gupta, and Singh, 1994; Watson et al., 1994). It was previously speculated that PF4 binds to cell surface heparan-sulfate proteoglycans and in this manner inhibits the activity of angiogenic growth factors such as basic fibroblast growth factor (Watson et al., 1994). [0012]
  • While reducing the present invention to practice and while searching for alternative anti-angiogenic factors or targets, the present inventors have uncovered a novel PF4 binding protein which participates in modulating anglogenesis. [0013]
  • Tumor Metastasis [0014]
  • The transition from a localized tumor to an invasive and metastatic tumor represents a landmark in the development of malignant disease, since it is usually associated with a markedly worse prognosis. The understanding of the processes that govern this transition is therefore of prime importance. [0015]
  • In breast cancer, the transition from a localized to an invasive/metastatic tumor is associated in many cases with the formation of fibrotic foci and desmoplasia, which is the presence of unusually dense collagenous stroma, within the primary tumor (Colpaert et al., 2001; Hasebe et al., 2000). A similar correlation may exist in other types of cancers such as colon and pancreatic cancers (Nishimura et al., 1998; Ellenrieder et al., 2000). These observations represent apparent paradoxes at first glance, since invasiveness has long been associated with the destruction of extracellular matrix by extracellular matrix degrading enzymes like metalo-proteases (Stamenkovic, 2000; Duffy et al., 2000) and heparanase (Vlodavsky and Friedmann, 2001). However, it is possible that deposition of excess extracellular matrix may stimulate in turn expression of matrix degrading enzymes that will contribute under certain circumstances to tumor invasion. In fact, there is some evidence that an increase in extracellular matrix deposition can indeed influence the production of extracellular matrix degrading enzymes (Schuppan et al., 2001; Swada et al., 2001). [0016]
  • Several prior art studies have attempted to develop agents to treat breast cancer metastases (Sauer et al., 2002) including a study by Kim et al., (2000) that described apicidin [cyclo (N-O-methyl-L-tryptophanyl-L-isoleucinyl-D-pipecolinyl-L-2-amino-8-oxodecanoyl)], a fungal metabolite that was identified as an antiprotozoal agent known to inhibit parasite histone deacetylase (HDAC), that can inhibit the H-ras-induced invasive phenotype of MCF10A human breast epithelial cells. Another agent is the polymeric form of fibronectin that was shown to reduce tumor growth and to posses antimetastatic activity when administered systemically to tumor-bearing mice (Yi and Ruoslahti, 2001). [0017]
  • While reducing the present invention to practice, the present inventors have also discovered that the lysyl oxidase protein, LOR-1, participates in metastasis and as such can be used as a target for inhibiting metastasis and reducing tumor invasiveness. [0018]
  • SUMMARY OF THE INVENTION
  • According to one aspect of the present invention there is provided a method of modulating angiogenesis in a mammalian tissue, the method comprising administering into the mammalian tissue a molecule capable of modifying a tissue level and/or activity of at least one type of lysyl-oxidase to thereby modulate angiogenesis in the mammalian tissue. [0019]
  • According to another aspect of the present invention there is provided a method of modulating angiogenesis in a mammalian tissue, the method comprising administering into the mammalian tissue a nucleic acid construct being capable of expressing a polypeptide having lysyl-oxidase activity to thereby modulate angiogenesis within the mammalian tissue. [0020]
  • According to yet another aspect of the present invention there is provided a pharmaceutical composition useful for modulating angiogenesis in mammalian tissue comprising, as an active ingredient, a molecule capable of modifying a level and/or activity of at least one type of lysyl-oxidase of the mammalian tissue and a pharmaceutically effective carrier. [0021]
  • According to another aspect of the present invention there is provided a method of modulating angiogenesis in a mammalian tissue, the method comprising administering into the mammalian tissue a molecule capable of modifying a tissue level and/or activity of a polypeptide at least 75% homologous to the polypeptide set forth in SEQ ID: 2 or 9 to thereby modulate angiogenesis within the mammalian tissue. According to further features in preferred embodiments of the invention described below, the molecule is an antibody or an antibody fragment capable of binding with, and at least partially inhibiting the activity of, the at least one polypeptide. [0022]
  • According to still further features in the described preferred embodiments the antibody or the antibody fragment is directed against at least a portion of the polypeptide set forth in SEQ ID NO: 2, 3, 6, 8 or 9. [0023]
  • According to still further features in the described preferred embodiments the molecule is a polynucleotide capable of down regulating expression of the at least one type of lysyl-oxidase. [0024]
  • According to still further features in the described preferred embodiments the polynucleotide is at least partially complementary with the polynucleotide set forth in SEQ ID NO: 1, 4, 5 or 7. [0025]
  • According to still further features in the described preferred embodiments the molecule is a polypeptide having lysyl-oxidase activity. [0026]
  • According to still further features in the described preferred embodiments the polypeptide is as set forth in SEQ ID NO: 2, 3, 6, 8 or 9. [0027]
  • According to another aspect of the present invention there is provided a method of modulating angiogenesis in a mammalian tissue, the method comprising administering into the mammalian tissue a nucleic acid construct being capable of expressing a polypeptide having lysyl-oxidase activity to thereby modulate angiogenesis within the mammalian tissue. [0028]
  • According to still further features in the described preferred embodiments the polypeptide is at least 75% homologous to the polypeptide set forth in SEQ ID NO: 2, 3, 6, 8 or 9. [0029]
  • According to still another aspect of the present invention there is provided method of identifying molecules capable of modulating angiogenesis, the method comprising: (a) isolating molecules which exhibit specific reactivity with at least one type of lysyl-oxidase; and (b) testing the molecules within mammalian tissue so as to determine the angiogenesis modulation activity thereof. [0030]
  • According to still further features in the described preferred embodiments step (a) is effected by binding assays and/or lysyl-oxidase activity assays. [0031]
  • According to an additional aspect of the present invention there is provided method of determining the malignancy of cancerous tissue, the method comprising (a) determining a lysyl-oxidase expression level and/or activity of the cancerous tissue; and (b) comparing the lysyl-oxidase expression level and/or activity with that determined for control tissue to thereby determine the malignancy of the cancerous tissue. [0032]
  • According to another aspect of the present invention there is provided method of inhibiting metastasis and fibrosis in a mammalian tissue, the method comprising administering into the mammalian tissue a molecule capable of downregulating a tissue level and/or activity of at least one type of lysyl-oxidase to thereby inhibit metastasis in the mammalian tissue [0033]
  • According to still another aspect of the present invention there is provided a pharmaceutical composition useful for inhibiting metastasis and fibrosis in mammalian tissue comprising, as an active ingredient, a molecule capable of downregulating a level and/or activity of at least one type of lysyl-oxidase of the mammalian tissue and a pharmaceutically effective carrier. [0034]
  • According to further features in preferred embodiments of the invention described below, the molecule is an antibody or an antibody fragment capable of binding with, and at least partially inhibiting the activity of, the at least one polypeptide. [0035]
  • According to still further features in the described preferred embodiments the antibody or the antibody fragment is directed against at least a portion of the polypeptide set forth in SEQ ID NO: 2, 3, 6, 8 or 9. [0036]
  • According to still further features in the described preferred embodiments the molecule is a polynucleotide capable of downregulating expression of the at least one type of lysyl-oxidase. [0037]
  • According to still further features in the described preferred embodiments the polynucleotide is at least partially complementary with the polynucleotide set forth in SEQ ID NO: 1, 4, 5 or 7. [0038]
  • According to still another aspect of the present invention there is provided method of identifying molecules capable of inhibiting metastasis and fibrosis, the method comprising: (a) screening and identifying molecules which exhibit specific reactivity with at least one type of lysyl-oxidase; and (b) testing the metastasis and fibrosis inhibitory potential of the said molecules. [0039]
  • According to still further features in the described preferred embodiments step (a) is effected by binding assays and/or lysyl-oxidase activity assays. [0040]
  • According to yet another aspect of the present invention there is provided a method for inhibiting metastasis and fibrosis in a mammalian tissue, the method comprising administering to the mammalian tissue a molecule capable of downregulating a tissue level and/or activity of a polypeptide at least 75% homologous to the polypeptide set forth in ID NO: 2 or 9, to thereby inhibit metastasis and fibrosis in a mammalian tissue. [0041]
  • The present invention successfully addresses the shortcomings of the presently known configurations by providing pharmaceutical compositions and methods that can be used to treat metastatic cancer, formation of tumor fibrosis and other disorders characterized by excessive or insufficient blood vessel formation. [0042]
  • Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. In case of conflict, the patent specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.[0043]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The invention is herein described, by way of example only, with reference to the accompanying drawings. With specific reference now to the drawings in detail, it is stressed that the particulars shown are by way of example and for purposes of illustrative discussion of the preferred embodiments of the present invention only, and are presented in the cause of providing what is believed to be the most useful and readily understood description of the principles and conceptual aspects of the invention. In this regard, no attempt is made to show structural details of the invention in more detail than is necessary for a fundamental understanding of the invention, the description taken with the drawings making apparent to those skilled in the art how the several forms of the invention may be embodied in practice. [0044]
  • In the Drawings: [0045]
  • FIG. 1 illustrates SDS-PAGE analysis of extracts of Pporcine aortic endothelial cells (PAE Cells) which were transfected with vector along (lane 1) or with vector containing the LOR-1 cDNA (lane 3) and metabolically labeled with [0046] 35S-methionine. Extracts from the vector transfected cells (lane 2), from vector containing LOR-1 cDNA transfected cells (lane 4) or from 35S-methionine labeled human umbilical vein endothelial cells (HUVEC) (lane 5) were purified on a PF4 affinity column. A Band corresponding in size to the original band observed in the HUVEC is evident (compare lanes 4 and 5); this band is absent in extracts of vector transfected cells.
  • FIG. 2 illustrates differential expression of LOR-1 in breast cancer derived cells of different metastatic potential. The metastatic potential of the cells increases from left to right, and is correlated to increased LOR-1 mRNA expression. The results correspond to northern blot analysis of LOR-1 mRNA expression. Data regarding the relative metastatic potential of the cell lines was derived from the literature. [0047]
  • FIG. 3 illustrates expression of recombinant LOR-1 in MCF-7 breast cancer cells (lane 1). Vector transfected MCF-7 cells (lane 2) and two clones of MCF-7 expressing recombinant LOR-1 ([0048] lane 3, clone 12, lane 4, clone 22) were grown for two days in serum free medium. The medium from an equal number of cells was collected, concentrated 30 fold using Centricon™, and 10 μl aliquots were electrophoresed using an SDS/PAGE gel. Proteins were blotted onto nitrocellulose, and LOR-1 protein was identified using an antibody directed against the C-terminal of LOR-1. A secondary alkaline-phosphatase coupled antibody and NBT-BICP staining were used to detect the primary bound antibody.
  • FIG. 4 illustrates tumor size as correlated to LOR-1 expression. Parental MCF-7 cells (par), MCF-7 cells transfected with pCDNA3 vector alone (vec) and two recombinant LOR-1 expressing MCF-7 cells ([0049] clones 12 and 24) were deposited under the skin of immune deficient mice (107/injection site) along with an estrogen slow release pellet. Six animals were used for each cell type implanted. The area of the tumors was measured every few days. Bars represent standard deviation from the mean.
  • FIGS. 5[0050] a-b illustrate anti-factor-8 immunostaining of tumors generated by MCF-7 cells transfected with the expression vector alone (FIG. 5a) or with an expression vector containing the LOR-1 cDNA (FIG. 5b). Counter staining was performed with hematoxylin-eosin (blue). Invasion of blood vessels into the tumor mass is more abundant in LOR-1 expressing tumors (FIG. 5b) as compared to tumors generated by control cells which do not express LOR-1 (FIG. 5a).
  • FIGS. 6[0051] a-d illustrate liver sections of Wilson's disease patients (FIGS. 6c-d) and normal patients (FIGS. 6a-b) probed with a LOR-1 sense probe (FIGS. 6a, 6 c) and antisense probe (FIGS. 6b, 6 d).
  • FIG. 7 illustrates results of a whole mount in-situ hybridization using a LOR-1 cDNA probe and a 4 day old chick embryo. Strong expression of LOR-1 mRNA is observed in amniotic blood vessels (arrow). [0052]
  • FIG. 8 illustrates sequence alignment of several lysyl oxidases including LOR-1. [0053]
  • FIGS. 9[0054] a-b. illustrate the expression of LOR-1 and LOR-2 in human breast cancer derived cells: Total RNA was prepared from confluent MCF-7 cells (MCF-7), MDA-MB-231 cells (MDA-231) and MDA-MB-435 cells (MDA-435) and was subjected to Northern blot analysis using a LOR-1 (FIG. 9a) and LOR-2 (FIG. 9b) specific cDNA probes. LOR-1 specific hybridization signal is seen in tumors derived from both MDA-231 and MDA-435 cells but not in tumors derived from MCF-7 cells. LOR-2 specific hybridization signal is seen only in tumors derived from MDA-435 cells. LOR-1 and LOR-2 non-specific hybridization signals are seen in all tumors' RNA in a band that corresponds to the 28S rRNA.
  • FIGS. 9[0055] c-f illustrate the expression pattern of LOR-1 in normal human breast (FIG. 9c), in in-situ non-invasive breast carcinoma (FIG. 9d), in grade-1 invasive ductal carcinoma (FIG. 9e) and in grade-3 invasive ductal carcinoma (FIG. 9f). A polyclonal affinity purified rabbit antibody directed against the C-terminal of LOR-1 was used to detect the expression of LOR-1. High level expression of LOR-1 protein is seen in the epithelium of the normal duct (FIG. 9c, open arrow); magnification×100. In in-situ non-invasive breast carcinoma (FIG. 9d) the cancer cells have filled the duct but are still confined to it. Many of the cells located at periphery of the tumor have lost their ability to express LOR-1, while at the center, the cells still express high levels of LOR-1 (FIG. 9d, empty arrow); magnification×200. In grade-1 invasive breast carcinoma the tumorigenic cells have formed pseudo-ducts (FIG. 9e, black arrows) but they do not express LOR-1 anymore. However, cells found in a nearby carcinoma in-situ express LOR-1 (FIG. 9e, empty arrow); magnification×200. In grade-3 invasive breast carcinoma the tumorigenic cells express large amounts of LOR-1 and the morphology is completely disorderly (FIG. 9f); magnification×200.
  • FIG. 10[0056] a illustrates the expression of recombinant LOR-1 in MCF-7 cells transfected with the expression vector alone (vec) or with an expression vector containing the LOR-1 cDNA clone 12 or 24 (#12 or #24, respectively). LOR-1 proteins were deteceted by Western blot with an antibody directed against the C-terminal of human LOR-1.
  • FIG. 10[0057] b illustrates post-translational processes of LOR-1. MCF-7/Tet-LOR1 cells grown in the presence of tetracycline were trypsinized and seeded into a 24-well dish (5×104 cells/well) in a serum free medium in the absence of tetracycline. Cell aliquots collected at the noted times following tetracycline removal and were analyzed for the presence of LOR-1 by Western blot as described in FIG. 10a.
  • FIG. 10[0058] c illustrates the growth rate of tumors derived from control or LOR-1 expressing MCF-7 cells. MCF-7 cells transfected with the expression vector alone (vec) or with an expression vector containing the LOR-1 cDNA clone 12 or 24 were injected into the mammary fat pads of female athymic nude mice. Each cell type was implanted in 8 animals. Tumor area was measured at the indicated times. Error bars represent the standard error of the mean. The experiment was terminated and the mice sacrificed when the tumor reached a diameter of about 1 cm.
  • FIG. 10[0059] d illustrates the relative size of tumor in mice 25 days following injection of MCF-7 cells. Shown are mice harboring tumors that developed from cells transfected with the expression vector alone (left) or with the expression vector containing LOR-1 cDNA clone 24 (center) or 12 (right).
  • FIGS. 11[0060] a-h illustrate fibrotic foci and collagen deposits in tumors derived from MCF-7 cells expressing recombinant LOR-1. Hematoxilin-eosin staining demonstrate a few necrotic foci in a tumor derived from MCF-7 cells transfected with expression vector alone (FIG. 11a, arrow, magnification×20) and numerous necrotic foci in a tumor derived from MCF-7 cells transfected with expression vector containing LOR-1 cDNA clone 12 (FIG. 11b, arrows, magnification×20).
  • FIG. 11[0061] c illustrates human keratin-7 immunostaining of tumors generated by MCF-7 cells transfected with expression vector containing clone 12. Counter staining was performed with hematoxylin-eosin (blue). Arrow points to nuclei of host cells concentrated in the fibrotic foci. No necrosis can be seen; magnification×40. FIGS. 11d-f illustrate collagen deposits in tumor cells as viewed by Masson's Trichrome stain. A few collagen deposits are seen in tumors generated from MCF-7 cells transfected with the expression vector alone (FIG. 11d, arrows, magnification×200). Thick collagen bundles are seen between tumor cells generated from MCF-7 cells transfected with expression vector containing clone 12 (FIG. 11e, magnification×200). The fibrotic area is full with collagen fibers and interspaced with host derived cells in tumors generated from MCF-7 cells transfected with expression vector containing clone 24 (FIG. 11f, magnification×200).
  • FIGS. 11[0062] g-h illustrate blood vessels stained with Masson's Trichrome in tumors generated from MCF-7 cells transfected with the expression vector alone (FIG. 11g) or expression vector containing clone 12 (FIG. 11h); magnification×400.
  • FIGS. 12[0063] a-d illustrate the deposition of collagen type-3 by reticulum stain in tumors generated from MCF-7 and C6 glioma cells transfected with expression vector alone (FIG. 12a, c) or vector expressing LOR-1 cDNA clone 12 (FIG. 12b,d); magnification×200.
  • FIGS. 13[0064] a-i illustrate the invasiveness of tumors derived from MCF-7 cells expressing recombinant LOR-1. Shown are histological sections of tumors generated from MCF-7 cells transfected with an expression vector alone (FIGS. 13a, b) or a vector expressing LOR-1 (FIGS. 13c-h). Sections were labeled with either a monoclonal antibody specific to human keratin-7 (FIGS. 13 a-d and f-h, blue purple stain) or with an antibody directed against LOR-1 (FIG. 13e, red stain). Counterstain was with hematoxylin (light blue) in all sections. Black arrows designate cytokeratin-7 positive tumor cells invading the tumor pseudo-capsule (FIG. 13c), infiltrating between adjacent muscle bundles (FIG. 13d), or invading the vasculature (FIGS. 13f, g) and peri-neural space of nerves (FIG. 13h). White arrows designate LOR-1 positive tumor cells infiltrating muscles located next to the tumor (FIG. 13g). Blood vessels v; nerves n; muscle fibers m; magnification×100 for FIGS. 13a-c, f, h and ×200 for FIGS. 13d, e, g.
  • DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • The present invention is of pharmaceutical compositions and methods that can be used to modulate angiogenesis and to inhibit tumor invasiveness and tumor fibrosis. Specifically, the present invention can be used to suppress tumor growth and metastasis as well as to treat disorders such as, for example, arthritis, diabetic retinopathy, psoriasis and vasculitis. [0065]
  • The principles and operation of the present invention may be better understood with reference to the drawings and accompanying descriptions. [0066]
  • Before explaining at least one embodiment of the invention in detail, it is to be understood that the invention is not limited in its application to the details of construction and the arrangement of the components set forth in the following description or illustrated in the drawings described in the Examples section. The invention is capable of other embodiments or of being practiced or carried out in various ways. Also, it is to be understood that the phraseology and terminology employed herein is for the purpose of description and should not be regarded as limiting. [0067]
  • As described in the Examples section which follows, the present inventors have uncovered a novel protein constituent of the angiogenic process. [0068]
  • This protein, which is termed LOR-1 herein (SEQ ID NO: 2) belongs to the lysyl-oxidase family of enzymes which catalyze the formation of covalent crosslinks between lysine residues on adjacent collagen or elastin fibrils. The lysyl oxidase family includes four genes (Saito et al., 1997; Kim et al., 1995; Reiser et al., 1992; Jang et al., 1999), the protein sequences of which are presented in SEQ ID NOs: 3, 6, 8 and 9. A homology comparison between several lysyl oxidase family members is presented in FIG. 8 which is further described in the Examples section which follows. [0069]
  • Each member of the lysyl-oxidase family of enzymes includes a highly conserved lysyl-oxidase domain, the activity of which is highly dependent on the presence of copper. [0070]
  • It should be noted that prior art studies have shown that removal of copper from tumor tissues leads to inhibition of angiogenesis (Rabinovitz, 1999; Yoshida et al., 1995). This further substantiates the role of the lysyl-oxidase family of enzymes in angiogenesis since presumably, removal of copper leads to inhibition of lysyl-oxidases. [0071]
  • Further support to the angiogenic activity of lysyl-oxidases is provided by the PF4-LOR-1 binding assays presented herein. As mentioned hereinabove, PF4 is an inhibitor of angiogenesis. As such, the anti-angiogenic activity exhibited by PF4 may be effected through LOR-1 inhibition, which, as demonstrated in the Examples section which follows, is highly expressed in the endothelial cells lining blood vessels. [0072]
  • Thus according to one aspect of the present invention there is provided a method of modulating angiogenesis [0073]
  • The method is effected by administering into the mammalian tissue a molecule capable of modifying a tissue level and/or activity of at least one type of lysyl-oxidase to thereby modulate angiogenesis in the mammalian tissue. [0074]
  • As used herein, the phrase “tissue level” refers to the level of lysyl-oxidase protein present in active form in the tissue at a given time point. Protein levels are determined by factors such as, transcription and/or translation rates, RNA or protein turnover and/or protein localization within the cell. As such any molecule which effects any of these factors can modify the tissue level of the lysyl-oxidase. [0075]
  • As used herein the term “activity” refers to an enzymatic activity of the lysyl-oxidase. A molecule which can modify the enzymatic activity may directly or indirectly alter substrate specificity of the enzyme or activity of the catalytic site thereof. [0076]
  • There are numerous examples of molecules which can specifically modify the tissue level and/or activity of a lysyl-oxidase. Such molecules can be categorized into lysyl-oxidase “upregulators” or “downregulators”. [0077]
  • Downregulators [0078]
  • An antibody (polyclonal, monoclonal or monospecific) or an antibody portion (e.g., Fab fragment) directed against at least a portion of a lysyl-oxidase (e.g., region spanning the catalytic site) can be used to specifically inhibit lysyl-oxidase activity when introduced into the mammalian tissue. As such, an antibody or an antibody fragment directed at a lysyl-oxidase can be used to suppress or arrest the formation of blood vessels, and to inhibit tumor fibrosis and metastasis. [0079]
  • Numerous examples of antibody inhibitors are known in the art, including inhibitors of angiogenesis which target angiogenic factors (Brooks et al., 1994; Brooks et al., 1998). [0080]
  • As is described below, various approaches can be used to reduce or abolish transcription or translation of a lysyl oxidase. These include anti-sense or ribozyme approaches as well as RNA interference. [0081]
  • An antisense molecule which can be used with the present invention includes a polynucleotide or a polynucleotide analog of at least 10 bases, preferably between 10 and 15, more preferably between 50 and 20 bases, most preferably, at least 17, at least 18, at least 19, at least 20, at least 22, at least 25, at least 30 or at least 40 bases which is hybridizable in vivo, under physiological conditions, with a portion of a polynucleotide strand encoding a polypeptide at least 50% homologous to SEQ ID NO: 1, 4, 5 or 7 or at least 75% homologous to an N-terminal portion thereof as determined using the BestFit software of the Wisconsin sequence analysis package, utilizing the Smith and Waterman algorithm, where gap creation penalty equals 8 and gap extension penalty equals 2. [0082]
  • The antisense oligonucleotides used by the present invention can be expressed from nucleic acid construct administered into the tissue, in which case inducible promoters are preferably used such that antisense expression can be switched on and off, or alternatively such oligonucleotides can be chemically synthesized and administered directly into the tissue, as part of, for example, a pharmaceutical composition. [0083]
  • The ability of chemically synthesizing oligonucleotides and analogs thereof having a selected predetermined sequence offers means for downmodulating gene expression. Four types of gene expression modulation strategies may be considered. [0084]
  • At the transcription level, antisense or sense oligonucleotides or analogs that bind to the genomic DNA by strand displacement or the formation of a triple helix, may prevent transcription. At the transcript level, antisense oligonucleotides or analogs that bind target mRNA molecules lead to the enzymatic cleavage of the hybrid by intracellular RNase H. In this case, by hybridizing to the targeted mRNA, the oligonucleotides or oligonucleotide analogs provide a duplex hybrid recognized and destroyed by the RNase H enzyme. Alternatively, such hybrid formation may lead to interference with correct splicing. As a result, in both cases, the number of the target mRNA intact transcripts ready for translation is reduced or eliminated. [0085]
  • At the translation level, antisense oligonucleotides or analogs that bind target mRNA molecules prevent, by steric hindrance, binding of essential translation factors (ribosomes), to the target mRNA, a phenomenon known in the art as hybridization arrest, disabling the translation of such mRNAs. [0086]
  • Several prior art studies have shown that antisense oligonucleotides can be effective in vivo. For example, antisense molecules have been used to arrest hematopoietic cell proliferation (Szczylik et al., 1991), growth (Calabretta et al. 1991), or entry into the S phase of the cell cycle (Heikhila et al., 1987) and to prevent receptor mediated responses (Burch and Mahan, 1991). [0087]
  • Several considerations must be taken into account when designing antisense oligonucleotides. For efficient in vivo inhibition of gene expression using antisense oligonucleotides or analogs, the oligonucleotides or analogs must fulfill the following requirements (i) sufficient specificity in binding to the target sequence; (ii) solubility in water; (iii) stability against intra- and extracellular nucleases; (iv) capability of penetration through the cell membrane; and (v) when used to treat an organism, low toxicity. The binding affinity of an antisense oligonucleotide can be predicted (Walton et al., 1999, Biotechnol Bioeng 65: 1-9; Matveeva et al., 1998, Nature Biotechnology 16, 1374-1375.) Unmodified oligonucleotides are typically impractical for use as antisense sequences since they have short in vivo half-lives, during which they are degraded rapidly by nucleases. Furthermore, they are difficult to prepare in more than milligram quantities. In addition, such oligonucleotides are poor cell membrane penetrants. [0088]
  • Thus it is apparent that in order to meet all the above listed requirements, oligonucleotide analogs need to be devised in a suitable manner. [0089]
  • For example, problems arising in connection with double-stranded DNA (dsDNA) recognition through triple helix formation have been diminished by a clever “switch back” chemical linking, whereby a sequence of polypurine on one strand is recognized, and by “switching back”, a homopurine sequence on the other strand can be recognized. Also, good helix formation has been obtained by using artificial bases, thereby improving binding conditions with regard to ionic strength and pH. [0090]
  • In addition, in order to improve half-life as well as membrane penetration, a large number of variations in polynucleotide backbones have been done, nevertheless with little success. [0091]
  • Oligonucleotides can be modified either in the base, the sugar or the phosphate moiety. These modifications include, for example, the use of methylphosphonates, monothiophosphates, dithiophosphates, phosphoramidates, phosphate esters, bridged phosphorothioates, bridged phosphoramidates, bridged methylenephosphonates, dephospho internucleotide analogs with siloxane bridges, carbonate bridges, carboxymethyl ester bridges, carbonate bridges, carboxymethyl ester bridges, acetamide bridges, carbamate bridges, thioether bridges, sulfoxy bridges, sulfono bridges, various “plastic” DNAs, anomeric bridges and borane derivatives (Cook, 1991). [0092]
  • International patent application WO 89/12060 discloses various building blocks for synthesizing oligonucleotide analogs, as well as oligonucleotide analogs formed by joining such building blocks in a defined sequence. The building blocks may be either “rigid” (i.e., containing a ring structure) or “flexible” (i.e., lacking a ring structure). In both cases, the building blocks contain a hydroxy group and a mercapto group, through which the building blocks are said to join to form oligonucleotide analogs. The linking moiety in the oligonucleotide analogs is selected from the group consisting of sulfide (—S—), sulfoxide (—SO—), and sulfone (—SO2-). [0093]
  • International patent application WO 92/20702 describe an acyclic oligonucleotide which includes a peptide backbone on which any selected chemical nucleobases or analogs are stringed and serve as coding characters as they do in natural DNA or RNA. These new compounds, known as peptide nucleic acids (PNAs), are not only more stable in cells than their natural counterparts, but also bind natural DNA and [0094] RNA 50 to 100 times more tightly than the natural nucleic acids cling to each other. PNA oligomers can be synthesized from the four protected monomers containing thymine, cytosine, adenine and guanine by Merrifield solid-phase peptide synthesis. In order to increase solubility in water and to prevent aggregation, a lysine amide group is placed at the C-terminal region.
  • Thus, antisense technology requires pairing of messenger RNA with an oligonucleotide to form a double helix that inhibits translation. The concept of antisense-mediated gene therapy was already introduced in 1978 for cancer therapy. This approach was based on certain genes that are crucial in cell division and growth of cancer cells. Synthetic fragments of genetic substance DNA can achieve this goal. Such molecules bind to the targeted gene molecules in RNA of tumor cells, thereby inhibiting the translation of the genes and resulting in dysfunctional growth of these cells. Other mechanisms have also been proposed. These strategies have been used, with some success in treatment of cancers, as well as other illnesses, including viral and other infectious diseases. [0095]
  • Antisense oligonucleotides are typically synthesized in lengths of 13-30 nucleotides. The life span of oligonucleotide molecules in blood is rather short. Thus, they have to be chemically modified to prevent destruction by ubiquitous nucleases present in the body. Phosphorothioates are very widely used modification in antisense oligonucleotide ongoing clinical trials. A new generation of antisense molecules consist of hybrid antisense oligonucleotide with a central portion of synthetic DNA while four bases on each end have been modified with 2′O-methyl ribose to resemble RNA. In preclinical studies in laboratory animals, such compounds have demonstrated greater stability to metabolism in body tissues and an improved safety profile when compared with the first-generation unmodified phosphorothioate. Dozens of other nucleotide analogs have also been tested in antisense technology. [0096]
  • RNA oligonucleotides may also be used for antisense inhibition as they form a stable RNA-RNA duplex with the target, suggesting efficient inhibition. However, due to their low stability RNA oligonucleotides are typically expressed inside the cells using vectors designed for this purpose. This approach is favored when attempting to target a mRNA that encodes an abundant and long-lived protein. [0097]
  • Recent scientific publications have validated the efficacy of antisense compounds in animal models of hepatitis, cancers, coronary artery restenosis and other diseases. The first antisense drug was recently approved by the FDA. The drug, Fomivirsen, was developed by Isis, and is indicated for local treatment of cytomegalovirus in patients with AIDS who are intolerant of or have a contraindication to other treatments for CMV retinitis or who were insufficiently responsive to previous treatments for CMV retinitis (Pharmacotherapy News Network). [0098]
  • Several antisense compounds are now in clinical trials in the United States. These include locally administered antivirals, systemic cancer therapeutics. Antisense therapeutics has the potential to treat many life-threatening diseases with a number of advantages over traditional drugs. Traditional drugs intervene after a disease-causing protein is formed. Antisense therapeutics, however, block mRNA transcription/translation and intervene before a protein is formed, and since antisense therapeutics target only one specific mRNA, they should be more effective with fewer side effects than current protein-inhibiting therapy. [0099]
  • A second option for disrupting gene expression at the level of transcription uses synthetic oligonucleotides capable of hybridizing with double stranded DNA. A triple helix is formed. Such oligonucleotides may prevent binding of transcription factors to the gene's promoter and therefore inhibit transcription. Alternatively, they may prevent duplex unwinding and, therefore, transcription of genes within the triple helical structure. [0100]
  • Ribozymes may also be used as down regulators. Ribozymes are being increasingly used for the sequence-specific inhibition of gene expression by the cleavage of mRNAs encoding proteins of interest. The possibility of designing ribozymes to cleave any specific target RNA has rendered them valuable tools in both basic research and therapeutic applications. In the therapeutics area, ribozymes have been exploited to target viral RNAs in infectious diseases, dominant oncogenes in cancers and specific somatic mutations in genetic disorders. Most notably, several ribozyme gene therapy protocols for HIV patients are already in [0101] Phase 1 trials (Welch et al., 1998). More recently, ribozymes have been used for transgenic animal research, gene target validation and pathway elucidation. Several ribozymes are in various stages of clinical trials. ANGIOZYME was the first chemically synthesized ribozyme to be studied in human clinical trials. ANGIOZYME specifically inhibits formation of the VEGF-r (Vascular Endothelial Growth Factor receptor), a key component in the angiogenesis pathway. Ribozyme Pharmaceuticals, Inc., as well as other firms have demonstrated the importance of anti-angiogenesis therapeutics in animal models. HEPTAZYME, a ribozyme designed to selectively destroy Hepatitis C Virus (HCV) RNA, was found effective in decreasing Hepatitis C viral RNA in cell culture assays (Ribozyme Pharmaceuticals, Incorporated).
  • Another mechanism of down regulating enzymes at the transcript level is RNA interference (RNAi), an approach which utilizes small interfering dsRNA (siRNA) molecules that are homologous to the target mRNA and lead to its degradation (Carthew, 2001). RNAi is an evolutionarily conserved surveillance mechanism that responds to double-stranded RNA by sequence-specific silencing of homologous genes (Fire et al., 1998; Zamore et al., 2000). RNAi is initiated by the dsRNA-specific endonuclease Dicer, which promotes cleavage of long dsRNA into double-stranded fragments between 21 and 25 nucleotides long, termed small interfering RNA (siRNAs) (Zamore et al., 2000; Elbashir et al., 2001; Hammond et al., 2000; Bernstein et al., 2001). siRNA are incorporated into a protein complex that recognizes and cleaves target mRNAs (Nykanen et al., 2001). [0102]
  • RNAi has been increasingly used for the sequence-specific inhibition of gene expression. The possibility of interfering with any specific target RNA has rendered RNAi a valuable tool in both basic research and therapeutic applications. RNAi was first used for gene silencing in nematodes (Fire et al., 1991;Guo & Kemphues, 1995). [0103]
  • Recent scientific publications have validated the efficacy of such short double stranded RNA molecules in inhibiting target mRNA expression and thus have clearly demonstrated the therapeutic potential of such molecules. For example, RNAi has been utilized to inhibit expression of hepatitis C (McCaffrey et al., 2002), HIV-1 (Jacque et al., 2002), cervical cancer cells (Jiang and Milner 2002) and leukemic cells (Wilda et al., 2002). [0104]
  • Several considerations must be taken into account when designing RNAi for in vivo administration into mammalian cells. Since the introduction of dsRNA into mammalian cells does not result in efficient Dicer-mediated generation of siRNA (Caplen et al., 2000; Ui-Tei et al., 2000) short siRNA duplexes of typically 21 to 25-base pairs are utilized to initiate target cleavage. [0105]
  • Such siRNA molecules can be chemically synthesized as 21 to 25-nucleotide siRNA duplexes (Elbashir et al., 2001; McCaffrey et al., 2002). Synthetic siRNA oligonucleotide duplexes can be prepared with either [0106] ribonucleotide 3′ overhangs or with deoxyribonucleotide 3′ overhangs (Hohjoh 2002). They can also be prepared as a sense-stranded DNA/antisense-stranded RNA hybrids or vise versa.
  • The siRNA used by the present invention can be transcribed in vitro from plasmids and administered into the tissue. Transcripts that include two self-complementary siRNAs annealed to form a loop region can be further processed by single-stranded ribonucleases and/or other proteins into a functional duplex siRNA molecule (Leirdal and Sioud, 2002). siRNA can also be prepared from dsRNA by Escherichia coli RNase III cleavage into endoribonuclease-prepared siRNA (esiRNA). [0107]
  • Since approaches for introducing synthetic siRNA into cells by lipofection can result in low transfection efficiencies in some cell types and/or short-term persistence of silencing effects, vector mediated methods have been developed. [0108]
  • Thus, siRNA molecules utilized by the present invention are preferably delivered into cell using retroviruses. Delivery of siRNA using retroviruses provides several advantages over methods, such as lipofection, since retroviral delivery is more efficient, uniform and immediately selects for stable “knock-down” cells (Devroe and Silver, 2002). [0109]
  • Thus, siRNA molecules of the present invention are preferably transcribed from expression vectors which can facilitate stable expression of the siRNA transcripts once introduced into a host cell. These vectors are engineered to express small hairpin RNAs (shRNAs), which are processed in-vivo into siRNA molecules capable of carrying out gene-specific silencing (Brummelkamp, 2002; Paddison et al. 2002; Paul et al., 2002; Yu et al., 2002. [0110]
  • An example of a suitable expression vector is the pSUPER™, which includes the polymerase-III H1-RNA gene promoter with a well defined start of transcription and a termination signal consisting of five thymidines in a row (T5) (Brummelkamp, 2002). Most importantly, the cleavage of the transcript at the termination site is at a site following the second uridine, thus yielding a transcript which resembles the ends of synthetic siRNAs, which also contain nucleotide overhangs. siRNA is cloned such that it includes the sequence of interest, i.e., LOR-1 separated by a short spacer from the reverse complement of the same sequence. The resulting transcript folds back on itself to form a stem-loop structure, which mediates LOR-1 RNAi. [0111]
  • Another suitable siRNA expression vector encodes the sense and antisense siRNA under the regulation of separate polIII promoters (Miyagishi and Taira (2002) Nature Biotech. 20:497-500). The siRNA, generated by this vector also includes a 5 thymidine termination signal. [0112]
  • Oligoribonucleotide sequences of the present invention can also be placed under bi-directional promoters to produce both the sense and antisense transcripts from the same promoter construct, thus simplifying the construction of expression vectors and achieving an equal molar ratio of cellular sense and antisense sequences. Examples for bi-directional promoters are disclosed in U.S. pat. application No. 20020108142. [0113]
  • The downregulators described hereinabove would be particularly useful for inhibiting angiogenesis in tumor tissue. It has been shown that PF4, a lysyl oxidase binding protein which inhibits angiogenesis in tumor tissue specifically accumulates in newly formed blood vessels of tumors (angiogenic vessels) but not in established blood vessels (Hansell et al., 1995; Reiser et al., 1992). [0114]
  • Newly formed angiogenic blood vessels are more permeable to proteins than established blood vessels because the major inducer of angiogenesis in many angiogenic diseases is VEGF, a growth factor which also functions as a potent blood vessel permeabilizing factor (VPF) (Neufeld et al., 1999). Tumor associated blood vessels are therefore in a permanent state of hyperpermeability due to deregulated over-expression of VEGF (Shweiki et al., 1995; Rak et al., 1995) and as such, a downregulator molecule used by the method of the present invention would be able to extravasate efficiently from tumor blood vessels but much less efficiently from normal stabilized blood vessels. [0115]
  • Upregulators [0116]
  • Several approaches can be utilized to increase the levels of lysyl oxidase and as such to enhance the formation of blood vessels. [0117]
  • For example, a nucleic acid construct including a constitutive, inducible or tissue specific promoter positioned upstream of a polynucleotide encoding a polypeptide having lysyl oxidase activity, such as the polypeptide set forth in SEQ ID NO: 2, 3, 6, 8 or 9 can be administered into a mammalian tissue. The lysyl oxidase expressed from this construct would substantially increase the levels of lysyl oxidase within the cells of the tissue and as such enhance angiogenesis. [0118]
  • The polynucleotide segments encoding the lysyl oxidase can be ligated into a commercially available expression vector system suitable for transforming mammalian cells and for directing the expression of this enzyme within the transformed cells. It will be appreciated that such commercially available vector systems can easily be modified via commonly used recombinant techniques in order to replace, duplicate or mutate existing promoter or enhancer sequences and/or introduce any additional polynucleotide sequences. [0119]
  • Suitable mammalian expression vectors include, but are not limited to, pcDNA3, pcDNA3.1(+/−), pZeoSV2(+/−), pSecTag2, pDisplay, pEF/myc/cyto, pCMV/myc/cyto, pCR3.1, which are available from Invitrogen, pCI which is available from Promega, pBK-RSV and pBK-CMV which are available from Stratagene, pTRES which is available from Clontech, and their derivatives. [0120]
  • Gene “knock-in” techniques can also be used to increase the expression levels of the lysyl oxidase. [0121]
  • Enhancer elements can be “knocked-in” adjacent to endogenous lysyl oxidase coding sequences to thereby increase transcription therefrom. [0122]
  • Further detail relating to the construction and use of knock-out and knock-in constructs is provided elsewhere (Fukushige and Ikeda, 1996; Bedell et al., 1997; Bermingham et al., 1996). [0123]
  • It will be appreciated that direct administration of a polypeptide exhibiting lysyl oxidase activity can also be utilized for enhancing angiogenesis. [0124]
  • It will be appreciated that affinity binding assays and/or activity assays, the principles of which are well known in the art, can be used to screen for novel compounds (e.g., substrate analogs) which can specifically regulate activity of lysyl oxidase and as such can be used with the present invention. [0125]
  • An assay suitable for use with this aspect of the present invention has been previously described in a study conducted by Bedell-Hogan et al., 1993. [0126]
  • As is clearly illustrated in the Examples section which follows, the present study also correlated expression levels of LOR-1 to the metastatic properties of breast cancer derived cell lines, indicating that LOR-1 may play additional roles in tumor invasiveness in addition to its role in angiogenesis. [0127]
  • Thus, the present invention also provides a method of inhibiting metastasis and/or fibrosis in a mammalian tissue. The method is effected by administering to the mammalian tissue a molecule capable of downregulating a tissue level and/or activity of at least one type of lysyl-oxidase. [0128]
  • The method of the present invention can be used to treat human patients that have been diagnosed with cancerous tumors, by administering any of the downregulating molecules described herein above, in order to reduce the tissue level and/or activity of at least one type of a lysyl oxidase. [0129]
  • As used herein, the phrase “cancerous tumor” refers to any malignant tumor within a human body including, but not limiting to, tumors with metastases. In addition, and without being bound to any particular type of cancerous tumor, the present invention is useful to treat breast cancer tumors, with or without metastases. [0130]
  • As used herein, the phrase “administering” refers to all modes of administration described hereinbelow with respect to the pharmaceutical compositions of the present invention. [0131]
  • These include, but not limit to, local administration at the tumor tissue, an organ where the cancerous tumor was diagnosed and/or related tissues that typically form metastases [Hortobagyi, 2002, Semin Oncol 29 (3 Suppl 11): 134-44; Morrow and Gradishar, 2002, 324: 410-4]. Examples of related tissue include lymph nodes adjacent to, for example, breast tissue and bones. [0132]
  • Administration can also be effected in a systemic manner in order to treat the affected tissue, i.e., the tissue where the cancerous tumor was formed and where metastases are present or likely to be formed with tumor progression. [0133]
  • Since any molecule capable of downregulating lysyl-oxidase activity can be utilized by the methods described hereinabove, the present invention also provide a method of identifying molecules capable of inhibiting metastasis and/or fibrosis. [0134]
  • This method is effected by screening and identifying molecules which exhibit specific reactivity with at least one type of lysyl-oxidase and testing a metastasis and/or fibrosis inhibitory potential of these molecules. [0135]
  • Numerous types of molecules can be screened for reactivity with at least one type of lysyl-oxidase, examples include, but are not limited to, molecules such as antisense oligonucleotides, siRNA and ribozymes that interact with a polynucleotide expressing a lysyl oxidase activity or molecules such as antibodies that interact with polypeptides having a lysyl oxidase activity. In addition, short peptides and other small molecules can also be screened by this method of the present invention. [0136]
  • Screening for cross reactivity can be effected by lysyl-oxidase enzymatic activity assays, by binding assays and the like. Examples of suitable assays are provided in Rodriguez et al., 2002, Arterioscler Thromb Vasc Biol 22: 1409-14; Wilson and Nock, 2002, Curr Opin Chem Biol 6: 81-5; Uetz, 2002, Curr Opin Chem Biol 6: 57-62; Stoll et al., 2002, Front Biosci 2002 7: c13-32). [0137]
  • Testing a metastatic phenotype of transformed tumor cells can be performed in-vitro since nearly all steps of the metastatic process, including attachment, matrix degradation and migration, can be modeled experimentally in-vitro by measuring invasion of a reconstituted basement membrane (RBM). Metastatic invasiveness of tumor cell can be modeled by migration of tumor cells into reconstituted basement membrane (RBM) in the presence and absence of a chemoattractant, such as fibroblast conditioned medium (FCM). The assay determines cells that have attached to the RBM, degraded the RBM enzymatically and, finally, cells that have penetrated the FCM side of the membrane. [0138]
  • Since in-vitro metastasis events correspond to steps observed in the metastatic spread of tumor cells through the basement membrane in-vivo, in-vitro invasiveness of cells can be assayed by the methods described in Albini et al., 1987 Cancer Research 47: 3239-3245, which is incorporated herein by reference in its entirety. Invasiveness assays and other methods for assessing metastatic affects, are described in Leyton et al., 1994 Cancer Research 54:3696-3699, which is incorporated by reference herein in its entirety. Reconstituted basement membrane preparations for use in accordance with the hereinabove described assays are readily available from numerous commercial suppliers. One suitable example membrane in this regard is “MATRIGEL” available from Collaborative Biomedical Products of Bedford, Mass. [0139]
  • In-vitro evaluation of tumor cell metastatic phenotype can also be effected by determining level and pattern of expression of one or more metastasis associated markers such protease markers, which are considered to be an integral part of tumor metastasis (see U.S. Pat. No.: 6,303,318). One example is the arachidonic acid, the release of which in cells can serve to indicate metastatic potential of a tumor (U.S. Pat. No: 6,316,416). In this regard, determining phospholipase A-2 (PLA[0140] 2) activity, and the activity or abundance of factors that affect the activity of PLA2, such as uteroglobin protein (U.S. Pat. No.: 6,316,416) can serve as an indication of metastatic potential.
  • Determining pattern and level of expression of metastasis-associated markers can be effected by one of several methods known in the art. [0141]
  • The presence or level of proteins indicative of metastatic potential of tumors can be determined in cells by conventional methods well known to those of skill in the art. For instance, the techniques for making and using antibody and other immunological reagents and for detecting particular proteins in samples using such reagents are described in current protocols in immunology, Coligan et al., Eds., John Wiley & Sons, New York (1995), which is incorporated by reference herein in parts pertinent to making and using reagents useful for determining specific proteins in samples. As another example, immunohistochemical methods for determining proteins in cells in tissues are described in [0142] Volume 2, Chapter 14 of current protocols in molecular biology, Ausubel et al., Eds., John Wiley & Sons, Inc. (1994), which is incorporated by reference herein in part pertinent to carrying out such determinations. Finally, Linnoila et al., A.J.C.P. 97(2): 235-243 (1992) and Peri et al., J. Clin. Invest. 92: 2099-2109 (1992), incorporated herein as referred to above, describe techniques that may need, in part, in this aspect of the present invention.
  • Metastatic potential can also be determined in vivo at the mRNA level. The presence and/or level of mRNA transcripts can be determined by a variety of methods known to those of skill in the art. A given mRNA may be detected in cells by hybridization to a specific probe. Such probes may be cloned DNAs or fragments thereof, RNA, typically made by in-vitro transcription, or oligonucleotide probes, usually generated by solid phase synthesis. Methods for generating and using probes suitable for specific hybridization are well known and used in the art. [0143]
  • A variety of controls may be usefully employed to improve accuracy in mRNA detection assays. For instance, samples may be hybridized to an irrelevant probe and treated with RNAse A prior to hybridization, to assess false hybridization. [0144]
  • In order to modulate angiogenesis or inhibit metastasis or tumor fibrosis, the molecules used by the present invention can be administered to the individual per se, or in a pharmaceutical composition where it is mixed with suitable carriers or excipients. [0145]
  • As used herein a “pharmaceutical composition” refers to a preparation of one or more of the active ingredients described herein with other chemical components such as physiologically suitable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate administration/targeting of a compound to a mammal. [0146]
  • As used herein the term “active ingredients”, refers to the preparation accountable for the biological effect, i.e. the upregulator/downregulator molecules used by the present invention to modulate angiogenesis and the downregulators molecules used by the present invention to inhibit metastasis and tumor fibrosis. [0147]
  • Hereinafter, the phrases “physiologically acceptable carrier” and “pharmaceutically acceptable carrier” are interchangeably used to refer to a carrier, such as, for example, a liposome, a virus, a micelle, or a protein, or a diluent which do not cause significant irritation to the mammal and do not abrogate the biological activity and properties of the active ingredient. An adjuvant is included under these phrases. [0148]
  • Herein the term “excipient” refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient. Examples, without limitation, of excipients, include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols. [0149]
  • Techniques for formulation and administration of compositions may be found in “Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, Pa., latest edition, which is incorporated herein by reference. [0150]
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, transnasal, intestinal or parenteral delivery, including intramuscular, subcutaneous and intramedullary injections as well as intrathecal, direct intraventricular, intravenous, inrtaperitoneal, intranasal, or intraocular injections. [0151]
  • For injection, the active ingredients of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological salt buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. [0152]
  • For oral administration, the compounds can be formulated readily by combining the active ingredient with pharmaceutically acceptable carriers well known in the art. Such carriers enable the active ingredient of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient. Pharmacological preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carbomethylcellulose; and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. [0153]
  • Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses. [0154]
  • Pharmaceutical compositions, which can be used orally, include push-fit capsules made of gelatin as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules may contain the active ingredients in admixture with filler such as lactose, binders such as starches, lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active ingredients may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for the chosen route of administration. [0155]
  • For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner. [0156]
  • The preparations described herein may be formulated for parenteral administration, e.g., by bolus injection or continuos infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers with optionally, an added preservative. The compositions may be suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. [0157]
  • Pharmaceutical compositions for parenteral administration include aqueous solutions of the active preparation in water-soluble form. Additionally, suspensions of the active ingredients may be prepared as appropriate oily or water based injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acids esters such as ethyl oleate, triglycerides or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the active ingredients to allow for the preparation of highly concentrated solutions. [0158]
  • Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water based solution, before use. [0159]
  • The preparation of the present invention may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides. [0160]
  • Pharmaceutical compositions suitable for use in context of the present invention include compositions wherein the active ingredients are contained in an amount effective to achieve the intended purpose. [0161]
  • The pharmaceutical composition may form a part of an article of manufacturing which also includes a packaging material for containing the pharmaceutical composition and a leaflet which provides indications of use for the pharmaceutical composition. [0162]
  • Thus, the present invention provides a method and pharmaceutical compositions useful modulating angiogenesis. [0163]
  • Such modulation activity can be used to treat arthritis (Koch, 1998; Paleolog and Fava, 1998), diabetic retinopathy (Miller et al., 1997), psoriasis (Detmar et al., 1994; Creamer et al., 1997) and vasculitis (Lie, 1992; Klipple and Riordan, 1989). [0164]
  • In addition, the present invention can also be used to treat disease characterized by fragile blood vessels, including Marfans syndrom, Kawasaki, Ehlers-Danlos, cutis-laxa, and takysu (Lie, 1992; Klipple and Riordan, 1989; Brahn et al., 1999; Cid et al., 1993; Hoffman et al., 1991). [0165]
  • It is possible that some of these diseases result from reduced or abolished lysyl oxidase activity which leads to the synthesis of a fragile extracellular matrix, and consequently, fragile blood vessels. [0166]
  • As such, administration of lysyl oxidase encoding sequences or polypeptides can be used to correct some of the manifestations of these diseases. [0167]
  • The present invention can also be used to treat diseases which are characterized by changes in the wall of blood vessels. For example, restenosis which is a common complication following balloon therapy, Fibromuscular dysplasia (Begelman, and Olin, 2000) and aortic stenosis (Palta et al., 2000) are all potentially treatable by the method of the present invention. [0168]
  • In addition, as is illustrated in the Examples section which follows, LOR-1 is more highly expressed in metastatic tumors and cell lines than in non-metastatic tumors and cell lines (FIGS. 3, 9, and [0169] 13). This suggests that levels of LOR-1 expression can be used as a diagnostic tool to determine the malignancy of cancer cells, as well as, to determine and implement suitable treatment regimens.
  • Additional objects, advantages, and novel features of the present invention will become apparent to one ordinarily skilled in the art upon examination of the following examples, which are not intended to be limiting. Additionally, each of the various embodiments and aspects of the present invention as delineated hereinabove and as claimed in the claims section below finds experimental support in the following examples. [0170]
  • EXAMPLES
  • Reference is now made to the following examples, which together with the above descriptions, illustrate the invention in a non limiting fashion. [0171]
  • Generally, the nomenclature used herein and the laboratory procedures utilized in the present invention include molecular, biochemical, microbiological and recombinant DNA techniques. Such techniques are thoroughly explained in the literature. See, for example, “Molecular Cloning: A laboratory Manual” Sambrook et al., (1989); “Current Protocols in Molecular Biology” Volumes I-III Ausubel, R. M., ed. (1994); Ausubel et al., “Current Protocols in Molecular Biology”, John Wiley and Sons, Baltimore, Md. (1989); Perbal, “A Practical Guide to Molecular Cloning”, John Wiley & Sons, New York (1988); Watson et al., “Recombinant DNA”, Scientific American Books, New York; Birren et al. (eds) “Genome Analysis: A Laboratory Manual Series”, Vols. 1-4, Cold Spring Harbor Laboratory Press, New York (1998); methodologies as set forth in U.S. Pat. Nos. 4,666,828; 4,683,202; 4,801,531; 5,192,659 and 5,272,057; “Cell Biology: A Laboratory Handbook”, Volumes I-III Cellis, J. E., ed. (1994); “Current Protocols in Immunology” Volumes I-III Coligan J. E., ed. (1994); Stites et al. (eds), “Basic and Clinical Immunology” (8th Edition), Appleton & Lange, Norwalk, Conn. (1994); Mishell and Shiigi (eds), “Selected Methods in Cellular Immunology”, W. H. Freeman and Co., New York (1980); available immunoassays are extensively described in the patent and scientific literature, see, for example, U.S. Pat. Nos. 3,791,932; 3,839,153; 3,850,752; 3,850,578; 3,853,987; 3,867,517; 3,879,262; 3,901,654; 3,935,074; 3,984,533; 3,996,345; 4,034,074; 4,098,876; 4,879,219; 5,011,771 and 5,281,521; “Oligonucleotide Synthesis” Gait, M. J., ed. (1984); “Nucleic Acid Hybridization” Hames, B. D., and Higgins S. J., eds. (1985); “Transcription and Translation” Hames, B. D., and Higgins S. J., eds. (1984); “Animal Cell Culture” Freshney, R. I., ed. (1986); “Immobilized Cells and Enzymes” IRL Press, (1986); “A Practical Guide to Molecular Cloning” Perbal, B., (1984) and “Methods in Enzymology” Vol. 1-317, Academic Press; “PCR Protocols: A Guide To Methods And Applications”, Academic Press, San Diego, Calif. (1990); Marshak et al., “Strategies for Protein Purification and Characterization—A Laboratory Course Manual” CSHL Press (1996); all of which are incorporated by reference as if fully set forth herein. Other general references are provided throughout this document. The procedures therein are believed to be well known in the art and are provided for the convenience of the reader. All the information contained therein is incorporated herein by reference. [0172]
  • Example 1 The role of LOR1 in Angiogenesis
  • A study was conducted in efforts to further substantiate and characterize the role of LOR-1 in angiogenes is. [0173]
  • Materials and Methods [0174]
  • Human recombinant platelet factor-4 (PF4, GenBank Accession number M20901) which was produced in bacteria and subsequently refolded was supplied by Dr. Maione of Repligen Corp. (Boston, USA). Estrogen slow release pellets were obtained from Innovative Research of America, Sarasota, Fla., USA. [0175]
  • Construction of LOR-1 expression vector, transfection into MCF-7 cells, and expression: The LOR-1 cDNA (SEQ ID NO: 1) was cloned into a pCDNA3.1-hygro expression vector (Invitrogen Inc., USA) under the control of a CMV promoter. Clones of cells expressing LOR-1 were selected using hygromycine and assayed for LOR-1 expression using the polyclonal antisera described below. [0176]
  • Construction of platelet factor-4 affinity columns and purification of LOR-1 on such columns: PF4 was coupled to sepharose using a modification of the method of Miron and Wilchek (Wilchek and Miron, 1982) as previously described for vascular endothelial growth factor (Soker et al., 1998). Serum free conditioned medium was collected from [0177] 35S-methionine labeled MCF-7 cells which over-expressed LOR-1. The conditioned medium was passed through the column twice. The column was washed with phosphate buffered saline (300 mM NaCl, pH-7.2) and eluted with PBS (containing 2M NaCl).
  • Experiments with nude mice: Modified or parental MCF-7 cells (10[0178] 7 cells per animal) were implanted under the skin of nude mice. A pellet of slow release estrogen was implanted 1 cm away as previously described (Zhang et al., 1995). Tumors were measured periodically, following which, tumors at least 1 cm in size were removed and immuno-histologicaly analyzed using a commercial antibody directed against a factor 8 like antigen which served as a specific marker for endothelial cells.
  • In-situ hybridization: Fragments encompassing nucleotides 922-1564 of LOR-1, nucleotides 976-1391 of LOL, nucleotides 400-950 of LO and nucleotides 1061-1590 of LOR-2 (as numbered from the ATG codon of these sequences) where each independently subcloned into the Bluescript SK and KS (Stratagene) vectors. A DIG cRNA labeling kit of Boehringer-Mannheim was used to transcribe sense (s) and antisense (as) digoxigenin-labeled cRNA probes from the T7 promoter of the Bluescript constructs. Hybridization and subsequent detection of hybridized probes was carried out essentially as previously described (Cohen et al., 2001). [0179]
  • Anti-LOR-1 polyclonal antisera: Antisera was generated by injecting a recombinant peptide containing the C-[0180] terminal 200 amino-acids of LOR-1 (amino acids 540-744 of SEQ ID NO: 2) into female rabbits. Serum was collected 10 days following each injection and an immunoglobulin fraction was purified using a protein A Sepharose affinity column (Pharmacia).
  • Results [0181]
  • LOR-1 purification: A PF4 affinity column was used to detect endothelial cell proteins which specifically interact with PF4. [0182]
  • Two PF4 binding proteins were detected in conditioned medium of human umbilical vein endothelial cells (HUVEC), whereas PF4 binding proteins were not detected in detergent extracts of endothelial cells. [0183]
  • Two liters of conditioned medium enabled a partial purification of one such binding protein which eluted from the column at a relatively high salt concentrations (0.4-0.5 M NaCl). [0184]
  • Further purification of this protein was performed using reverse phase high pressure liquid chromatography and SDS/PAGE chromatography. The PF4 binding protein did not bind to heparin nor was it a heparan-sulfate proteoglycan since heparinase digestion failed to change its mobility in SDS/PAGE experiments. [0185]
  • Partial sequencing and database comparison revealed that the PF4 binding protein of the present invention (LOR-1) belongs to a family of proteins containing a lysyl-oxidase like domain (Kim et al., 1995; Kim et al., 1999). Lysyl-oxidases are copper dependent enzymes that participate in the synthesis of the extracellular matrix by catalyzing the formation of covalent bonds between lysines of adjacent collagen or elastin fibers. [0186]
  • The full length amino acid sequence of LOR-1 (as deduced from the isolated cDNA sequence) displayed a high degree of identity to WS9-14, a protein over expressed in senescent fibroblasts, in several types of adherent cells (but not in non-adherent cells) and in fibroblasts, in which it was correlated to pro-collagen I-a1 expression levels, as well as being induced by TGF-β and inhibited by phorbol esters and retinoic acid (Saito et al., 1997). [0187]
  • The role of LOR-1 in tumor development: Recombinant LOR-1 expressed in PAE cells specifically bound with the PF4 affinity column (FIG. 1). Since LOR-1 is a member of the LO family it was hypothesized that it participates in ECM formation during angiogenesis. Furthermore it was also hypothesized that PF4 suppresses or inhibits the pro-angiogenic activity of LOR-1 thus inhibiting the later stages of blood vessel formation and as a result limiting tumor growth. [0188]
  • LOR-1 expression: In-situ hybridization demonstrated that LOR-1 is expressed in a wide variety of tissues and cell types including fibroblasts, adipocytes, nerve cells, endothelial cells and a variety of epithelial cells. Several cell types, such as liver hepatocytes, did not express LOR-1; of the 4 LO family members examined (all except for LoxC) LOR-1 was the only one expressed in endothelial cells of blood vessels. [0189]
  • LOR-1 and cancer: as shown in FIG. 2, a direct correlation between the expression levels of LOR-1 and the metastatic properties of breast cancer derived cell lines was demonstrated herein. [0190]
  • Since the epithelial cells which line the milk ducts of normal breast tissue (from which most breast tumors arise) express large amounts of LOR-1 it is possible that the less metastatic lines lost LOR-1 expression rather than gained it. [0191]
  • To substantiate its role in metastasis, LOR-1 cDNA was expressed in non-metastatic breast cancer derived MCF-7 cell lines which do not normally express LOR-1. The expression of LOR-1 was examined using rabbit polyclonal antibodies generated as described above (FIG. 3). [0192]
  • A control cell line which was transfected with an empty expression vector, and an MCF-7 cell line expressing LOR-1 were implanted under the skin of immune deficient mice along with an estrogen slow release pellet as described above. Estrogen was added since the development of tumors from this non-metastatic cell line is estrogen dependent (Mcleskey et al., 1993). [0193]
  • The rate of tumor development in the mice was continuously monitored (FIG. 4); [0194] tumors 1 cm in size were excised and subjected to histological analysis as described above. Interestingly, the rate of tumor development varied between the two cell lines, with some tumors exhibiting slower growth in the LOR-1 expressing MCF-7 and yet other exhibiting slower growth in the control cells.
  • In order to overcome expression level problems, LOR-1 cDNA was placed under the control of a tetracycline induced promoter (The TET-off system). Such a construct will enable to determine conclusively whether the reduced rate of tumor growth observed in LOR-1 expressing cells is indeed caused by LOR-1. [0195]
  • Tumors expressing large amounts of LOR-1 were sectioned and stained with an antibody directed against factor 8 like antigen, a specific marker of endothelial cells. Control tumor tissue predominantly stained at the capsule around the tumor while in the LOR-1 expressing tumors pronounced staining was observed in inner regions of the tumor tissue (FIG. 5[0196] a and FIG. 5b respectively).
  • The role of LOR-1 in Wilson's disease and in other chronic liver diseases: Normal and diseased liver tissue were probed with LOR-1 sense (FIGS. 6[0197] a and 6 c) and antisense probes (FIGS. 6b and 6 d). Normal liver tissues expresses very low levels of LOR-1 (FIG. 6b). However, fibrotic liver tissues such as those observed in Wilson's disease, exhibit a strong increase in hepatocyte expression of LOR-1 (FIG. 6d).
  • Expression of LOR-1 in chick embryos: FIG. 7 illustrates LOR-1 expression of LOR-1 mRNA in blood vessels of a developing chick embryo. Whole-mount in-situ hybridization of 4 day old chick embryos revealed LOR-1 mRNA expression in blood vessels located in the amnion (arrow). [0198]
  • The Lysyl oxidase family: A homology comparison between five members of the lysyl oxidase family which includes the LO and LOL subfamily and the LOR-1 and LOR-2 subfamily revealed a strong homology at the C-terminal portion which includes the conserved lysyl oxidase motif. LOR-1 and LOR-2 are characterized by long N-terminal stretches which are not found in LO and LOL. [0199]
  • Example 2 MCF-7 Breast Cancer Cells Expressing Recombinant Lysyl Oxidase Related Protein-1 (LOR-1) form Invasive Tumors Characterized by Extensive Fibrosis
  • A study was conducted in efforts to further substantiate and characterize the role of LOR-1 in inhibiting metastasis and tumor fibrosis. [0200]
  • Materials and Methods [0201]
  • Estrogen pellets (17β-estradiol, 0.72 mg/pellet, 60-days release) were from Innovative Research of America, FL, USA. Masson Trichrome stain kit was purchased from Bio-Optica (Milano, Italy), Reverse Transcriptase, G418 were from GIBCO BRL (U.K.), Hygromycin B, Tetracycline hydrochloride, Reticulum stain kit fast green and Sirius red (direct red 80) were from Sigma (USA) , Restriction enzymes, T4 ligase were from New England Biolabs (USA). The bacterial expression vector pQE-30 and the nickel affinity column were obtained from Qiagen (Germany) , [0202] 32p-dATP was purchased from NEN (USA). Monoclonal anti cytokeratin-7 (CAM 5.2) coupled to FITC was acquired from Becton Dickinson (USA), and monoclonal mouse anti vimentin (clone V9) was purchased from DAKO Denmark). Anti FITC alkaline phosphates-conjugated was purchased from ROCHE (USA), CAS block, citrate and EDTA antigen retrieval buffers were from Zymed (USA).
  • Cell culture: MCF-7 breast cancer cells were kindly provided by Dr. Hadasa Degani (Weizmann Institute, Israel). The MDA-MB-435 breast cancer cell line was kindly provided by Dr. Israel Vlodavsky (Technion, Israel). The MDA-MB-23 1 cells were kindly provided by Dr. Michael Klagsbrun (Harvard University, USA). These cell lines were routinely cultured in Dulbecco's modified eagle medium supplemented with gentamicin, amphotericin, glutamine and 10% Fetal Calf Serum (FCS). Human umbilical vein derived endothelial cells were isolated and cultured as described (Neufeld and Gospodarowics, 1988). Tissue culture media, sera, and cell culture supplements were from Beth-Haemek Biological Industries, Israel, or from Gibco-BRL. MCF-7 TetOff cells (Clontech, USA) containing the tetracycline trans-activator (tTA) were grown in DMEM medium containing 10% Tet system approved fetal calf serum (Clontech), in the presence of 100 μg/ml G418, 150 μg/ml Hygromycin B and 1 μg/ml Tetracyclin. [0203]
  • Cloning of the LOR1 and LOR2 cDNA: Total RNA (4 μg) from HUVEC cells (for LOR1) or melanoma cells (for LOR2) was reversed transcribed using MMLV reverse transcriptase (GIBCO BRL) as described (Chomczynski and Sacchi, 1987). The LOR-1 and LOR-2 cDNAs were amplified using the Expand Long High Fidelity PCR system (ROCHE) and the following pairs of amplification primers: For LOR-1 SEQ ID NOs: 10 and 11, and for LOR-2 SEQ ID NOs: 12 and 13. The 2.3 kb cDNA of LOR-1 (SEQ ID NO: 1) and the 2.26 KB of LOR-2 (SEQ ID NO: 4) were subcloned into the pGEM-T Easy vector (Promega) by T-A cloning. [0204]
  • Generation of polyclonal antibodies against human LOR1: A cDNA fragment containing nucleotides 1641-2253 of LOR1 (SEQ ID NO: 14) was amplified using the Expand High Fidelity PCR kit and a pair of amplification primers (SEQ ID NOs: 15 and 16). The PCR product was subcloned into the pGEM-T Easy vector (Promega) by T-A cloning. [0205]
  • A 613 bp LOR1 cDNA fragment (SEQ ID NO: 14) was digested with the Sph-I and Hind III restriction enzymes and ligated into the bacterial expression vector pQE-30, which added an in-frame sequence encoding a six histidine (6×His) tag to the 5′ end of the insert. The resulting plasmid was used to produce a recombinant, 6×His tagged 23 kDa peptide (SEQ ID NO: 17). The peptide was purified from bacterial cell extracts using nickel affinity chromatography, and further purified using SDS-PAGE. The gel was electroblotted onto nitrocellulose and the band containing the peptide was cut out from the blot, solubilized in DMSO, and used to immunize rabbits. Antiserum was affinity purified on protein-A sepharose followed by affinity purification on a column to which the recombinant peptide was coupled using a previously described method (Wilchek and Miron, 1982). The antibody was eluted from the column using 0.1 M glycine at [0206] pH 3.
  • Transfections: To constitutively express LOR1, the full length LOR1 cDNA (SEQ ID NO: 1), was digested out of the pGEM-T easy vector (promega) with Hind III and XbaI (which were incorporated into the primers used for the cloning of the LOR1 cDNA) and ligated into the mammalian expression vector pcDNA3.1 Hygro (Invitrogen, USA) to generate the expression vector pcDNA-LOR1. Empty pCDNA3. 1 Hygro plasmid or pcDNA-LOR1 plasmid (10-20 μg) were stably transfected into MCF-7 cells using electroporation with a BioRad gene pulser (960 μF, 0.28 V). Stable transfectants were selected using 300 μg/ml hygromycin B. Clones expressing recombinant LOR1 were obtained in two consecutive stable transfections and screened for LOR1 expression using our anti-LOR-1 polyclonal antibodies. Conditioned medium was collected after 48 hours from transfected cells and LOR-1 expression was monitored using western blot is analysis (FIG. 10A). [0207]
  • To inducively express LOR1, full length LOR1 cDNA (SEQ ID NO: 1) was cloned intQ the pTET-Splice vector (Clontech), which enables an inducible expression under the control of tetracycline (Tet off system). The pTET-Splice plasmid DNA was digested with Hind III and SpeI and ligated to the 2.3 kb HLOR1 cDNA fragment which was rescued out of the pCDNA-LOR1 plasmid using Hind III and XbaI. The resultant plasmid, which was designated as pTET-LOR1, was co-transfected into MCF-7 TetOff cells along with pTK-Hygro at a ratio of 20:1 respectively. LOR-1 expressing cells were selected in medium containing 100 μg/ml G418, 150 μg/ml hygromycin B and 1 μg/ml tetracycline. Stable transfectants were screened for inducible expression of LOR1 using western blot analysis 48 hours following removal of the tetracycline from the growth media. The clone having the highest induction levels in the absence of tetracycline and lowest basal expression levels in the presence of tetracycline was selected and designated MCF-7/Tet-LOR1. [0208]
  • C6 glioma cells were transfected and screened for LOR1 expression as described above. [0209]
  • Northern blot Analysis: Total RNA was extracted from cultured cells using Tri-Reagent (MRC, Cincinnati) according to the manufacture's instructions. Total RNA (15 μg) was loaded on a 1.2% agarose gel and Northern blot analysis was carried out as previously described (Cohen et al., 2001). LOR-1 and LOR-2 [0210] 32P labeled cDNA fragments; nucleotides 1-660 of SEQ ID NO: 18 and 1061-1590 of SEQ ID NO: 19 (respectively) were used as probes.
  • Protein blot analysis: Serum free conditioned media (40 μl) was separated on a 8% SDS-PAGE gel and the proteins were electroblotted onto a nitrocellulose filter using semi-dry electroblotting. The filter was blocked for 1 hour at room temperature with TBST buffer containing 10 mM Tris-HCl (pH-7.0), 0.15 M NaCl, and 0.3% Tween-20 supplemented with 10% low fat milk. The filter was incubated over night at 4° C. with affinity purified rabbit anti-LOR1 polyclonal antibody in TBST (1:2500). The blot was subsequently washed 3 times in TBST and incubated with goat anti-rabbit IgG peroxidase-conjugated secondary antibodies for 1 hour at room temperature. Bound antibody was visualized using the ECL detection system (Biological Industries, Israel). [0211]
  • Nude Mice Experiments: Slow release pellets containing 17β-estradiol (0.72 mg/pellet, 60 day release, Innovative Research) were pre-implanted subcutaneously in 6-8 weeks old female athymic nude mice (CD1). MCF-7 cells (10[0212] 7 cells/mouse) were injected into the mammary fat pads. Tumor size was measured with a caliper once or twice a week, and tumor volume was determined using the following formula: volume=width2×length×0.52 (O'Reilly et al., 1999). Mice were sacrificed 4 weeks following injection of the MCF-7 cells. In other experiments, the tumors were excised when reaching a diameter of 0.8 cm. The primary tumor, the liver and the lungs were removed, weighted, fixed in 10% buffered formalin and embedded in paraffin.
  • The development of tumors from C6 glioma cells expressing recombinant LOR-1 was also studied. Cells (2×10[0213] 5 cells/animal) transfected with a control expression vector or with an expression vector containing LOR1 cDNA were injected subcutaneously in the hind limb. Mice were sacrificed 3 weeks after the injection of the cells. The primary tumors were removed, fixed in 10% buffered formalin, and embedded in paraffin for analysis.
  • Histology and Immunohistochemistry: Formalin-fixed, paraffin-embedded tissues were cut into serial sections of 5 μm each and used for immunohistochemistry. Sections were deparaffinized by heating to 60° C. for 1 hour, washed twice with xylen for 5 min. and rehydrated by consecutive washes in 100%, 95%, and 70% ethanol followed by a wash in water. Endogenous peroxidase activity was inhibited by a 15 minute incubation with 3% hydrogen peroxide in methanol, followed by washes with water and PBS. The sections were then antigen retrieved by heating twice for 10 minutes in a microwave oven to 90° C. in citrate buffer at pH-6.2 (for cytokeratin and vimentin antibodies) or in 1 mM EDTA buffer (for LOR1 antibody); blocking was performed using CAS block (zymed). Following blocking, the sections were incubated for 1.5 hr at room temperature with the following antibodies, all diluted with in antibody diluent reagent solution (Zymed): affinity purified anti-LOR1 antibody (1:30-1:50), monoclonal anti human cytokeratin-7 antibodies-FITC conjugated (1:50), or with monoclonal antibodies directed against vimentin (1:50) . The sections were then washed 3 times with TBST, and secondary detection was applied using anti FITC alkaline phosphates-conjugate (Roche) at a 1:200 dilution (for anti cytokeratin) or DAKO Envision detection system (anti rabbit or anti mouse—HRP). Sections were developed in 3-amino-9-ethylcarbazole (AEC, DAKO) solution, counterstained by Hematoxylin, and photographed under a microscope. In control experiments the primary antibodies were omitted. Masson Trichrome and reticulum stains were according to manufacture's protocol. For Sirius red stain sections were incubated for 5 min. with 0.03% (w/v) fast green solution, rinsed twice with 1% acetic acid, incubated for 15 min. in 0.1% Sirius red solution, rinsed as above, dehydrated and examined under polarizing light microscope. [0214]
  • Results [0215]
  • LOR-1 is expressed in highly metastatic breast cancer derived cell types but not in non-metastatic MCF-7 cells: Desmoplasia and formation of fibrotic foci in breast cancer tumors is associated with the transition from a localized, relatively benign tumor to an invasive/metastatic tumor (Colpaert et al., 2001; Hasebe et al., 2000). Lysyl-oxidases contribute to the deposition of collagen by covalently cross-linking collagen monomers (Smith-Mungo and Kagan, 1998). To find out whether expression of lysyl-oxidases is associated with the invasive/metastatic phenotype several human breast cancer derived cell types have been screened for the expression of lysyl-oxidases. Northern blot analysis revealed that the LOR-1 gene is expressed in the highly malignant, hormone independent MDA-MB-231 and MDA-MB-435 cells (Price et al., 1990) but not in hormone dependent non-metastatic MCF-7 cells (FIG. 9[0216] a). LOR-2 on the other hand was expressed only in MDA-MB-435 cells but not in the MDA-MB-231 or in MCF-7 cells (FIG. 9b).
  • Highly [0217] malignant grade 1 breast carcinoma cells do not express LOR-1 while highly malignant cells of grade 3 carcinomas express LOR-1: LOR-1 expression was confined to the milk ducts in both normal breast (FIG. 9c) and in in-situ ductal carcinoma (FIG. 9d). However, in grade 1 well-differentiated ductal breast carcinoma, wherein the tumor cells migrate out of the ducts to form pseudo ducts, the cells do not express LOR-1 (FIG. 9e, black arrows). On the other hand, the cells of grade 3 ductal breast carcinoma tumors, a highly malignant tumor characterized by desmoplasia, express high levels of LOR-1 (FIG. 9f).
  • Tumors generated in mice from LOR-1 MCF-7 transfected cells have a slower growth rate. In order to determine whether LOR-1 expression contributes to the progression of breast tumors and to the invasive/metastatic phenotype, non-invasive MCF-7 cells have been transfected with an expression plasmid containing LOR-1 cDNA (SEQ ID NO: 1). Several transfectant clones have been isolated and selected for their LOR-1 expression. The conditioned medium of two LOR-1 expressing clones (clone 12 and 24) and of a clone transfected with an expression vector alone (vec) was assayed with LOR-1 antibodies directed against the C-terminal portion of LOR-1 (SEQ ID NO: 17). Western blot analysis revealed higher levels of LOR-1 expression in [0218] clone 12 cells as compared with clone 24 cells, and no expression in cells transfected with the expression vector alone (FIG. 10a).
  • In addition, both LOR-1 expressing cells displayed extra protein bands of about 70 kDa (FIGS. 10[0219] a, b) suggesting that LOR-1 may undergo proteolytic processing like other members of the lysyl-oxidase family (Borel et al., 2001; Panchenko et al., 1996). To verify that the low molecular weight forms are produced as a result of post-translational processing, LOR-1 cDNA has been expressed in MCF-7 cells under the control of a Tetracycline inducible promoter (Shockett and Schatz, 1996). It can be seen that once the tetracycline inhibition is removed, the cells start to produce full length LOR-1 which is then converted into a shorter, 70 kDa C-terminal containing form (FIG. 10b). It is not yet clear whether all of these forms are enzymatically active.
  • To substantiate its role in tumor growth and metastasis LOR-1 producing cells and cells transfected with expression vector alone were pre-implanted subcutaneously in nude mice (described above). Interestingly, the growth rate of tumors containing LOR-1 expressing cells was retarded as compared with that of tumors developed from parental cells (not shown) or cells transfected with the expression vector alone (FIGS. 10[0220] c, d).
  • To determine if the decreased tumor growth rate was a result of slower proliferation, the proliferation rate of empty vector transfected MCF-7 with that of [0221] clone 12 and clone 24 cells were also compared; significant differences in their rates of proliferation was not detected (data not shown).
  • Tumors that develop from LOR-1 producing MCF-7 cells contain many necrotic and fibrotic foci rich in collagen deposits: Hematoxilin-eosin staining of tumor sections revealed major necrosis in tumors generated from LOR-1 expressing MCF-7 cells (FIG. 11[0222] b) and only a few necrotic areas in tumors generated from parental (not shown) or MCF-7 cells transfected with expression vector alone (FIG. 11a).
  • LOR-1 expressing tumors also contained extensive fibrotic areas mainly composed of host derived cells such as mouse fibroblasts rather than MCF-7 cells. These cells are easily distinguishable from the host cells since they do not react with an antibody against human keratin 7 (FIG. 11[0223] c).
  • Since LOR-1 was shown to oxidize lysine residues on collagen-I (Vadasz et al., 2002) it was anticipated that it may induce collagen cross-linking and deposition. However, this activity is probably depended on the existence of collagen since LOR-1 expressing MCF-7 cells grown in culture do not produce more collagen than parental MCF-7 cells (data not shown). To further understand the involvement of LOR-1 in tumor progression tumors derived from LOR-1 expressing MCF-7 cells have been stained with Mason's Trichrome, a reagent that reacts primarily with collagen-I and produces an azure color (Pinder et al., 1994). (1) Tumors that developed from parental or MCF-7 cells transfected by an expression vector alone contained limited amounts of collagen that was scattered between the tumor cells (FIG. 11[0224] d, arrows). In-contrast, tumors that developed from clone 12 or clone 24 LOR-1 expressing MCF-7 cells contained denser deposits of collagen between the tumor cells (FIG. 11e). Furthermore, the fibrotic and necrotic areas in these tumors appeared choke full with collagen fibers (FIG. 11f). In addition, while the blood vessels within tumors that developed from vector-transfected MCF-7 cells remained almost unstained (FIG. 11g, arrows), the blood vessels in tumors derived from LOR-1 expressing MCF-7 cells were sheathed by a thick layer of collagen (FIG. 11h, arrows).
  • These experiments indicate that LOR-1 affects both collagen-I production and deposition in MCF-7 derived breast cancer tumors. To substantiate the involvement of LOR-1 in collagen deposition C6 glioma cells were also subcutaneously injected into mice. While tumors generated from C6-glioma cells did not contain large amounts of collagen, tumors generated from LOR-1 expressing C6-glioma cells were rich with collagen (data not shown). These results indicate a more general effect for LOR-1 on collagen-I deposits in tumors. In addition, Reticulum staining of collagen-III revealed that tumors generated from LOR-1 expressing MCF-7 or C6-glioma cells contained thicker and higher concentrations of collagen-Ill fibers (FIG. 12[0225] b, 12 d) as compared with tumors generated from cells transfected with the expression vector alone (FIGS. 12a, c).
  • Expression of LOR-1 in MCF-7 cells transforms the cells into invasive cells in-vivo: It was reported that the appearance of fibrotic foci in breast cancer tumors correlates with their degree of invasiveness (Hasebe et al., 2000). To substantiate the involvement of LOR-1 in tumor invasiveness human keratin-7 staining was employed. Tumors generated from MCF-7 cells transfected by the expression vector alone were surrounded by thick capsules with sharp borders. No staining of human keratin-7 was observed within the capsule (FIG. 13[0226] a) or in between blood vessels, nerves and muscles located adjacent to the capsules (FIG. 13b, arrows). In contrast, in tumors generated from LOR-1 expressing MCF-7 cells, human keratin-7 positive cells were observed within the capsule (FIG. 13c). Furthermore, in many areas the tumor cells migrated on-mass through the capsule and invaded muscles (FIG. 13d), nerves (FIG. 13e) and blood vessels (FIG. 13f). The invading cells were identified as the transfected LOR-1 expressing MCF-7 cells using an anti-LOR-1 antibody (FIG. 13g). These observations provide a strong evidence that the production of LOR-1 by breast cancer tumor cells contribute to the transition from localized non-invasive tumors to invasive tumors. Furthermore, aggregates of tumor cells were also detected inside lymph vessels adjacent to the tumors indicating that the LOR-1 expressing MCF-7 cells are metastatic (Luna, 1968).
  • Although the invention has been described in conjunction with specific embodiments thereof, it is evident that many alternatives, modifications and variations will be apparent to those skilled in the art. Accordingly, it is intended to embrace all such alternatives, modifications and variations that fall within the spirit and broad scope of the appended claims. All publications, patents, patent applications and sequences identified by their accession numbers mentioned in this specification are herein incorporated in their entirety by reference into the specification, to the same extent as if each individual publication, patent, patent application or sequence identified by their accession number was specifically and individually indicated to be incorporated herein by reference. In addition, citation or identification of any reference in this application shall not be construed as an admission that such reference is available as prior art to the present invention. [0227]
  • REFERENCES CITED (Additional References are Cited in the Text)
  • 1. Folkman, J. (1990) What is the evidence that tumors are angiogenesis dependent. J. Nat. Cancer Inst. 82, 4-7. [0228]
  • 2. Hanahan, D. and Folkman, J. (1996) Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 86, 353-364. [0229]
  • 3. Boehm, T., Folkman, J., Browder, T., and Oreilly, M. S. (1997) Antiangiogenic therapy of experimental cancer does not induce acquired drug resistance. [0230] Nature 390, 404-407.
  • 4. Bergers, G., Javaherian, K., Lo, K. M., Folkman, J., and Hanahan, D. (1999) Effects of angiogenesis inhibitors on multistage carcinogenesis in mice. Science 284, 808-812. [0231]
  • 5. Zetter, B. R. (1998) Angiogenesis and tumor metastasis. Annu. Rev. Med. 49:407-424, 407-424. [0232]
  • 6. Weidner, N. (1998) Tumoural vascularity as a prognostic factor in cancer patients: The evidence continues to grow. J. Pathol. 184, 119-122. [0233]
  • 7. Degani, H., Gusis, V., Weinstein, D., Fields, S., and Strano, S. (1997) Mapping pathophysiological features of breast tumors by MRI at high spatial resolution. Nature Med. 3, 780-782. [0234]
  • 8. Guidi, A. J., Schnitt, S. J., Fischer, L., Tognazzi, K., Harris, J. R., Dvorak, H. F., and Brown, L. F. (1997) Vascular permeability factor (vascular endothelial growth factor) expression and angiogenesis in patients with ductal carcinoma in situ of the breast. [0235] Cancer 80, 1945-1953.
  • 9. Balsari, A., Maier, J. A. M., Colnaghi, M. I., and Menard, S. (1999) Correlation between tumor vascularity, vascular endothelial growth factor production by tumor cells, serum vascular endothelial growth factor levels, and serum angiogenic activity in patients with breast carcinoma. Lab. Invest. 79, 897-902. [0236]
  • 10. Klauber, N., Parangi, S., Flynn, E., Hamel, E., and D'Amato, R. J. (1997) Inhibition of angiogenesis and breast cancer in mice by the microtubule inhibitors 2-methoxyestradiol and taxol. Cancer Res. 57, 81-86. [0237]
  • 11. Harris, A. L., Zhang, H. T., Moghaddam, A., Fox, S., Scott, P., Pattison, A., Gatter, K., Stratford, I., and Bicknell, R. (1996) Breast cancer angiogenesis—New approaches to therapy via antiangiogenesis, hypoxic activated drugs, and vascular targeting. Breast Cancer Res. Treat. 38, 97-108. [0238]
  • 12. Weinstatsaslow, D. L., Zabrenetzky, V. S., Vanhoutte, K., Frazier, W. A., Roberts, D. D., and Steeg, P. S. (1994) Transfection of [0239] thrombospondin 1 complementary DNA into a human breast carcinoma cell line reduces primary tumor growth, metastatic potential, and angiogenesis. Cancer Res. 54, 6504-6511.
  • 13. Neufeld, G., Cohen, T., Gengrinovitch, S., and Poltorak, Z. (1999) Vascular endothelial growth factor (VEGF) and its receptors. FASEB J. 13, 9-22. [0240]
  • 14. Brooks, P. C., Montgomery, A. M. P., Rosenfeld, M., Reisfeld, R. A., Hu, T. H., Klier, G., and Cheresh, D. A. (1994) Integrin alpha(v)beta(3) antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels. Cell 79, 1157-1164. [0241]
  • 15. Brooks, P. C., Silletti, S., Von Schalscha, T. L., Friedlander, M., and Cheresh, D. A. (1998) Disruption of angiogenesis by PEX, a noncatalytic metalloproteinase fragment with integrin binding activity. Cell 92, 391-400. [0242]
  • 16. O'Reilly, M. S., Boehm, T., Shing, Y., Fukai, N., Vasios, G., Lane, W. S., Flynn, E., Birkhead, J. R., Olsen, B. R., and Folkman, J. (1997) Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell 88, 277-285. [0243]
  • 17. Oreilly, M. S., Holmgren, L., Chen, C., and Folkman, J. (1996) Angiostatin induces and sustains dormancy of human primary tumors in mice. Nature Med. 2, 689-692. [0244]
  • 18. Tanaka, T., Manome, Y., Wen, P., Kufe, D. W., and Fine, H. A. (1997) Viral vector-mediated transduction of a modified [0245] platelet factor 4 cDNA inhibits angiogenesis and tumor growth. Nature Med. 3, 437-442.
  • 19. Maione, T. E., Gray, G. S., Petro, J., Hunt, A. J., Donner, A. L., Bauer, S. I., Carson, H. F., and Sharpe, R. J. (1990) Inhibition of angiogenesis by recombinant human platelet factor-4 and related peptides. Science 247, 77-79. [0246]
  • 20. Neufeld, G., Akiri, G., and Vadasz, Z. (2000) in Platelet Factor 4 (PF4).The Cytokine Reference: A compendium of cytokines and other mediators of host defence (Oppenheim, J. J. and Feldmann, M. eds) Academic Press. [0247]
  • 21. Gengrinovitch, S., Greenberg, S. M., Cohen, T., Gitay-Goren, H., Rockwell, P., Maione, T. E., Levi, B., and Neufeld, G. (1995) Platelet factor-4 inhibits the mitogenic activity of VEGF-121 and VEGF-165 using several concurrent mechanisms. J. Biol. Chem. 270, 15059-15065. [0248]
  • 22. Brown, K. J. and Parish, C. R. (1994) Histidine-rich glycoprotein and [0249] platelet factor 4 mask heparan sulfate proteoglycans recognized by acidic and basic fibroblast growth factor. Biochemistry 33, 13918-13927.
  • 23. Gupta, S. K. and Singh, J. P. (1994) Inhibition of endothelial cell. [0250]
  • Proliferation by platelet factor-4 involves a unique action on S phase progression. J. Cell Biol. 127, 1121-1127. [0251]
  • 24. Watson, J. B., Getzler, S. B., and Mosher, D. F. (1994) [0252] Platelet factor 4 modulates the mitogenic activity of basic fibroblast growth factor. J. Clin. Invest. 94, 261-268.
  • 25. Maione, T. E., Gray, G. S., Hunt, A. J., and Sharpe, R. J. (1991) Inhibition of tumor growth in mice by an analogue of [0253] platelet factor 4 that lacks affinity for heparin and retains potent angiostatic activity. Cancer Res. 51, 2077-2083.
  • 26. Sharpe, R. J., Byers, H. R., Scott, C. F., Bauer, S. I., and Maione, T. E. (1990) Growth inhibition of murine melanoma and human colon carcinoma by recombinant [0254] human platelet factor 4. J. Natl. Cancer Inst. 82, 848-853.
  • 27. Saito, H., Papaconstantinou, J., Sato, H., and Goldstein, S. (1997) Regulation of a novel gene encoding a lysyl oxidase-related protein in cellular adhesion and senescence. J. Biol. Chem. 272, 8157-8160. [0255]
  • 28. Kim, Y., Boyd, C. D., and Csiszar, K. (1995) A new gene with sequence and structural similarity to the gene encoding human lysyl oxidase. J. Biol. Chem. 270, 7176-7182. [0256]
  • 29. Kim, Y. H., Peyrol, S., So, C. K., Boyd, C. D., and Csiszar, K. (1999) Coexpression of the lysyl oxidase-like gene (LOXL) and the gene encoding type III procollagen in induced liver fibrosis. J. Cell Biochem. 72, 181-188. [0257]
  • 30. Rabinovitz, M. (1999) Angiogenesis and its inhibition: the copper connection. J. Natl. Cancer Inst. 91, 1689-1690. [0258]
  • 31. Hansell, P., Maione, T. E., and Borgstrom, P. (1995) Selective binding of [0259] platelet factor 4 to regions of active angiogenesis in vivo. Amer. J. Physiol-Heart. Circ. Phy. 38, H829-H836.
  • 32. Reiser, K., McCormick, R. J., and Rucker, R. B. (1992) Enzymatic and nonenzymatic cross-linking of collagen and elastin. FASEB J. 6, 2439-2449. [0260]
  • 33. Jang, W., Hua, A., Spilson, S. V., Miller, W., Roe, B. A., and Meisler, M. H. (1999) Comparative sequence of human and mouse BAC clones from the mnd2 region of chromosome 2p 13. Genome Res. 9, 53-61. [0261]
  • 34. Yoshida, D., Ikeda, Y., and Nakazawa, S. (1995) Copper chelation inhibits tumor angiogenesis in the experimental 9L gliosarcoma model. Neurosurgery 37, 287-292. [0262]
  • 35. Borgstroem, P., Discipio, R., and Maione, T. E. (1998) [0263] Recombinant platelet factor 4, an angiogenic marker for human breast carcinoma. Anticancer Res. 18, 4035-4041.
  • 36. Shweiki, D., Neeman, M., Itin, A., and Keshet, E. (1995) Induction of vascular endothelial growth factor expression by hypoxia and by glucose deficiency in multicell spheroids: Implications for tumor angiogenesis. Proc. Natl. Acad. Sci. USA 92, 768-772. [0264]
  • 37. Rak, J., Mitsuhashi, Y., Bayko, L., Filmus, J., Shirasawa, S., Sasazuki, T., and Kerbel, R. S. (1995) Mutant ras oncogenes upregulate VEGF/VPF expression: Implications for induction and inhibition of tumor angiogenesis. Cancer Res. 55, 4575-4580. [0265]
  • 38. Koch, A. E. (1998) Angiogenesis—Implications for rheumatoid arthritis. Arthritis Rheum. 41, 951-962. [0266]
  • 39. Paleolog, E. M. and Fava, R. A. (1998) Angiogenesis in rheumatoid arthritis: implications for future therapeutic strategies. Springer Semin. Immunopathol. 20, 73-94. [0267]
  • 40. Miller, J. W., Adamis, A. P., and Aiello, L. P. (1997) Vascular endothelial growth factor in ocular neovascularization and proliferative diabetic retinopathy. Diabetes Metab. Rev. 13, 37-50. [0268]
  • 41. Detmar, M., Brown, L. F., Claffey, K. P., Yee, K. T., Kocher, O., Jackman, R. W., Berse, B., and Dvorak, H. F. (1994) Overexpression of vascular permeability factor/vascular endothelial growth factor and its receptors in psoriasis. J. Exp. Med. 180, 1141-1146. [0269]
  • 42. Creamer, D., Allen, M. H., Sousa, A., Poston, R., and Barker, J. N. W. N. (1997) Localization of endothelial proliferation and microvascular expansion in active plaque psoriasis. Br. J. Dermatol. 136, 859-865. [0270]
  • 43. Lie, J. T. (1992) Vasculitis simulators and vasculitis look-alikes. Curr. Opin. Rheumatol. 4, 47-55. [0271]
  • 44. Klipple, G. L. and Riordan, K. K. (1989) Rare inflammatory and hereditary connective tissue diseases. Rheum. Dis. Clin. North Am. 15, 383-398. [0272]
  • 45. Brahn, E., Lehman, T. J. A., Peacock, D. J., Tang, C., and Banquerigo, M. L. (1999) Suppression of coronary vasculitis in a murine model of Kawasaki disease using an angiogenesis inhibitor. Clin. Immunol. Immunopathol. 90, 147-151. [0273]
  • 46. Cid, M. C., Grant, D. S., Hoffman, G. S., Auerbach, R., Fauci, A. S., and Kleinman, H. K. (1993) Identification of Haptoglobin as an Angiogenic Factor in Sera from Patients with Systemic Vasculitis. J. Clin. Invest. 91, 977-985. [0274]
  • 47. Hoffman, G. S., Filie, J. D., Schumacher, H. R., Jr., Ortiz-Bravo, E., Tsokos, M. G., Marini, J. C., Kerr, G. S., Ling, Q. H., and Trentham, D. E. (1991) Intractable vasculitis, resorptive osteolysis, and immunity to type I collagen in type VIII Ehlers-Danlos syndrome. Arthritis Rheum. 34, 1466-1475. [0275]
  • 48. Bauters, C. and Isner, J. M. (1997) The biology of restenosis. Prog. Cardiovasc. Dis. 40, 107-116. [0276]
  • 49. Begelman, S. M. and Olin, J. W. (2000) Fibromuscular dysplasia. Curr. Opin. Rheumatol. 12, 41-47. [0277]
  • 50. Palta, S., Pai, A. M., Gill, K. S., and Pai, R. G. (2000) New insights into the progression of aortic stenosis : implications for secondary prevention. Circulation 101, 2497-2502. [0278]
  • 51. Wilchek, M. Miron, T., 1982. Immobilization of enzymes and affinity ligands onto agarose via stable and uncharged carbamate linkages. Biochem. Int. 4, 629-635. [0279]
  • 52. Soker, S., Takashima, S., Miao, H. Q., Neufeld, G., Klagsbrun, M., 1998. Neuropilin-1 is expressed by endothelial and tumor cells as an isoform specific receptor for vascular endothelial growth factor. Cell 92, 735-745. [0280]
  • 53. Zhang, H. T., Craft, P., Scott, P. A. E., Ziche, M., Weich, H. A., Harris, A. L., Bicknell, R., 1995. Enhancement of tumor growth and vascular density by transfection of vascular endothelial cell growth factor into MCF-7 human breast carcinoma cells. J. Nat. Cancer Inst. 87, 213-219. [0281]
  • 54. Cohen, T., Gluzman-Poltorak, Z., Brodzky, A., Meytal, V., Sabo, E., Misselevich, I., Hassoun, M., Boss, J. H., Resnick, M., Shneyvas, D., Eldar, S., Neufeld, G., 2001. Neuroendocrine Cells along the Digestive Tract Express Neuropilin-2. Biochem. Biophys. Res. Commun. 284, 395-403. [0282]
  • 55. Mcleskey, S. W., Kurebayashi, J., Honig, S. F., Zwiebel, J., Lippman, M. E., Dickson, R. B., Kern, F. G., 1993. Fibroblast Growth Factor-4 Transfection of MCF-7 Cells Produces Cell Lines That Are Tumorigenic and Metastatic in Ovariectomized or Tamoxifen-Treated Athymic Nude Mice. Cancer Res. 53, 2168-2177. [0283]
  • 56. Nakamura et al. Cancer Res 60(3), 760-5, 2000. [0284]
  • 57. Szczylik et al (1991) Selective inhibition of leukemia cell proliferation by BCR-ABL antisense oligodeoxynucleotides. Science 253:562. [0285]
  • 58. Calabretta et al. (1991) Normal and leukemic hematopoietic cell manifest differential sensitivity to inhibitory effects of c-myc antisense oligodeoxynucleotides: an in vitro study relevant to bone marrow purging. Proc. Natl. Acad. Sci. USA 88:2351. [0286]
  • 59. Heikhila et al. (1987) A c-myc antisense oligodeoxynucleotide inhibits entry into S phase but not progress from G(0) to G(1). Nature, 328:445. [0287]
  • 60. Burch and Mahan (1991) Oligodeoxynucleotides antisense to the interleukin I receptor m RNA block the effects of interleukin I in cultured murine and human fibroblasts and in mice. J. Clin. Invest. 88:1190. [0288]
  • 61. Cook (1991) Medicinal chemistry of antisense oligonucleotides-future opportunities. Anti-Cancer Drug Design 6:585. [0289]
  • 62. Carthew R W. Gene silencing by double-stranded RNA. Curr Opin Cell Biol April 2001; 13(2):244-8. [0290]
  • 63. S. and Ikeda, J. E.: Trapping of mammalian promoters by Cre-lox site-specific recombination. DNA Res 3 (1996) 73-80. [0291]
  • 64. Bedell, M. A., Jenkins, N. A. and Copeland, N. G.: Mouse models of human disease. Part I: Techniques and resources for genetic analysis in mice. Genes and Development 11 (1997) 1-11. [0292]
  • 65. Bermingham, J. J., Scherer, S. S., O'Connell, S., Arroyo, E., Kalla, K. A., Powell, F. L. and Rosenfeld, M. G.: Tst-1/October-6/SCIP regulates a unique step in peripheral myelination and is required for normal respiration. Genes Dev 10 (1996) 1751-62. [0293]
  • 66. Welch P. J., Barber J. R., and Wong-Staal F. (1998) Expression of ribozymes in gene transfer systems to modulate target RNA levels. Curr. Opin. Biotechnol., 9(5):486-496. [0294]
  • 67. Bedell-Hogan, D., Trackman, P., Abrams, W., Rosenbloom, J., and Kagan, H. (1993) Oxidation, cross-linking, and insolubilization of recombinant tropoelastin by purified lysyl oxidase. [0295] J. Biol. Chem. 268, 10345-10350)
  • 68. Colpaert, C., Vermeulen, P., Van Marck, E., and Dirix, L. (2001) The presence of a fibrotic focus is an independent predictor of early metastasis in lymph node-negative breast cancer patients. Am. J. Surg. Pathol. 25, 1557 Hasebe, T., Mukai, K., Tsuda, H., and Ochiai, A. (2000) New prognostic histological parameter of invasive ductal carcinoma of the breast: Clinicopathological significance of fibrotic focus. [0296] Pathology International 50, 263-272
  • 69. Nishimura, R., Hasebe, T., Tsubono, Y., Ono, M., Sugitoh, M., Arai, T., and Mukai, K. (1998) The fibrotic focus in advanced colorectal carcinoma: a hitherto unrecognized histological predictor for liver metastasis. Virchows Arch. 433, 517-522 [0297]
  • 70. Ellenrieder, V., Alber, B., Lacher, U., Hendler, S. F., Menke, A., Boeck, W., Wagner, M., Wilda, M., Friess, H., Buchler, M., Adler, G., and Gress, T. M. (2000) Role of MT-MMPs and MMP-2 in pancreatic cancer progression. Int. J. Cancer 85, 14-20 [0298]
  • 71. Stamenkovic, I. (2000) Matrix metalloproteinases in tumor invasion and metastasis. Semin. Cancer Biol. 10, 415-433 [0299]
  • 72. Duffy, M. J., Maguire, T. M., Hill, A., McDermott, E., and O'Higgins, N. (2000) Metalloproteinases: role in breast carcinogenesis, invasion and metastasis. Breast Cancer Res. 2, 252-257 [0300]
  • 73. Vlodavsky, I. and Friedmann, Y. (2001) Molecular properties and involvement of heparanase in cancer metastasis and angiogenesis. J. Clin. Invest 108, 341-347 [0301]
  • 74. Schuppan, D., Ruehl, M., Somasundaram, R., and Hahn, E. G. (2001) Matrix as a modulator of hepatic fibrogenesis. Semin. Liver Dis. 21, 351-372 [0302]
  • 75. Sawada, S., Murakami, K., Murata, J., Tsukada, K., and Saiki, I. (2001) Accumulation of extracellular matrix in the liver induces high metastatic potential of hepatocellular carcinoma to the lung. Int. J. Oncol. 19, 65-70 [0303]
  • 76. Fire, A., Xu, S., Montgomery, M. K., Kostas, S. A., Driver, S. E., and Mello, C. C. (1998) Potent and specific genetic interference by double-stranded RNA in [0304] Caenorhabditis elegans. Nature 391, 806-811.
  • 77. Zamore, P. D., Tuschl, T., Sharp, P. A., and Bartel, D. P. (2000) RNAi: double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals. Cell 101, 25-33. [0305]
  • 78. Elbashir, S. M., Lendeckel, W. and Tuschl, T. (2001) RNA interference is mediated by 21- and 22-nucleotide RNAs. Genes Dev. 15, 188-200. [0306]
  • 79. Hammond, S. M., Bernstein, E., Beach, D. and Hannon, G. J. (2000) An RNA-directed nuclease mediates post-transcriptional gene silencing in Drosophila cells. Nature 404, 293-296 . [0307]
  • 80. Bernstein, E., Caudy, A. A., Hammond, S. M. and Hannon, G. J. (2001) Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 409, 363-366. [0308]
  • 81. Nykanen, A., Haley, B. and Zamore, P. D. (2001) ATP requirements and small interfering RNA structure in the RNA interference pathway. Cell 107, 309-321 [0309]
  • 82. Caplen, N. J., Fleenor, J., Fire, A. and Morgan, R. A. (2000) [0310]
  • 83. dsRNA-mediated gene silencing in cultured Drosophila cells: a tissue culture model for the analysis of RNA interference. Gene 252, 95-105. [0311]
  • 84. Ui-Tei, K., Zenno, S., Miyata, Y. and Saigo, K. (2000) Sensitive assay of RNA interference in Drosophila and Chinese hamster cultured cells using firefly luciferase gene as target. FEBS Lett. 479, 79-82. [0312]
  • 85. McCaffrey, A. P., Meuse, L., Pham, T -T. T., Conklin, D. S., Hannon, G. J., and Kay, M. A. (2002) Gene expression: RNA interference in adult mice. Nature 418, 38-39. [0313]
  • 86. Jacque, J -M., Triques, K., and Stevenson, M. (2002) Modulation of HIV-1 replication by RNA interference. Nature 418, 435-438. [0314]
  • 87. Jiang, M., and Milner, J. (2002) Selective silencing of viral gene expression in HPV-positive human cervical carcinoma cells treated with siRNA, aprimer of RNA interference. [0315] Oncogene 21, 6041-8.
  • 88. Wilda, M., Fuchs, U., Wossmann, W., and Borkhardt, A. (2002) Killing of leukemic cells with a BCR/ABL fusion gene by RNA interference (RNAi). [0316] Oncogene 21, 5716-24.
  • 89. Hohjoh, H. (2002) RNA interference (RNA(i)) induction with various types of synthetic oligonucleotide duplexes in cultured human cells. FEBS Lett. 521, 195-9. [0317]
  • 90. Leirdal, M., and Sioud, M. (2002) Gene silencing in mammalian cells by preformed small RNA duplexes. Biochem Biophys Res Commun 295, 744-8. [0318]
  • 91. Yang, D., Buchholz, F., Huang, Z., Goga, A., Chen, C. Y., Brodsky, F. M., and Bishop, J. M. (2002) Short RNA duplexes produced by hydrolysis with [0319] Escherichia coli RNase III mediate effective RNA interference in mammalian cells. Proc. Natl. Acad. Sci. U.S.A. 99, 9942-7.
  • 92. Brummelkamp, T. R., Bernards, R., Agami, R. (2002) A system for stable expression of short interfering RNAs in mammalian cells. Science 296: 550-53 [0320]
  • 93. Paddison, P. J., Caudy, A. A., Bernstein, E., Hannon, G. J., Conklin, D. S. (2002) Short hairpin RNAs (shRNAs) induce sequence-specific silencing in mammalian cells. Genes Dev. 16:948-58 [0321]
  • 94. Paul et al. (2002) Nature Biotech. 20: 505-08, [0322]
  • 95. Yu, J. Y., DeRuiter, S. L., Turner, D. L. (2002) RNA interference by expression of short-interfering RNAs and hairpin RNAs in mammalian cells. Proc. Natl. Acad. Sci. USA 99: 6047-52 [0323]
  • 96. Pinder, S. E., Ellis, I. O., Galea, M., O'Rouke, S., Blamey, R. W., and Elston, C. W. (1994) Pathological prognostic factors in breast cancer. III. Vascular invasion: relationship with recurrence and survival in a large study with long-term follow-up. [0324] Histopathology 24, 41-47
  • 97. Luna, L. G. (1968) Manual of histologic staining methods of the armed forces institute of pathology, McGraw-Hill, New-York. [0325]
  • 1 19 1 2325 DNA Homo sapiens 1 atggagaggc ctctgtgctc ccacctctgc agctgcctgg ctatgctggc cctcctgtcc 60 cccctgagcc tggcacagta tgacagctgg ccccattacc ccgagtactt ccagcaaccg 120 gctcctgagt atcaccagcc ccaggccccc gccaacgtgg ccaagattca gctgcgcctg 180 gctgggcaga agaggaagca cagcgagggc cgggtggagg tgtactatga tggccagtgg 240 ggcaccgtgt gcgatgacga cttctccatc cacgctgccc acgtcgtctg ccgggagctg 300 ggctatgtgg aggccaagtc ctggactgcc agctcctcct acggcaaggg agaagggccc 360 atctggttag acaatctcca ctgtactggc aacgaggcga cccttgcagc atgcacctcc 420 aatggctggg gcgtcactga ctgcaagcac acggaggatg tcggtgtggt gtgcagcgac 480 aaaaggattc ctgggttcaa atttgacaat tcgttgatca accagataga gaacctgaat 540 atccaggtgg aggacattcg gattcgagcc atcctctcaa cctaccgcaa gcgcacccca 600 gtgatggagg gctacgtgga ggtgaaggag ggcaagacct ggaagcagat ctgtgacaag 660 cactggacgg ccaagaattc ccgcgtggtc tgcggcatgt ttggcttccc tggggagagg 720 acatacaata ccaaagtgta caaaatgttt gcctcacgga ggaagcagcg ctactggcca 780 ttctccatgg actgcaccgg cacagaggcc cacatctcca gctgcaagct gggcccccag 840 gtgtcactgg accccatgaa gaatgtcacc tgcgagaatg ggctaccggc cgtggtgggt 900 tgtgtgcctg ggcaggtctt cagccctgac ggaccctcaa gattccggaa agcgtacaag 960 ccagagcaac ccctggtgcg actgagaggc ggtgcctaca tcggggaggg ccgcgtggag 1020 gtgctcaaaa atggagaatg ggggaccgtc tgcgacgaca agtgggacct ggtgtcggcc 1080 agtgtggtct gcagagagct gggctttggg agtgccaaag aggcagtcac tggctcccga 1140 ctggggcaag ggatcggacc catccacctc aacgagatcc agtgcacagg caatgagaag 1200 tccattatag actgcaagtt caatgccgag tctcagggct gcaaccacga ggaggatgct 1260 ggtgtgagat gcaacacccc tgccatgggc ttgcagaaga agctgcgcct gaacggcggc 1320 cgcaatccct acgagggccg agtggaggtg ctggtggaga gaaacgggtc ccttgtgtgg 1380 gggatggtgt gtggccaaaa ctggggcatc gtggaggcca tggtggtctg ccgccagctg 1440 ggcctgggat tcgccagcaa cgccttccag gagacctggt attggcacgg agatgtcaac 1500 agcaacaaag tggtcatgag tggagtgaag tgctcgggaa cggagctgtc cctggcgcac 1560 tgccgccacg acggggagga cgtggcctgc ccccagggcg gagtgcagta cggggccgga 1620 gttgcctgct cagaaaccgc ccctgacctg gtcctcaatg cggagatggt gcagcagacc 1680 acctacctgg aggaccggcc catgttcatg ctgcagtgtg ccatggagga gaactgcctc 1740 tcggcctcag ccgcgcagac cgaccccacc acgggctacc gccggctcct gcgcttctcc 1800 tcccagatcc acaacaatgg ccagtccgac ttccggccca agaacggccg ccacgcgtgg 1860 atctggcacg actgtcacag gcactaccac agcatggagg tgttcaccca ctatgacctg 1920 ctgaacctca atggcaccaa ggtggcagag ggccacaagg ccagcttctg cttggaggac 1980 acagaatgtg aaggagacat ccagaagaat tacgagtgtg ccaacttcgg cgatcagggc 2040 atcaccatgg gctgctggga catgtaccgc catgacatcg actgccagtg ggttgacatc 2100 actgacgtgc cccctggaga ctacctgttc caggttgtta ttaaccccaa cttcgaggtt 2160 gcagaatccg attactccaa caacatcatg aaatgcagga gccgctatga cggccaccgc 2220 atctggatgt acaactgcca cataggtggt tccttcagcg aagagacgga aaaaaagttt 2280 gagcacttca gcgggctctt aaacaaccag ctgtccccgc agtaa 2325 2 774 PRT Homo sapiens 2 Met Glu Arg Pro Leu Cys Ser His Leu Cys Ser Cys Leu Ala Met Leu 1 5 10 15 Ala Leu Leu Ser Pro Leu Ser Leu Ala Gln Tyr Asp Ser Trp Pro His 20 25 30 Tyr Pro Glu Tyr Phe Gln Gln Pro Ala Pro Glu Tyr His Gln Pro Gln 35 40 45 Ala Pro Ala Asn Val Ala Lys Ile Gln Leu Arg Leu Ala Gly Gln Lys 50 55 60 Arg Lys His Ser Glu Gly Arg Val Glu Val Tyr Tyr Asp Gly Gln Trp 65 70 75 80 Gly Thr Val Cys Asp Asp Asp Phe Ser Ile His Ala Ala His Val Val 85 90 95 Cys Arg Glu Leu Gly Tyr Val Glu Ala Lys Ser Trp Thr Ala Ser Ser 100 105 110 Ser Tyr Gly Lys Gly Glu Gly Pro Ile Trp Leu Asp Asn Leu His Cys 115 120 125 Thr Gly Asn Glu Ala Thr Leu Ala Ala Cys Thr Ser Asn Gly Trp Gly 130 135 140 Val Thr Asp Cys Lys His Thr Glu Asp Val Gly Val Val Cys Ser Asp 145 150 155 160 Lys Arg Ile Pro Gly Phe Lys Phe Asp Asn Ser Leu Ile Asn Gln Ile 165 170 175 Glu Asn Leu Asn Ile Gln Val Glu Asp Ile Arg Ile Arg Ala Ile Leu 180 185 190 Ser Thr Tyr Arg Lys Arg Thr Pro Val Met Glu Gly Tyr Val Glu Val 195 200 205 Lys Glu Gly Lys Thr Trp Lys Gln Ile Cys Asp Lys His Trp Thr Ala 210 215 220 Lys Asn Ser Arg Val Val Cys Gly Met Phe Gly Phe Pro Gly Glu Arg 225 230 235 240 Thr Tyr Asn Thr Lys Val Tyr Lys Met Phe Ala Ser Arg Arg Lys Gln 245 250 255 Arg Tyr Trp Pro Phe Ser Met Asp Cys Thr Gly Thr Glu Ala His Ile 260 265 270 Ser Ser Cys Lys Leu Gly Pro Gln Val Ser Leu Asp Pro Met Lys Asn 275 280 285 Val Thr Cys Glu Asn Gly Leu Pro Ala Val Val Gly Cys Val Pro Gly 290 295 300 Gln Val Phe Ser Pro Asp Gly Pro Ser Arg Phe Arg Lys Ala Tyr Lys 305 310 315 320 Pro Glu Gln Pro Leu Val Arg Leu Arg Gly Gly Ala Tyr Ile Gly Glu 325 330 335 Gly Arg Val Glu Val Leu Lys Asn Gly Glu Trp Gly Thr Val Cys Asp 340 345 350 Asp Lys Trp Asp Leu Val Ser Ala Ser Val Val Cys Arg Glu Leu Gly 355 360 365 Phe Gly Ser Ala Lys Glu Ala Val Thr Gly Ser Arg Leu Gly Gln Gly 370 375 380 Ile Gly Pro Ile His Leu Asn Glu Ile Gln Cys Thr Gly Asn Glu Lys 385 390 395 400 Ser Ile Ile Asp Cys Lys Phe Asn Ala Glu Ser Gln Gly Cys Asn His 405 410 415 Glu Glu Asp Ala Gly Val Arg Cys Asn Thr Pro Ala Met Gly Leu Gln 420 425 430 Lys Lys Leu Arg Leu Asn Gly Gly Arg Asn Pro Tyr Glu Gly Arg Val 435 440 445 Glu Val Leu Val Glu Arg Asn Gly Ser Leu Val Trp Gly Met Val Cys 450 455 460 Gly Gln Asn Trp Gly Ile Val Glu Ala Met Val Val Cys Arg Gln Leu 465 470 475 480 Gly Leu Gly Phe Ala Ser Asn Ala Phe Gln Glu Thr Trp Tyr Trp His 485 490 495 Gly Asp Val Asn Ser Asn Lys Val Val Met Ser Gly Val Lys Cys Ser 500 505 510 Gly Thr Glu Leu Ser Leu Ala His Cys Arg His Asp Gly Glu Asp Val 515 520 525 Ala Cys Pro Gln Gly Gly Val Gln Tyr Gly Ala Gly Val Ala Cys Ser 530 535 540 Glu Thr Ala Pro Asp Leu Val Leu Asn Ala Glu Met Val Gln Gln Thr 545 550 555 560 Thr Tyr Leu Glu Asp Arg Pro Met Phe Met Leu Gln Cys Ala Met Glu 565 570 575 Glu Asn Cys Leu Ser Ala Ser Ala Ala Gln Thr Asp Pro Thr Thr Gly 580 585 590 Tyr Arg Arg Leu Leu Arg Phe Ser Ser Gln Ile His Asn Asn Gly Gln 595 600 605 Ser Asp Phe Arg Pro Lys Asn Gly Arg His Ala Trp Ile Trp His Asp 610 615 620 Cys His Arg His Tyr His Ser Met Glu Val Phe Thr His Tyr Asp Leu 625 630 635 640 Leu Asn Leu Asn Gly Thr Lys Val Ala Glu Gly His Lys Ala Ser Phe 645 650 655 Cys Leu Glu Asp Thr Glu Cys Glu Gly Asp Ile Gln Lys Asn Tyr Glu 660 665 670 Cys Ala Asn Phe Gly Asp Gln Gly Ile Thr Met Gly Cys Trp Asp Met 675 680 685 Tyr Arg His Asp Ile Asp Cys Gln Trp Val Asp Ile Thr Asp Val Pro 690 695 700 Pro Gly Asp Tyr Leu Phe Gln Val Val Ile Asn Pro Asn Phe Glu Val 705 710 715 720 Ala Glu Ser Asp Tyr Ser Asn Asn Ile Met Lys Cys Arg Ser Arg Tyr 725 730 735 Asp Gly His Arg Ile Trp Met Tyr Asn Cys His Ile Gly Gly Ser Phe 740 745 750 Ser Glu Glu Thr Glu Lys Lys Phe Glu His Phe Ser Gly Leu Leu Asn 755 760 765 Asn Gln Leu Ser Pro Gln 770 3 757 PRT Homo sapiens 3 Met Met Trp Pro Gln Pro Pro Thr Phe Ser Leu Phe Leu Leu Leu Leu 1 5 10 15 Leu Ser Gln Ala Pro Ser Ser Arg Pro Gln Ser Ser Gly Thr Lys Lys 20 25 30 Leu Arg Leu Val Gly Pro Ala Asp Arg Pro Glu Glu Gly Arg Leu Glu 35 40 45 Val Leu His Gln Gly Gln Trp Gly Thr Val Cys Asp Asp Asp Phe Ala 50 55 60 Leu Gln Glu Ala Thr Val Ala Cys Arg Gln Leu Gly Phe Glu Ser Ala 65 70 75 80 Leu Thr Trp Ala His Ser Ala Lys Tyr Gly Gln Gly Glu Gly Pro Ile 85 90 95 Trp Leu Asp Asn Val Arg Cys Leu Gly Thr Glu Lys Thr Leu Asp Gln 100 105 110 Cys Gly Ser Asn Gly Trp Gly Ile Ser Asp Cys Arg His Ser Glu Asp 115 120 125 Val Gly Val Val Cys His Pro Arg Arg Gln His Gly Tyr His Ser Glu 130 135 140 Lys Val Ser Asn Ala Leu Gly Pro Gln Gly Arg Arg Leu Glu Glu Val 145 150 155 160 Arg Leu Lys Pro Ile Leu Ala Ser Ala Lys Arg His Ser Pro Val Thr 165 170 175 Glu Gly Ala Val Glu Val Arg Tyr Asp Gly His Trp Arg Gln Val Cys 180 185 190 Asp Gln Gly Trp Thr Met Asn Asn Ser Arg Val Val Cys Gly Met Leu 195 200 205 Gly Phe Pro Ser Gln Thr Ser Val Asn Ser His Tyr Tyr Arg Lys Val 210 215 220 Trp Asn Leu Lys Met Lys Asp Pro Lys Ser Arg Leu Asn Ser Leu Thr 225 230 235 240 Lys Lys Asn Ser Phe Trp Ile His Arg Val Asp Cys Phe Gly Thr Glu 245 250 255 Pro His Leu Ala Lys Cys Gln Val Gln Val Ala Pro Gly Arg Gly Lys 260 265 270 Leu Arg Pro Ala Cys Pro Gly Gly Met His Ala Val Val Ser Cys Val 275 280 285 Ala Gly Pro His Phe Arg Arg Gln Lys Pro Lys Pro Thr Arg Lys Glu 290 295 300 Ser His Ala Glu Glu Leu Lys Val Arg Leu Arg Ser Gly Ala Gln Val 305 310 315 320 Gly Glu Gly Arg Val Glu Val Leu Met Asn Arg Gln Trp Gly Thr Val 325 330 335 Cys Asp His Arg Trp Asn Leu Ile Ser Ala Ser Val Val Cys Arg Gln 340 345 350 Leu Gly Phe Gly Ser Ala Arg Glu Ala Leu Phe Gly Ala Gln Leu Gly 355 360 365 Gln Gly Leu Gly Pro Ile His Leu Ser Glu Val Arg Cys Arg Gly Tyr 370 375 380 Glu Arg Thr Leu Gly Asp Cys Leu Ala Leu Glu Gly Ser Gln Asn Gly 385 390 395 400 Cys Gln His Ala Asn Asp Ala Ala Val Arg Cys Asn Ile Pro Asp Met 405 410 415 Gly Phe Gln Asn Lys Val Arg Leu Ala Gly Gly Arg Asn Ser Glu Glu 420 425 430 Gly Val Val Glu Val Gln Val Glu Val Asn Gly Gly Pro Arg Trp Gly 435 440 445 Thr Val Cys Ser Asp His Trp Gly Leu Thr Glu Ala Met Val Thr Cys 450 455 460 Arg Gln Leu Gly Leu Gly Phe Ala Asn Phe Ala Leu Lys Asp Thr Trp 465 470 475 480 Tyr Trp Gln Gly Thr Pro Glu Ala Lys Glu Val Val Met Ser Gly Val 485 490 495 Arg Cys Ser Gly Thr Glu Met Ala Leu Gln Gln Cys Gln Arg His Gly 500 505 510 Pro Val His Cys Ser His Gly Pro Gly Arg Phe Ser Ala Gly Val Ala 515 520 525 Cys Met Asn Ser Ala Pro Asp Leu Val Met Asn Ala Gln Leu Val Gln 530 535 540 Glu Thr Ala Tyr Leu Glu Asp Arg Pro Leu Ser Met Leu Tyr Cys Ala 545 550 555 560 His Glu Glu Asn Cys Leu Ser Lys Ser Ala Asp His Met Asp Trp Pro 565 570 575 Tyr Gly Tyr Arg Arg Leu Leu Arg Phe Ser Ser Gln Ile Tyr Asn Leu 580 585 590 Gly Arg Ala Asp Phe Arg Pro Lys Ala Gly Arg His Ser Trp Ile Trp 595 600 605 His Gln Cys His Arg His Asn His Ser Ile Glu Val Phe Thr His Tyr 610 615 620 Asp Leu Leu Thr Leu Asn Gly Ser Lys Val Ala Glu Gly His Lys Ala 625 630 635 640 Ser Phe Cys Leu Glu Asp Thr Asn Cys Pro Ser Gly Val Gln Arg Arg 645 650 655 Tyr Ala Cys Ala Asn Phe Gly Glu Gln Gly Val Ala Val Gly Cys Trp 660 665 670 Asp Thr Tyr Arg His Asp Ile Asp Cys Gln Trp Val Asp Ile Thr Asp 675 680 685 Val Gly Pro Gly Asp Tyr Ile Phe Gln Val Val Val Asn Pro Thr Asn 690 695 700 Asp Val Ala Glu Ser Asp Phe Ser Asn Asn Met Ile Arg Cys Arg Cys 705 710 715 720 Lys Tyr Asp Gly Gln Arg Val Trp Leu His Asn Cys His Thr Gly Asp 725 730 735 Ser Tyr Arg Ala Asn Ala Glu Leu Ser Leu Glu Gln Glu Gln Arg Leu 740 745 750 Arg Asn Asn Leu Ile 755 4 2262 DNA Homo sapiens 4 atgcgacctg tcagtgtctg gcagtggagc ccctgggggc tgctgctgtg cctgctgtgc 60 agttcgtgct tggggtctcc gtccccttcc acgggccctg agaagaaggc cgggagccag 120 gggcttcggt tccggctggc tggcttcccc aggaagccct acgagggccg cgtggagata 180 cagcgagctg gtgaatgggg caccatctgc gatgatgact tcacgctgca ggctgcccac 240 atcctctgcc gggagctggg cttcacagag gccacaggct ggacccacag tgccaaatat 300 ggccctggaa caggccgcat ctggctggac aacttgagct gcagtgggac cgagcagagt 360 gtgactgaat gtgcctcccg gggctggggg aacagtgact gtacgcacga tgaggatgct 420 ggggtcatct gcaaagacca gcgcctccct ggcttctcgg actccaatgt cattgaggta 480 gagcatcacc tgcaagtgga ggaggtgcga attcgacccg ccgttgggtg gggcagacga 540 cccctgcccg tgacggaggg gctggtggaa gtcaggcttc ctgacggctg gtcgcaagtg 600 tgcgacaaag gctggagcgc ccacaacagc cacgtggtct gcgggatgct gggcttcccc 660 agcgaaaaga gggtcaacgc ggccttctac aggctgctag cccaacggca gcaacactcc 720 tttggtctgc atggggtggc gtgcgtgggc acggaggccc acctctccct ctgttccctg 780 gagttctatc gtgccaatga caccgccagg tgccctgggg ggggccctgc agtggtgagc 840 tgtgtgccag gccctgtcta cgcggcatcc agtggccaga agaagcaaca acagtcgaag 900 cctcaggggg aggcccgtgt ccgtctaaag ggcggcgccc accctggaga gggccgggta 960 gaagtcctga aggccagcac atggggcaca gtctgtgacc gcaagtggga cctgcatgca 1020 gccagcgtgg tgtgtcggga gctgggcttc gggagtgctc gagaagctct gagtggcgct 1080 cgcatggggc agggcatggg tgctatccac ctgagtgaag ttcgctgctc tggacaggag 1140 ctctccctct ggaagtgccc ccacaagaac atcacagctg aggattgttc acatagccag 1200 gatgccgggg tccggtgcaa cctaccttac actggggcag agaccaggat ccgactcagt 1260 gggggccgca gccaacatga ggggcgagtc gaggtgcaaa tagggggacc tgggcccctt 1320 cgctggggcc tcatctgtgg ggatgactgg gggaccctgg aggccatggt ggcctgtagg 1380 caactgggtc tgggctacgc caaccacggc ctgcaggaga cctggtactg ggactctggg 1440 aatataacag aggtggtgat gagtggagtg cgctgcacag ggactgagct gtccctggat 1500 cagtgtgccc atcatggcac ccacatcacc tgcaagagga cagggacccg cttcactgct 1560 ggagtcatct gttctgagac tgcatcagat ctgttgctgc actcagcact ggtgcaggag 1620 accgcctaca tcgaagaccg gcccctgcat atgttgtact gtgctgcgga agagaactgc 1680 ctggccagct cagcccgctc agccaactgg ccctatggtc accggcgtct gctccgattc 1740 tcctcccaga tccacaacct gggacgagct gacttcaggc ccaaggctgg gcgccactcc 1800 tgggtgtggc acgagtgcca tgggcattac cacagcatgg acatcttcac tcactatgat 1860 atcctcaccc caaatggcac caaggtggct gagggccaca aagctagttt ctgtctcgaa 1920 gacactgagt gtcaggagga tgtctccaag cggtatgagt gtgccaactt tggagagcaa 1980 ggcatcactg tgggttgctg ggatctctac cggcatgaca ttgactgtca gtggattgac 2040 atcacggatg tgaagccagg aaactacatt ctccaggttg tcatcaaccc aaactttgaa 2100 gtagcagaga gtgactttac caacaatgca atgaaatgta actgcaaata tgatggacat 2160 agaatctggg tgcacaactg ccacattggt gatgccttca gtgaagaggc caacaggagg 2220 tttgaacgct accctggcca gaccagcaac cagattatct aa 2262 5 1725 DNA Homo sapiens 5 atggctctgg cccgaggcag ccggcagctg ggggccctgg tgtggggcgc ctgcctgtgc 60 gtgctggtgc acgggcagca ggcgcagccc gggcagggct cggaccccgc ccgctggcgg 120 cagctgatcc agtgggagaa caacgggcag gtgtacagct tgctcaactc gggctcagag 180 tacgtgccgg ccggacctca gcgctccgag agtagctccc gggtgctgct ggccggcgcg 240 ccccaggccc agcagcggcg cagccacggg agcccccggc gtcggcaggc gccgtccctg 300 cccctgccgg ggcgcgtggg ctcggacacc gtgcgcggcc aggcgcggca cccattcggc 360 tttggccagg tgcccgacaa ctggcgcgag gtggccgtcg gggacagcac gggcatggcc 420 ctggcccgca cctccgtctc ccagcaacgg cacgggggct ccgcctcctc ggtctcggct 480 tcggccttcg ccagcaccta ccgccagcag ccctcctacc cgcagcagtt cccctacccg 540 caggcgccct tcgtcagcca gtacgagaac tacgaccccg cgtcgcggac ctacgaccag 600 ggtttcgtgt actaccggcc cgcgggcggc ggcgtgggcg cgggggcggc ggccgtggcc 660 tcggcggggg tcatctaccc ctaccagccc cgggcgcgct acgaggagta cggcggcggc 720 gaagagctgc ccgagtaccc gcctcagggc ttctacccgg cccccgagag gccctacgtg 780 ccgccgccgc cgccgccccc cgacggcctg gaccgccgct actcgcacag tctgtacagc 840 gagggcaccc ccggcttcga gcaggcctac cctgaccccg gtcccgaggc ggcgcaggcc 900 catggcggag acccacgcct gggctggtac ccgccctacg ccaacccgcc gcccgaggcg 960 tacgggccgc cgcgcgcgct ggagccgccc tacctgccgg tgcgcagctc cgacacgccc 1020 ccgccgggtg gggagcggaa cggcgcgcag cagggccgcc tcagcgtagg cagcgtgtac 1080 cggcccaacc agaacggccg cggtctccct gacttggtcc cagaccccaa ctatgtgcaa 1140 gcatccactt atgtgcagag agcccacctg tactccctgc gctgtgctgc ggaggagaag 1200 tgtctggcca gcacagccta tgcccctgag gccaccgact acgatgtgcg ggtgctactg 1260 cgcttccccc agcgcgtgaa gaaccagggc acagcagact tcctccccaa ccggccacgg 1320 cacacctggg agtggcacag ctgccaccag cattaccaca gcatggacga gttcagccac 1380 tacgacctac tggatgcagc cacaggcaag aaggtggccg agggccacaa ggccagtttc 1440 tgcctggagg acagcacctg tgacttcggc aacctcaagc gctatgcatg cacctctcat 1500 acccagggcc tgagcccagg ctgctatgac acctacaatg cggacatcga ctgccagtgg 1560 atcgacataa ccgacgtgca gcctgggaac tacatcctca aggtgcacgt gaacccaaag 1620 tatattgttt tggagtctga cttcaccaac aacgtggtga gatgcaacat tcactacaca 1680 ggtcgctacg tttctgcaac aaactgcaaa attgtccaat cctga 1725 6 574 PRT Homo sapiens 6 Met Ala Leu Ala Arg Gly Ser Arg Gln Leu Gly Ala Leu Val Trp Gly 1 5 10 15 Ala Cys Leu Cys Val Leu Val His Gly Gln Gln Ala Gln Pro Gly Gln 20 25 30 Gly Ser Asp Pro Ala Arg Trp Arg Gln Leu Ile Gln Trp Glu Asn Asn 35 40 45 Gly Gln Val Tyr Ser Leu Leu Asn Ser Gly Ser Glu Tyr Val Pro Ala 50 55 60 Gly Pro Gln Arg Ser Glu Ser Ser Ser Arg Val Leu Leu Ala Gly Ala 65 70 75 80 Pro Gln Ala Gln Gln Arg Arg Ser His Gly Ser Pro Arg Arg Arg Gln 85 90 95 Ala Pro Ser Leu Pro Leu Pro Gly Arg Val Gly Ser Asp Thr Val Arg 100 105 110 Gly Gln Ala Arg His Pro Phe Gly Phe Gly Gln Val Pro Asp Asn Trp 115 120 125 Arg Glu Val Ala Val Gly Asp Ser Thr Gly Met Ala Leu Ala Arg Thr 130 135 140 Ser Val Ser Gln Gln Arg His Gly Gly Ser Ala Ser Ser Val Ser Ala 145 150 155 160 Ser Ala Phe Ala Ser Thr Tyr Arg Gln Gln Pro Ser Tyr Pro Gln Gln 165 170 175 Phe Pro Tyr Pro Gln Ala Pro Phe Val Ser Gln Tyr Glu Asn Tyr Asp 180 185 190 Pro Ala Ser Arg Thr Tyr Asp Gln Gly Phe Val Tyr Tyr Arg Pro Ala 195 200 205 Gly Gly Gly Val Gly Ala Gly Ala Ala Ala Val Ala Ser Ala Gly Val 210 215 220 Ile Tyr Pro Tyr Gln Pro Arg Ala Arg Tyr Glu Glu Tyr Gly Gly Gly 225 230 235 240 Glu Glu Leu Pro Glu Tyr Pro Pro Gln Gly Phe Tyr Pro Ala Pro Glu 245 250 255 Arg Pro Tyr Val Pro Pro Pro Pro Pro Pro Pro Asp Gly Leu Asp Arg 260 265 270 Arg Tyr Ser His Ser Leu Tyr Ser Glu Gly Thr Pro Gly Phe Glu Gln 275 280 285 Ala Tyr Pro Asp Pro Gly Pro Glu Ala Ala Gln Ala His Gly Gly Asp 290 295 300 Pro Arg Leu Gly Trp Tyr Pro Pro Tyr Ala Asn Pro Pro Pro Glu Ala 305 310 315 320 Tyr Gly Pro Pro Arg Ala Leu Glu Pro Pro Tyr Leu Pro Val Arg Ser 325 330 335 Ser Asp Thr Pro Pro Pro Gly Gly Glu Arg Asn Gly Ala Gln Gln Gly 340 345 350 Arg Leu Ser Val Gly Ser Val Tyr Arg Pro Asn Gln Asn Gly Arg Gly 355 360 365 Leu Pro Asp Leu Val Pro Asp Pro Asn Tyr Val Gln Ala Ser Thr Tyr 370 375 380 Val Gln Arg Ala His Leu Tyr Ser Leu Arg Cys Ala Ala Glu Glu Lys 385 390 395 400 Cys Leu Ala Ser Thr Ala Tyr Ala Pro Glu Ala Thr Asp Tyr Asp Val 405 410 415 Arg Val Leu Leu Arg Phe Pro Gln Arg Val Lys Asn Gln Gly Thr Ala 420 425 430 Asp Phe Leu Pro Asn Arg Pro Arg His Thr Trp Glu Trp His Ser Cys 435 440 445 His Gln His Tyr His Ser Met Asp Glu Phe Ser His Tyr Asp Leu Leu 450 455 460 Asp Ala Ala Thr Gly Lys Lys Val Ala Glu Gly His Lys Ala Ser Phe 465 470 475 480 Cys Leu Glu Asp Ser Thr Cys Asp Phe Gly Asn Leu Lys Arg Tyr Ala 485 490 495 Cys Thr Ser His Thr Gln Gly Leu Ser Pro Gly Cys Tyr Asp Thr Tyr 500 505 510 Asn Ala Asp Ile Asp Cys Gln Trp Ile Asp Ile Thr Asp Val Gln Pro 515 520 525 Gly Asn Tyr Ile Leu Lys Val His Val Asn Pro Lys Tyr Ile Val Leu 530 535 540 Glu Ser Asp Phe Thr Asn Asn Val Val Arg Cys Asn Ile His Tyr Thr 545 550 555 560 Gly Arg Tyr Val Ser Ala Thr Asn Cys Lys Ile Val Gln Ser 565 570 7 1254 DNA Homo sapiens 7 atgcgcttcg cctggaccgt gctcctgctc gggcctttgc agctctgcgc gctagtgcac 60 tgcgcccctc ccgccgccgg ccaacagcag cccccgcgcg agccgccggc ggctccgggc 120 gcctggcgcc agcagatcca atgggagaac aacgggcagg tgttcagctt gctgagcctg 180 ggctcacagt accagcctca gcgccgccgg gacccgggcg ccgccgtccc tggtgcagcc 240 aacgcctccg cccagcagcc ccgcactccg atcctgctga tccgcgacaa ccgcaccgcc 300 gcggcgcgaa cgcggacggc cggctcatct ggagtcaccg ctggccgccc caggcccacc 360 gcccgtcact ggttccaagc tggctactcg acatctagag cccgcgaagc tggcgcctcg 420 cgcgcggaga accagacagc gccgggagaa gttcctgcgc tcagtaacct gcggccgccc 480 agccgcgtgg acggcatggt gggcgacgac ccttacaacc cctacaagta ctctgacgac 540 aacccttatt acaactacta cgatacttat gaaaggccca gacctggggg caggtaccgg 600 cccggatacg gcactggcta cttccagtac ggtctcccag acctggtggc cgacccctac 660 tacatccagg cgtccacgta cgtgcagaag atgtccatgt acaacctgag atgcgcggcg 720 gaggaaaact gtctggccag tacagcatac agggcagatg tcagagatta tgatcacagg 780 gtgctgctca gatttcccca aagagtgaaa aaccaaggga catcagattt cttacccagc 840 cgaccaagat attcctggga atggcacagt tgtcatcaac attaccacag tatggatgag 900 tttagccact atgacctgct tgatgccaac acccagagga gagtggctga aggccacaaa 960 gcaagtttct gtcttgaaga cacatcctgt gactatggct accacaggcg atttgcatgt 1020 actgcacaca cacagggatt gagtcctggc tgttatgata cctatggtgc agacatagac 1080 tgccagtgga ttgatattac agatgtaaaa cctggaaact atatcctaaa ggtcagtgta 1140 aaccccagct acctggttcc tgaatctgac tataccaaca atgttgtgcg ctgtgacatt 1200 cgctacacag gacatcatgc gtatgcctca ggctgcacaa tttcaccgta ttag 1254 8 417 PRT Homo sapiens 8 Met Arg Phe Ala Trp Thr Val Leu Leu Leu Gly Pro Leu Gln Leu Cys 1 5 10 15 Ala Leu Val His Cys Ala Pro Pro Ala Ala Gly Gln Gln Gln Pro Pro 20 25 30 Arg Glu Pro Pro Ala Ala Pro Gly Ala Trp Arg Gln Gln Ile Gln Trp 35 40 45 Glu Asn Asn Gly Gln Val Phe Ser Leu Leu Ser Leu Gly Ser Gln Tyr 50 55 60 Gln Pro Gln Arg Arg Arg Asp Pro Gly Ala Ala Val Pro Gly Ala Ala 65 70 75 80 Asn Ala Ser Ala Gln Gln Pro Arg Thr Pro Ile Leu Leu Ile Arg Asp 85 90 95 Asn Arg Thr Ala Ala Ala Arg Thr Arg Thr Ala Gly Ser Ser Gly Val 100 105 110 Thr Ala Gly Arg Pro Arg Pro Thr Ala Arg His Trp Phe Gln Ala Gly 115 120 125 Tyr Ser Thr Ser Arg Ala Arg Glu Ala Gly Ala Ser Arg Ala Glu Asn 130 135 140 Gln Thr Ala Pro Gly Glu Val Pro Ala Leu Ser Asn Leu Arg Pro Pro 145 150 155 160 Ser Arg Val Asp Gly Met Val Gly Asp Asp Pro Tyr Asn Pro Tyr Lys 165 170 175 Tyr Ser Asp Asp Asn Pro Tyr Tyr Asn Tyr Tyr Asp Thr Tyr Glu Arg 180 185 190 Pro Arg Pro Gly Gly Arg Tyr Arg Pro Gly Tyr Gly Thr Gly Tyr Phe 195 200 205 Gln Tyr Gly Leu Pro Asp Leu Val Ala Asp Pro Tyr Tyr Ile Gln Ala 210 215 220 Ser Thr Tyr Val Gln Lys Met Ser Met Tyr Asn Leu Arg Cys Ala Ala 225 230 235 240 Glu Glu Asn Cys Leu Ala Ser Thr Ala Tyr Arg Ala Asp Val Arg Asp 245 250 255 Tyr Asp His Arg Val Leu Leu Arg Phe Pro Gln Arg Val Lys Asn Gln 260 265 270 Gly Thr Ser Asp Phe Leu Pro Ser Arg Pro Arg Tyr Ser Trp Glu Trp 275 280 285 His Ser Cys His Gln His Tyr His Ser Met Asp Glu Phe Ser His Tyr 290 295 300 Asp Leu Leu Asp Ala Asn Thr Gln Arg Arg Val Ala Glu Gly His Lys 305 310 315 320 Ala Ser Phe Cys Leu Glu Asp Thr Ser Cys Asp Tyr Gly Tyr His Arg 325 330 335 Arg Phe Ala Cys Thr Ala His Thr Gln Gly Leu Ser Pro Gly Cys Tyr 340 345 350 Asp Thr Tyr Gly Ala Asp Ile Asp Cys Gln Trp Ile Asp Ile Thr Asp 355 360 365 Val Lys Pro Gly Asn Tyr Ile Leu Lys Val Ser Val Asn Pro Ser Tyr 370 375 380 Leu Val Pro Glu Ser Asp Tyr Thr Asn Asn Val Val Arg Cys Asp Ile 385 390 395 400 Arg Tyr Thr Gly His His Ala Tyr Ala Ser Gly Cys Thr Ile Ser Pro 405 410 415 Tyr 9 752 PRT Homo sapiens 9 Met Arg Pro Val Ser Val Trp Gln Trp Ser Pro Trp Gly Leu Leu Leu 1 5 10 15 Cys Leu Leu Cys Ser Ser Cys Leu Gly Ser Pro Ser Pro Ser Thr Gly 20 25 30 Pro Glu Lys Lys Ala Gly Ser Gln Gly Leu Arg Phe Arg Leu Ala Gly 35 40 45 Phe Pro Arg Lys Pro Tyr Glu Gly Arg Val Glu Ile Gln Arg Ala Gly 50 55 60 Glu Trp Gly Thr Ile Cys Asp Asp Asp Phe Thr Leu Gln Ala Ala His 65 70 75 80 Ile Leu Cys Arg Glu Leu Gly Phe Thr Glu Ala Thr Gly Trp Thr His 85 90 95 Ser Ala Lys Tyr Gly Pro Gly Thr Gly Arg Ile Trp Leu Asp Asn Leu 100 105 110 Ser Cys Ser Gly Thr Glu Gln Ser Val Thr Glu Cys Ala Ser Arg Gly 115 120 125 Trp Gly Asn Ser Asp Cys Thr His Asp Glu Asp Ala Gly Val Ile Cys 130 135 140 Lys Asp Gln Arg Leu Pro Gly Phe Ser Asp Ser Asn Val Ile Glu Val 145 150 155 160 Glu His His Leu Gln Val Glu Glu Val Arg Ile Arg Pro Ala Val Gly 165 170 175 Trp Gly Arg Arg Pro Leu Pro Val Thr Glu Gly Leu Val Glu Val Arg 180 185 190 Leu Pro Asp Gly Trp Ser Gln Val Cys Asp Lys Gly Trp Ser Ala His 195 200 205 Asn Ser His Val Val Cys Gly Met Leu Gly Phe Pro Ser Glu Lys Arg 210 215 220 Val Asn Ala Ala Phe Tyr Arg Leu Leu Ala Gln Arg Gln Gln His Ser 225 230 235 240 Phe Gly Leu His Gly Val Ala Cys Val Gly Thr Glu Ala His Leu Ser 245 250 255 Leu Cys Ser Leu Glu Phe Tyr Arg Ala Asn Asp Thr Ala Arg Cys Pro 260 265 270 Gly Gly Gly Pro Ala Val Val Ser Cys Val Pro Gly Pro Val Tyr Ala 275 280 285 Ala Ser Ser Gly Gln Lys Lys Gln Gln Gln Ser Lys Pro Gln Gly Glu 290 295 300 Ala Arg Val Arg Leu Lys Gly Gly Ala His Pro Gly Glu Gly Arg Val 305 310 315 320 Glu Val Leu Lys Ala Ser Thr Trp Gly Thr Val Cys Asp Arg Lys Trp 325 330 335 Asp Leu His Ala Ala Ser Val Val Cys Arg Glu Leu Gly Phe Gly Ser 340 345 350 Ala Arg Glu Ala Leu Ser Gly Ala Arg Met Gly Gln Gly Met Gly Ala 355 360 365 Ile His Leu Ser Glu Val Arg Cys Ser Gly Gln Glu Leu Ser Leu Trp 370 375 380 Lys Cys Pro His Lys Asn Ile Thr Ala Glu Asp Cys Ser His Ser Gln 385 390 395 400 Asp Ala Gly Val Arg Cys Asn Leu Pro Tyr Thr Gly Ala Glu Thr Arg 405 410 415 Ile Arg Leu Ser Gly Gly Arg Ser Gln His Glu Gly Arg Val Glu Val 420 425 430 Gln Ile Gly Gly Pro Gly Pro Leu Arg Trp Gly Leu Ile Cys Gly Asp 435 440 445 Asp Trp Gly Thr Leu Glu Ala Met Val Ala Cys Arg Gln Leu Gly Leu 450 455 460 Gly Tyr Ala Asn His Gly Leu Gln Glu Thr Trp Tyr Trp Asp Ser Gly 465 470 475 480 Asn Ile Thr Glu Val Val Met Ser Gly Val Arg Cys Thr Gly Thr Glu 485 490 495 Leu Ser Leu Asp Gln Cys Ala His His Gly Thr His Ile Thr Cys Lys 500 505 510 Arg Thr Gly Thr Arg Phe Thr Ala Gly Val Ile Cys Ser Glu Thr Ala 515 520 525 Ser Asp Leu Leu Leu His Ser Ala Leu Val Gln Glu Thr Ala Tyr Ile 530 535 540 Glu Asp Arg Pro Leu His Met Leu Tyr Cys Ala Ala Glu Glu Asn Cys 545 550 555 560 Leu Ala Ser Ser Ala Arg Ser Ala Asn Trp Pro Tyr Gly His Arg Arg 565 570 575 Leu Leu Arg Phe Ser Ser Gln Ile His Asn Leu Gly Arg Ala Asp Phe 580 585 590 Arg Pro Lys Ala Gly Arg His Ser Trp Val Trp His Glu Cys His Gly 595 600 605 His Tyr His Ser Met Asp Ile Phe Thr His Tyr Asp Ile Leu Thr Pro 610 615 620 Asn Gly Thr Lys Val Ala Glu Gly His Lys Ala Ser Phe Cys Leu Glu 625 630 635 640 Asp Thr Glu Cys Gln Glu Asp Val Ser Lys Arg Tyr Glu Cys Ala Asn 645 650 655 Phe Gly Glu Gln Gly Ile Thr Val Gly Cys Trp Asp Leu Tyr Arg His 660 665 670 Asp Ile Asp Cys Gln Trp Ile Asp Ile Thr Asp Val Lys Pro Gly Asn 675 680 685 Tyr Ile Leu Gln Val Val Ile Asn Pro Asn Phe Glu Val Ala Glu Ser 690 695 700 Asp Phe Thr Asn Asn Ala Met Lys Cys Asn Cys Lys Tyr Asp Gly His 705 710 715 720 Arg Ile Trp Val His Asn Cys His Ile Gly Asp Ala Phe Ser Glu Glu 725 730 735 Ala Asn Arg Arg Phe Glu Arg Tyr Pro Gly Gln Thr Ser Asn Gln Ile 740 745 750 10 36 DNA Artificial sequence Single strand DNA oligonucleotide 10 cgcaagcttg gatccgggat ggagaggcct ctgtgc 36 11 37 DNA Artificial sequence Single strand DNA oligonucleotide 11 cgctctagag gatccttact gcggggacag ctggttg 37 12 21 DNA Artificial sequence Single strand DNA oligonucleotide 12 gccatgcgac ctgtcagtgt c 21 13 18 DNA Artificial sequence Single strand DNA oligonucleotide 13 gggcagtggc acttagat 18 14 613 DNA Homo sapiens 14 ccctgacctg gtcctcaatg cggagatggt gcagcagacc acctacctgg aggaccggcc 60 catgttcatg ctgcagtgtg ccatggagga gaactgcctc tcggcctcag ccgcgcagac 120 cgaccccacc acgggctacc gccggctcct gcgcttctcc tcccagatcc acaacaatgg 180 ccagtccgac ttccggccca agaacggccg ccacgcgtgg atctggcacg actgtcacag 240 gcactaccac agcatggagg tgttcaccca ctatgacctg ctgaacctca atggcaccaa 300 ggtggcagag ggccacaagg ccagcttctg cttggaggac acagaatgtg aaggagacat 360 ccagaagaat tacgagtgtg ccaacttcgg cgatcagggc atcaccatgg gctgctggga 420 catgtaccgc catgacatcg actgccagtg ggttgacatc actgacgtgc cccctggaga 480 ctacctgttc caggttgtta ttaaccccaa cttcgaggtt gcagaatccg attactccaa 540 caacatcatg aaatgcagga gccgctatga cggccaccgc atctggatgt acaactgcca 600 cataggtggt tcc 613 15 30 DNA Artificial sequence Single strand DNA oligonucleotide 15 acatgcatgc cctgacctgg tcctcaatgc 30 16 30 DNA Artificial sequence Single strand DNA oligonucleotide 16 cccaagcttg gaaccaccta tgtggcagtt 30 17 210 PRT Artificial sequence Protein sequence derived from fusion of LOR-1 fragment (1641-2253) to a 5′ 6xHis tag 17 His His His His His His Pro Asp Leu Val Leu Asn Ala Glu Met Val 1 5 10 15 Gln Gln Thr Thr Tyr Leu Glu Asp Arg Pro Met Phe Met Leu Gln Cys 20 25 30 Ala Met Glu Glu Asn Cys Leu Ser Ala Ser Ala Ala Gln Thr Asp Pro 35 40 45 Thr Thr Gly Tyr Arg Arg Leu Leu Arg Phe Ser Ser Gln Ile His Asn 50 55 60 Asn Gly Gln Ser Asp Phe Arg Pro Lys Asn Gly Arg His Ala Trp Ile 65 70 75 80 Trp His Asp Cys His Arg His Tyr His Ser Met Glu Val Phe Thr His 85 90 95 Tyr Asp Leu Leu Asn Leu Asn Gly Thr Lys Val Ala Glu Gly His Lys 100 105 110 Ala Ser Phe Cys Leu Glu Asp Thr Glu Cys Glu Gly Asp Ile Gln Lys 115 120 125 Asn Tyr Glu Cys Ala Asn Phe Gly Asp Gln Gly Ile Thr Met Gly Cys 130 135 140 Trp Asp Met Tyr Arg His Asp Ile Asp Cys Gln Trp Val Asp Ile Thr 145 150 155 160 Asp Val Pro Pro Gly Asp Tyr Leu Phe Gln Val Val Ile Asn Pro Asn 165 170 175 Phe Glu Val Ala Glu Ser Asp Tyr Ser Asn Asn Ile Met Lys Cys Arg 180 185 190 Ser Arg Tyr Asp Gly His Arg Ile Trp Met Tyr Asn Cys His Ile Gly 195 200 205 Gly Ser 210 18 660 DNA Artificial sequence Northern blot probe consisted of nucleotides 1-660 of the LOR-1 cDNA 18 atggagaggc ctctgtgctc ccacctctgc agctgcctgg ctatgctggc cctcctgtcc 60 cccctgagcc tggcacagta tgacagctgg ccccattacc ccgagtactt ccagcaaccg 120 gctcctgagt atcaccagcc ccaggccccc gccaacgtgg ccaagattca gctgcgcctg 180 gctgggcaga agaggaagca cagcgagggc cgggtggagg tgtactatga tggccagtgg 240 ggcaccgtgt gcgatgacga cttctccatc cacgctgccc acgtcgtctg ccgggagctg 300 ggctatgtgg aggccaagtc ctggactgcc agctcctcct acggcaaggg agaagggccc 360 atctggttag acaatctcca ctgtactggc aacgaggcga cccttgcagc atgcacctcc 420 aatggctggg gcgtcactga ctgcaagcac acggaggatg tcggtgtggt gtgcagcgac 480 aaaaggattc ctgggttcaa atttgacaat tcgttgatca accagataga gaacctgaat 540 atccaggtgg aggacattcg gattcgagcc atcctctcaa cctaccgcaa gcgcacccca 600 gtgatggagg gctacgtgga ggtgaaggag ggcaagacct ggaagcagat ctgtgacaag 660 19 530 DNA Artificial sequence Northern blot probe consisted of nucleotides 1061-1590 of the LOR-2 cDNA 19 gagaagctct gagtggcgct cgcatggggc agggcatggg tgctatccac ctgagtgaag 60 ttcgctgctc tggacaggag ctctccctct ggaagtgccc ccacaagaac atcacagctg 120 aggattgttc acatagccag gatgccgggg tccggtgcaa cctaccttac actggggcag 180 agaccaggat ccgactcagt gggggccgca gccaacatga ggggcgagtc gaggtgcaaa 240 tagggggacc tgggcccctt cgctggggcc tcatctgtgg ggatgactgg gggaccctgg 300 aggccatggt ggcctgtagg caactgggtc tgggctacgc caaccacggc ctgcaggaga 360 cctggtactg ggactctggg aatataacag aggtggtgat gagtggagtg cgctgcacag 420 ggactgagct gtccctggat cagtgtgccc atcatggcac ccacatcacc tgcaagagga 480 cagggacccg cttcactgct ggagtcatct gttctgagac tgcatcagat 530

Claims (35)

What is claimed is:
1. A method of modulating angiogenesis in a mammalian tissue, the method comprising administering into the mammalian tissue a molecule capable of modifying a tissue level and/or activity of at least one type of lysyl-oxidase to thereby modulate angiogenesis in the mammalian tissue.
2. The method of claim 1, wherein said molecule is an antibody or an antibody fragment capable of binding with, and at least partially inhibiting said activity of, said at least one polypeptide.
3. The method of claim 2, wherein said antibody or said antibody fragment is directed against at least an antigenic portion of the polypeptide set forth in SEQ ID NO: 2, 3, 6, 8 or 9.
4. The method of claim 1, wherein said molecule is a polynucleotide capable of downregulating expression of said at least one type of lysyl-oxidase.
5. The method of claim 4, wherein said polynucleotide is at least partially complementary with the polynucleotide set forth in SEQ ID NO: 1, 4, 5 or 7.
6. The method of claim 1, wherein said molecule is a polypeptide having lysyl-oxidase activity.
7. The method of claim 6, wherein said polypeptide is as set forth in SEQ ID NO: 2, 3, 6, 8 or 9.
8. A method of modulating angiogenesis in a mammalian tissue, the method comprising administering into the mammalian tissue a nucleic acid construct being capable of expressing a polypeptide having lysyl-oxidase activity to thereby modulate angiogenesis within the mammalian tissue.
9. The method of claim 8, wherein said polypeptide is at least 75% homologous to the polypeptide set forth in SEQ ID NO: 2, 3, 6, 8 or 9.
10. A method of identifying molecules capable of modulating angiogenesis, the method comprising:
(a) isolating molecules which exhibit specific reactivity with at least one type of lysyl-oxidase; and
(b) testing said molecules within mammalian tissue so as to determine the angiogenesis modulation activity thereof.
11. The method of claim 10, wherein step (a) is effected by binding assays and/or lysyl-oxidase activity assays.
12. A pharmaceutical composition useful for modulating angiogenesis in mammalian tissue, the composition-of matter comprising, as an active ingredient, a molecule capable of modifying a level and/or activity of at least one type of lysyl-oxidase of the mammalian tissue and a pharmaceutically effective carrier.
13. The pharmaceutical composition of claim 12, wherein said molecule is an antibody or an antibody fragment capable of binding with, and at least partially inhibiting said activity of, said at least one type of lysyl-oxidase.
14. The pharmaceutical composition of claim 13, wherein said antibody or said antibody fragment is directed against at least a portion of the polypeptide set forth in SEQ ID NO: 2, 3, 6, 8 or 9.
15. The pharmaceutical composition of claim 12, wherein said molecule is a polynucleotide capable of downregulating expression of said at least one type of lysyl-oxidase.
16. The pharmaceutical composition of claim 15, wherein said polynucleotide is at least partially complementary with the polynucleotide set forth in SEQ ID NO: 1, 4, 5 or 7.
17. The pharmaceutical composition of claim 12, wherein said molecule is a polypeptide having lysyl-oxidase activity.
18. The pharmaceutical composition of claim 12, wherein said polypeptide is as set forth SEQ ID NO: 2, 3, 6, 8 or 9.
19. A method of modulating angiogenesis in a mammalian tissue, the method comprising administering into the mammalian tissue a molecule capable of modifying a tissue level and/or a lysyl-oxidase activity of a polypeptide at least 75% homologous to the polypeptide set forth in SEQ ID NO: 2 to thereby modulate angiogenesis in the mammalian tissue.
20. A method of modulating angiogenesis in a mammalian tissue, the method comprising administering into the mammalian tissue a nucleic acid construct being capable of expressing a polypeptide at least 75% homologous to the polypeptide set forth in SEQ ID NO: 2 to thereby modulate angiogenesis within the mammalian tissue.
21. A method of determining the malignancy of cancerous tissue, the method comprising
(a) determining a lysyl-oxidase expression level and/or activity of the cancerous tissue; and
(b) comparing said lysyl-oxidase expression level and/or activity with that determined for control tissue to thereby determine the malignancy of the cancerous tissue.
22. A method of inhibiting metastasis and/or fibrosis in a mammalian tissue, the method comprising administering to the mammalian tissue a molecule capable of downregulating a tissue level and/or activity of at least one type of lysyl-oxidase to thereby inhibit metastasis in the mammalian tissue.
23. The method of claim 22, wherein said molecule is an antibody or an antibody fragment capable of specifically binding with said at least one type of lysyl-oxidase.
24. The method of claim 22, wherein said antibody or said antibody fragment is directed against at least an antigenic portion of the polypeptide set forth in SEQ ID NO: 2, 3, 6, 8 or 9.
25. The method of claim 22, wherein said molecule is a polynucleotide capable of downregulating expression of said at least one type of lysyl-oxidase.
26. The method of claim 25, wherein said polynucleotide is at least partially complementary with the polynucleotide set forth in SEQ ID NO: 1, 4, 5 or 7.
27. A method of identifying molecules capable of inhibiting metastasis and/or fibrosis, the method comprising:
(a) screening and identifying molecules which exhibit specific reactivity with at least one type of lysyl-oxidase; and
(b) testing a metastasis and/or fibrosis inhibitory potential of said molecules.
28. The method of claim 27, wherein step (a) is effected by binding assays and/or lysyl-oxidase activity assays.
29. The method of claim 27, wherein step (b) is effected in-vivo or in-vitro.
30. A pharmaceutical composition useful for inhibiting metastasis and/or fibrosis in mammalian tissue, comprising, as an active ingredient, a molecule capable of downregulating a level and/or activity of at least one type of lysyl-oxidase present within the mammalian tissue and a pharmaceutically effective carrier.
31. The pharmaceutical composition of claim 30, wherein said molecule is an antibody or an antibody fragment capable of specifically binding with said at least one type of lysyl-oxidase.
32. The pharmaceutical composition of claim 31, wherein said antibody or said antibody fragment is directed against at least an antigenic portion of the polypeptide set forth in SEQ ID NO: 2, 3, 6, 8 or 9.
33. The pharmaceutical composition of claim 30, wherein said molecule is a polynucleotide capable of downregulating expression of said at least one type of lysyl-oxidase.
34. The pharmaceutical composition of claim 33, wherein said polynucleotide is at least partially complementary with the polynucleotide set forth in SEQ ID NO: 1, 4, 5 or 7.
35. A method of inhibiting metastasis and fibrosis in a mammalian tissue, the method comprising administering to the mammalian tissue a molecule capable of downregulating a tissue level and/or a lysyl-oxidase activity of a polypeptide at least 75% homologous to the polypeptide set forth in ID NO: 2 or 9, to thereby inhibit metastasis and fibrosis in the mammalian tissue.
US10/305,348 2000-08-08 2002-11-27 Pharmaceutical compositions and methods useful for modulating angiogenesis and inhibiting metastasis and tumor fibrosis Abandoned US20030114410A1 (en)

Priority Applications (17)

Application Number Priority Date Filing Date Title
US10/305,348 US20030114410A1 (en) 2000-08-08 2002-11-27 Pharmaceutical compositions and methods useful for modulating angiogenesis and inhibiting metastasis and tumor fibrosis
PT08020753T PT2062919E (en) 2002-11-27 2003-11-27 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
ES08020753.3T ES2584847T3 (en) 2002-11-27 2003-11-27 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and evaluating the malignancy of colon cancer tumors
EP08020752A EP2062918A3 (en) 2002-11-27 2003-11-27 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
EP08020753.3A EP2062919B1 (en) 2002-11-27 2003-11-27 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
EP03777136A EP1572100A4 (en) 2002-11-27 2003-11-27 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
US10/536,440 US8163494B2 (en) 2002-11-27 2003-11-27 Method for assessing metastatic properties of breast cancer
PCT/IL2003/001008 WO2004047720A2 (en) 2002-11-27 2003-11-27 Methods of inhibiting angiogensis via modification of a lysyl oxidase
AU2003286391A AU2003286391A1 (en) 2002-11-27 2003-11-27 Methods of inhibiting angiogensis via modification of a lysyl oxidase
DK08020753.3T DK2062919T3 (en) 2002-11-27 2003-11-27 Pharmaceutical compositions and methods useful for the modulation of angiogenesis, inhibition of metastasis and tumor fibrosis, and evaluate malignancy of coloncancertumorer
US12/571,167 US8168180B2 (en) 2002-11-27 2009-09-30 Methods and compositions for modulating angiogenesis
HK09110803.7A HK1131165A1 (en) 2002-11-27 2009-11-19 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
US13/412,544 US20120202206A1 (en) 2002-11-27 2012-03-05 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
US13/416,976 US8815823B2 (en) 2002-11-27 2012-03-09 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
US14/311,110 US20140302499A1 (en) 2002-11-27 2014-06-20 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
CY20161100760T CY1117943T1 (en) 2002-11-27 2016-08-03 PHARMACEUTICAL COMPOSITIONS AND METHODS USED FOR CONSTRUCTION OF VASCULATION, SUSPENSION OF TRANSFER AND INFECTION OF NEoplasm,
US15/881,339 US20190040470A1 (en) 2002-11-27 2018-01-26 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US22373900P 2000-08-08 2000-08-08
PCT/IL2001/000728 WO2002011667A2 (en) 2000-08-08 2001-08-07 Pharmaceutical compositions and methods useful for modulating angiogenesis
US10/305,348 US20030114410A1 (en) 2000-08-08 2002-11-27 Pharmaceutical compositions and methods useful for modulating angiogenesis and inhibiting metastasis and tumor fibrosis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2001/000728 Continuation-In-Part WO2002011667A2 (en) 2000-08-08 2001-08-07 Pharmaceutical compositions and methods useful for modulating angiogenesis

Related Child Applications (3)

Application Number Title Priority Date Filing Date
PCT/IL2003/001008 Continuation-In-Part WO2004047720A2 (en) 2002-11-27 2003-11-27 Methods of inhibiting angiogensis via modification of a lysyl oxidase
US10536440 Continuation-In-Part 2003-11-27
US10/536,440 Continuation-In-Part US8163494B2 (en) 2002-11-27 2003-11-27 Method for assessing metastatic properties of breast cancer

Publications (1)

Publication Number Publication Date
US20030114410A1 true US20030114410A1 (en) 2003-06-19

Family

ID=32392444

Family Applications (7)

Application Number Title Priority Date Filing Date
US10/305,348 Abandoned US20030114410A1 (en) 2000-08-08 2002-11-27 Pharmaceutical compositions and methods useful for modulating angiogenesis and inhibiting metastasis and tumor fibrosis
US10/536,440 Expired - Fee Related US8163494B2 (en) 2002-11-27 2003-11-27 Method for assessing metastatic properties of breast cancer
US12/571,167 Expired - Fee Related US8168180B2 (en) 2002-11-27 2009-09-30 Methods and compositions for modulating angiogenesis
US13/412,544 Abandoned US20120202206A1 (en) 2002-11-27 2012-03-05 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
US13/416,976 Expired - Fee Related US8815823B2 (en) 2002-11-27 2012-03-09 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
US14/311,110 Abandoned US20140302499A1 (en) 2002-11-27 2014-06-20 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
US15/881,339 Abandoned US20190040470A1 (en) 2002-11-27 2018-01-26 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors

Family Applications After (6)

Application Number Title Priority Date Filing Date
US10/536,440 Expired - Fee Related US8163494B2 (en) 2002-11-27 2003-11-27 Method for assessing metastatic properties of breast cancer
US12/571,167 Expired - Fee Related US8168180B2 (en) 2002-11-27 2009-09-30 Methods and compositions for modulating angiogenesis
US13/412,544 Abandoned US20120202206A1 (en) 2002-11-27 2012-03-05 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
US13/416,976 Expired - Fee Related US8815823B2 (en) 2002-11-27 2012-03-09 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
US14/311,110 Abandoned US20140302499A1 (en) 2002-11-27 2014-06-20 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
US15/881,339 Abandoned US20190040470A1 (en) 2002-11-27 2018-01-26 Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors

Country Status (9)

Country Link
US (7) US20030114410A1 (en)
EP (3) EP2062919B1 (en)
AU (1) AU2003286391A1 (en)
CY (1) CY1117943T1 (en)
DK (1) DK2062919T3 (en)
ES (1) ES2584847T3 (en)
HK (1) HK1131165A1 (en)
PT (1) PT2062919E (en)
WO (1) WO2004047720A2 (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030232400A1 (en) * 2002-12-20 2003-12-18 Susan Radka Methods of screening subjects for expression of soluble receptors of vascular endothelial growth factor (VEGF) for use in managing treatment and determining prognostic outcome
US20040014113A1 (en) * 2002-05-31 2004-01-22 The Regents Of The University Of California Method for efficient RNA interference in mammalian cells
US20050164968A1 (en) * 2001-05-18 2005-07-28 Sirna Therapeutics, Inc. RNA interference mediated inhibition of ADAM33 gene expression using short interfering nucleic acid (siNA)
US20060127402A1 (en) * 2002-11-27 2006-06-15 Technion Research & Development Foundation Ltd. Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
WO2006128740A2 (en) * 2005-06-02 2006-12-07 Centelion Anti-vascular methods and therapies employing lysyl oxidase inhibitors
US20070021365A1 (en) * 2005-06-21 2007-01-25 The Board Of Trustees Of The Leland Stanford Junior University Inhibition of Lysyl oxidase for treating tumor growth and diagnostics relating thereto
US20070225242A1 (en) * 2005-06-21 2007-09-27 The Board Of Trustees Of The Leland Stanford Junior University Method and composition for treating and preventing tumor metastasis in vivo
US20090053224A1 (en) * 2007-08-02 2009-02-26 Arresto Biosciences Lox and loxl2 inhibitors and uses thereof
US20100203062A1 (en) * 2009-02-06 2010-08-12 Ingeborg Stalmans Methods and Compositions for Treatment of Neovascularization
US20100209415A1 (en) * 2009-01-06 2010-08-19 Victoria Smith Chemotherapeutic methods and compositions
US20100317721A1 (en) * 2007-07-15 2010-12-16 Technion Research & Development Foundation Ltd. Compositions and Methods for Treating Tumors, Fibrosis, and Pulmonary Alveolar Proteinosis
US20110044981A1 (en) * 2009-08-21 2011-02-24 Spangler Rhyannon Methods and compositions for treatment of pulmonary fibrotic disorders
US20110044907A1 (en) * 2009-08-21 2011-02-24 Derek Marshall In vivo screening assays
US20110076739A1 (en) * 2009-08-21 2011-03-31 Mccauley Scott Catalytic domains from lysyl oxidase and loxl2
US20110076285A1 (en) * 2009-09-29 2011-03-31 Ingeborg Stalmans Methods and Compositions For Treatment of Ocular Fibrosis
US20110076272A1 (en) * 2009-08-21 2011-03-31 Victoria Smith Therapeutic methods and compositions
WO2011097513A1 (en) * 2010-02-04 2011-08-11 Gilead Biologics, Inc Antibodies that bind to lysyl oxidase-like 2 (loxl2) and methods of use therefor
US20150218563A1 (en) * 2004-07-01 2015-08-06 Dan Littman Compositions and methods for modulation of rorgammat functions
EP3388078A1 (en) * 2015-03-27 2018-10-17 Immatics Biotechnologies GmbH Novel peptides and combination of peptides for use in immunotherapy against various tumors (seq id 23 - mmp-002)
CN111118142A (en) * 2020-01-17 2020-05-08 上海市同济医院 Serum lysyl oxidase as a hierarchical diagnostic biomarker for renal interstitial fibrosis
US11407810B2 (en) 2015-03-27 2022-08-09 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against various tumors

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2359854B1 (en) * 2000-08-08 2013-10-09 Technion Research and Development Foundation, Ltd. Pharmaceutical compositions and methods useful for modulating angiogenesis
US20140186340A1 (en) 2011-04-08 2014-07-03 Gilead Biologics, Inc. Methods and Compositions for Normalization of Tumor Vasculature by Inhibition of LOXL2
EP2714744A1 (en) * 2011-06-01 2014-04-09 Gilead Biologics, Inc. Lysyl oxidase-like 2 assay and methods of use thereof
US10040868B2 (en) 2014-07-24 2018-08-07 Yeda Research And Development Co. Ltd. Antibodies targeted against LOXL-2 for the treatment of collagen-associated pathologies
CN109311978B (en) * 2016-06-06 2022-05-10 艾斯克立必恩股份有限公司 DSG2 monoclonal antibody and use thereof
US11471497B1 (en) 2019-03-13 2022-10-18 David Gordon Bermudes Copper chelation therapeutics

Citations (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3791932A (en) * 1971-02-10 1974-02-12 Akzona Inc Process for the demonstration and determination of reaction components having specific binding affinity for each other
US3839153A (en) * 1970-12-28 1974-10-01 Akzona Inc Process for the detection and determination of specific binding proteins and their corresponding bindable substances
US3850752A (en) * 1970-11-10 1974-11-26 Akzona Inc Process for the demonstration and determination of low molecular compounds and of proteins capable of binding these compounds specifically
US3850578A (en) * 1973-03-12 1974-11-26 H Mcconnell Process for assaying for biologically active molecules
US3853987A (en) * 1971-09-01 1974-12-10 W Dreyer Immunological reagent and radioimmuno assay
US3867517A (en) * 1971-12-21 1975-02-18 Abbott Lab Direct radioimmunoassay for antigens and their antibodies
US3879262A (en) * 1972-05-11 1975-04-22 Akzona Inc Detection and determination of haptens
US3901654A (en) * 1971-06-21 1975-08-26 Biological Developments Receptor assays of biologically active compounds employing biologically specific receptors
US3935074A (en) * 1973-12-17 1976-01-27 Syva Company Antibody steric hindrance immunoassay with two antibodies
US3984533A (en) * 1975-11-13 1976-10-05 General Electric Company Electrophoretic method of detecting antigen-antibody reaction
US3996345A (en) * 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4034074A (en) * 1974-09-19 1977-07-05 The Board Of Trustees Of Leland Stanford Junior University Universal reagent 2-site immunoradiometric assay using labelled anti (IgG)
US4098876A (en) * 1976-10-26 1978-07-04 Corning Glass Works Reverse sandwich immunoassay
US4485088A (en) * 1982-03-26 1984-11-27 Bio-Products, Inc. Method of treatment of fibrotic lesions by topical administration of lathyrogenic drugs
US4666828A (en) * 1984-08-15 1987-05-19 The General Hospital Corporation Test for Huntington's disease
US4683202A (en) * 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4801531A (en) * 1985-04-17 1989-01-31 Biotechnology Research Partners, Ltd. Apo AI/CIII genomic polymorphisms predictive of atherosclerosis
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5011771A (en) * 1984-04-12 1991-04-30 The General Hospital Corporation Multiepitopic immunometric assay
US5021404A (en) * 1988-04-20 1991-06-04 The Children's Medical Center Corporation Angiostatic collagen modulators
US5192659A (en) * 1989-08-25 1993-03-09 Genetype Ag Intron sequence analysis method for detection of adjacent and remote locus alleles as haplotypes
US5272057A (en) * 1988-10-14 1993-12-21 Georgetown University Method of detecting a predisposition to cancer by the use of restriction fragment length polymorphism of the gene for human poly (ADP-ribose) polymerase
US5281521A (en) * 1992-07-20 1994-01-25 The Trustees Of The University Of Pennsylvania Modified avidin-biotin technique
US5545807A (en) * 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US5545806A (en) * 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5625126A (en) * 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5633425A (en) * 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) * 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5721138A (en) * 1992-12-15 1998-02-24 Sandford University Apolipoprotein(A) promoter and regulatory sequence constructs and methods of use
US5879219A (en) * 1997-01-17 1999-03-09 Penjuke; Daniel Balloon inflation and illumination device
US6300092B1 (en) * 1999-01-27 2001-10-09 Millennium Pharmaceuticals Inc. Methods of use of a novel lysyl oxidase-related protein
US6303318B1 (en) * 1997-03-19 2001-10-16 The Board Of Trustees Of The University Of Arkansas Methods for the early diagnosis of ovarian cancer
US6316416B1 (en) * 1995-03-07 2001-11-13 Steven R. Patierno Uteroglobin therapy for epithelial cell cancer
US6326174B1 (en) * 1994-12-02 2001-12-04 The Scripps Research Institute Enzymatic DNA molecules
US20020123476A1 (en) * 1991-03-19 2002-09-05 Emanuele R. Martin Therapeutic delivery compositions and methods of use thereof
US20020128218A1 (en) * 1991-03-19 2002-09-12 Emanuele R. Martin Therapeutic delivery compositions and methods of use thereof
US20030017068A1 (en) * 2000-01-20 2003-01-23 Ignacio Larrain Valve arrangement
US20030096980A1 (en) * 1991-11-26 2003-05-22 Brian Froehler Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
US20030099213A1 (en) * 2001-11-29 2003-05-29 Gui-Jung Lee Wireless radio data protective device for private/public network wireless packet data services and authentication method according to internet connection request of mobile terminals receiving the services

Family Cites Families (136)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4036945A (en) 1976-05-03 1977-07-19 The Massachusetts General Hospital Composition and method for determining the size and location of myocardial infarcts
US4331647A (en) 1980-03-03 1982-05-25 Goldenberg Milton David Tumor localization and therapy with labeled antibody fragments specific to tumor-associated markers
ATE8130T1 (en) * 1980-08-25 1984-07-15 Kabivitrum Ab PEPTIDE SUBSTRATES FOR DETERMINING PROTEASE ACTIVITY.
US4879219A (en) 1980-09-19 1989-11-07 General Hospital Corporation Immunoassay utilizing monoclonal high affinity IgM antibodies
DE3278334D1 (en) * 1981-10-23 1988-05-19 Genetics Int Inc Sensor for components of a liquid mixture
DE3572485D1 (en) 1984-12-22 1989-09-28 Thomae Gmbh Dr K Tetrahydro-benzothiazoles, their production and their use as intermediates or drugs
US4627445A (en) * 1985-04-08 1986-12-09 Garid, Inc. Glucose medical monitoring system
US4965288A (en) * 1988-02-25 1990-10-23 Merrell Dow Pharmaceuticals Inc. Inhibitors of lysyl oxidase
US5059714A (en) * 1988-02-25 1991-10-22 Merrell Dow Pharmaceuticals Inc. Inhibitors of lysyl oxidase
US5252608A (en) * 1988-02-25 1993-10-12 Merrell Dow Pharmaceuticals Inc. Inhibitors of lysyl oxidase
US4943593A (en) * 1988-02-25 1990-07-24 Merrell Dow Pharmaceuticals Inc. Inhibitors of lysyl oxidase
US5182297A (en) * 1988-02-25 1993-01-26 Merrell Dow Pharmaceuticals Inc. Inhibitors of lysyl oxidase
US5021456A (en) * 1988-02-25 1991-06-04 Merrell Dow Pharmaceuticals Inc. Inhibitors of lysyl oxidase
US5216141A (en) 1988-06-06 1993-06-01 Benner Steven A Oligonucleotide analogs containing sulfur linkages
US5120764A (en) * 1988-11-01 1992-06-09 Merrell Dow Pharmaceuticals Inc. Inhibitors of lysyl oxidase
CA2006008C (en) 1988-12-20 2000-02-15 Donald J. Kessler Method for making synthetic oligonucleotides which bind specifically to target sites on duplex dna molecules, by forming a colinear triplex, the synthetic oligonucleotides and methods of use
US5262035A (en) 1989-08-02 1993-11-16 E. Heller And Company Enzyme electrodes
US4997854A (en) * 1989-08-25 1991-03-05 Trustees Of Boston University Anti-fibrotic agents and methods for inhibiting the activity of lysyl oxidase in-situ using adjacently positioned diamine analogue substrates
KR100272077B1 (en) 1990-08-29 2000-11-15 젠팜인터내셔날,인코포레이티드 Transgenic non-human animals capable of producing heterologous antibodies
US5641484A (en) * 1990-12-04 1997-06-24 Board Of Regents, The University Of Texas System Methods for the suppression of neu mediated tumors by adenoviral E1A and SV40 large T antigen
DK51092D0 (en) 1991-05-24 1992-04-15 Ole Buchardt OLIGONUCLEOTIDE ANALOGUE DESCRIBED BY PEN, MONOMERIC SYNTHONES AND PROCEDURES FOR PREPARING THEREOF, AND APPLICATIONS THEREOF
US6015562A (en) * 1992-09-22 2000-01-18 American Cyanamid Company Targeted forms of methyltrithio antitumor agents
AU6049796A (en) 1995-06-07 1996-12-30 Fibromed, Inc. Lysyloxidase inhibitors
AU724579B2 (en) 1996-04-01 2000-09-28 Sankyo Company Limited Anti-Fas recombinant antibodies and DNA therefor
AUPO156596A0 (en) 1996-08-09 1996-09-05 University Of Sydney, The Synthetic polynucleotides
DE69809391T2 (en) * 1997-02-06 2003-07-10 Therasense Inc SMALL VOLUME SENSOR FOR IN-VITRO DETERMINATION
PL199014B1 (en) 1997-08-08 2008-08-29 Univ California Monoclonal antibody specifically fixing to αvß6 integrine, a hybridome producing it and application of antibody
US6277622B1 (en) * 1997-08-11 2001-08-21 The University Of Sydney Synthetic polynucleotides
US6225118B1 (en) 1997-10-01 2001-05-01 Biocure Limited Multicellular in vitro assay of angiogenesis
EP1029048A2 (en) * 1997-11-05 2000-08-23 Baylor College Of Medicine Sequences for targeting metastatic cells
US6140056A (en) * 1999-01-27 2000-10-31 Millennium Pharmaceuticals, Inc. MSP-18 protein and nucleic acid molecules and uses therefor
US20020072089A1 (en) 1999-11-23 2002-06-13 Holtzman Douglas A. Novel ITALY, Lor-2, STRIFE, TRASH, BDSF, LRSG, and STMST protein and nucleic acid molecules and uses therefor
WO1999065928A2 (en) 1998-06-19 1999-12-23 Genzyme Corporation Polynucleotide population isolated from non-metastatic and metastatic breast tumor tissues
US20040058355A1 (en) * 1998-09-30 2004-03-25 Millennium Pharmaceuticals, Inc. Novel 21910, 56634, 55053, 2504, 15977, 14760, 25501, 17903, 3700, 21529, 26176, 26343, 56638, 18610, 33217, 21967, H1983, M1983, 38555 or 593 molecules and uses therefor
US6534261B1 (en) * 1999-01-12 2003-03-18 Sangamo Biosciences, Inc. Regulation of endogenous gene expression in cells using zinc finger proteins
US7108982B1 (en) 1999-02-19 2006-09-19 University Of Iowa Research Foundation Diagnostics and the therapeutics for macular degeneration
US20030149997A1 (en) * 1999-02-19 2003-08-07 Hageman Gregory S. Diagnostics and therapeutics for arterial wall disruptive disorders
GB9906114D0 (en) * 1999-03-18 1999-05-12 Camco Int Uk Ltd A method of applying a wear-resistant layer to a surface of a downhole component
US20030152926A1 (en) * 1999-08-11 2003-08-14 Eos Biotechnology, Inc. Novel methods of diagnosis of angiogenesis, compositions and methods of screening for angiogenesis modulators
US6808707B2 (en) * 2000-02-04 2004-10-26 Matrix Design Wound healing compositions and methods using tropoelastin and lysyl oxidase
EP1328630A2 (en) 2000-04-14 2003-07-23 Incyte Genomics, Inc. Secreted proteins
AU2001261194A1 (en) 2000-05-03 2001-11-12 University Of Hawaii Novel members of the lysyl oxidases family of amine oxidases related applications
WO2001092495A2 (en) 2000-05-26 2001-12-06 Millennium Pharmaceuticals, Inc. A human lysyl ixodase (47765) and uses thereof
US7700359B2 (en) * 2000-06-02 2010-04-20 Novartis Vaccines And Diagnostics, Inc. Gene products differentially expressed in cancerous cells
GB0014185D0 (en) * 2000-06-09 2000-08-02 Novartis Res Found Compound and method
US20090233270A9 (en) * 2000-08-02 2009-09-17 St Croix Brad Secreted and cytoplasmic tumor endothelial markers
US20030114410A1 (en) * 2000-08-08 2003-06-19 Technion Research And Development Foundation Ltd. Pharmaceutical compositions and methods useful for modulating angiogenesis and inhibiting metastasis and tumor fibrosis
MXPA03001196A (en) * 2000-08-08 2004-04-05 Wyeth Corp A novel member of the lysyl oxidase gene family.
EP2359854B1 (en) * 2000-08-08 2013-10-09 Technion Research and Development Foundation, Ltd. Pharmaceutical compositions and methods useful for modulating angiogenesis
US20020156263A1 (en) * 2000-10-05 2002-10-24 Huei-Mei Chen Genes expressed in breast cancer
AU2002241520A1 (en) * 2000-11-28 2003-03-03 University Of Cincinnati Blood assessment of injury
US7112668B2 (en) * 2001-01-23 2006-09-26 Curagen Corporation Polypeptides and nucleic acids encoded thereby
US20040029114A1 (en) * 2001-01-24 2004-02-12 Eos Technology, Inc. Methods of diagnosis of breast cancer, compositions and methods of screening for modulators of breast cancer
WO2002061092A2 (en) 2001-01-29 2002-08-08 Bayer Aktiengesellschaft Regulation of human lysyl oxidase
WO2002079492A2 (en) 2001-02-14 2002-10-10 Protein Design Labs, Inc. Methods of diagnosis of angiogenesis, compositions and methods of screening for angiogenesis modulators
US20020182274A1 (en) * 2001-03-21 2002-12-05 Kung-Ming Lu Methods for inhibiting cancer growth, reducing infection and promoting general health with a fermented soy extract
EP1463928A2 (en) 2001-04-18 2004-10-06 Protein Design Labs Methods of diagnosis of lung cancer, compositions and methods of screening for modulators of lung cancer
US20030211076A1 (en) 2001-05-10 2003-11-13 Wande Li Compositions and methods for treatment of proliferative disorders
US20030092037A1 (en) * 2001-07-18 2003-05-15 Osamu Matsuzaki Elk1 phosphorylation related gene
FR2828206B1 (en) 2001-08-03 2004-09-24 Centre Nat Rech Scient USE OF LYSYL OXIDASE INHIBITORS FOR CELL CULTURE AND TISSUE ENGINEERING
US20030129672A1 (en) 2001-08-29 2003-07-10 Dyer Richard Dennis Method for identifying metalloenzyme inhibitors
US20040248835A1 (en) * 2001-10-26 2004-12-09 Anja Krebs Use of a double-stranded ribonucleic acid for treating an infection with a positivestrand rna-virus
WO2003051388A2 (en) * 2001-12-18 2003-06-26 Mondobiotech Laboratories Anstalt Pharmaceutical composition of interferon gamma or pirfenidone with molecular diagnostics for the improved treatment of interstitial lung diseases
US7186540B2 (en) * 2001-12-27 2007-03-06 National Institute of Advanced Indusrtial Science and Technology Thermostable glutaminase and thermostable glutaminase gene
AU2003217966A1 (en) * 2002-03-07 2003-10-08 Licentia, Ltd. Lymphatic and blood endothelial cell genes
US7655397B2 (en) * 2002-04-25 2010-02-02 The United States Of America As Represented By The Department Of Health And Human Services Selections of genes and methods of using the same for diagnosis and for targeting the therapy of select cancers
WO2004002427A2 (en) * 2002-06-27 2004-01-08 The General Hospital Corporation Methods for the treatment or prevention of obesity
WO2004016758A2 (en) * 2002-08-15 2004-02-26 Genzyme Corporation Brain endothelial cell expression patterns
WO2004023973A2 (en) 2002-09-12 2004-03-25 Incyte Corporation Molecules for diagnostics and therapeutics
EP1556402B1 (en) * 2002-09-25 2011-06-22 University of Massachusetts In vivo gene silencing by chemically modified and stable sirna
US20050259483A1 (en) 2002-09-30 2005-11-24 Oncotherapy Science, Inc. Genes and polypeptides relating to prostate cancers
US20050260639A1 (en) 2002-09-30 2005-11-24 Oncotherapy Science, Inc. Method for diagnosing pancreatic cancer
WO2004048938A2 (en) 2002-11-26 2004-06-10 Protein Design Labs, Inc. Methods of detecting soft tissue sarcoma, compositions and methods of screening for soft tissue sarcoma modulators
CA2508348C (en) * 2002-12-06 2016-07-12 Millennium Pharmaceuticals, Inc. Methods for the identification, assessment, and treatment of patients with proteasome inhibition therapy
US20060188889A1 (en) * 2003-11-04 2006-08-24 Christopher Burgess Use of differentially expressed nucleic acid sequences as biomarkers for cancer
PT1581812E (en) 2003-01-06 2008-09-22 Wyeth Corp Compositions and methods for diagnosing and treating colon cancers
US20050181375A1 (en) * 2003-01-10 2005-08-18 Natasha Aziz Novel methods of diagnosis of metastatic cancer, compositions and methods of screening for modulators of metastatic cancer
US7393531B2 (en) 2003-01-21 2008-07-01 Arius Research Inc. Cytotoxicity mediation of cells evidencing surface expression of MCSP
WO2004078124A2 (en) 2003-03-03 2004-09-16 Board Of Regents, The University Of Texas System Methods and compositions involving mda-7
US8383158B2 (en) * 2003-04-15 2013-02-26 Abbott Cardiovascular Systems Inc. Methods and compositions to treat myocardial conditions
AT500650B1 (en) 2003-04-17 2009-11-15 Altropus Gmbh IMMUNOGENIC RECOMBINANT ANTIBODY
EP1628679A2 (en) 2003-05-23 2006-03-01 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumors of glial origin
JP2007516693A (en) * 2003-06-09 2007-06-28 ザ・リージェンツ・オブ・ザ・ユニバーシティ・オブ・ミシガン Compositions and methods for the treatment and diagnosis of cancer
FR2855969B1 (en) 2003-06-13 2012-11-30 Coletica STIMULATION OF THE ACTIVITY OF A LYSYL OXIDASE ISOFORM TO FIGHT CERTAIN PATHOLOGIES DUE TO AN INCOMPLETE, ABSENT OR DISORGANIZED ELASTOGENESIS
FR2855968B1 (en) * 2003-06-13 2012-11-30 Coletica STIMULATION OF THE SYNTHESIS AND ACTIVITY OF A LYSYL OXIDASE-LIKE LOXL ISOFORM TO STIMULATE THE FORMATION OF ELASTIC FIBERS
US10179935B2 (en) * 2003-07-17 2019-01-15 Pacific Edge Limited Markers for detection of gastric cancer
EP1670494A1 (en) 2003-09-16 2006-06-21 Hadasit Medical Research Services & Development Ltd. Glatiramer acetate for use as an immuno-modulatory agent
WO2005035754A1 (en) 2003-10-14 2005-04-21 Chugai Seiyaku Kabushiki Kaisha Double specific antibodies substituting for functional protein
EP1690932A4 (en) 2003-10-20 2008-03-05 Teijin Pharma Ltd Bone and/or joint disease-associated gene
US20070054278A1 (en) * 2003-11-18 2007-03-08 Applera Corporation Polymorphisms in nucleic acid molecules encoding human enzyme proteins, methods of detection and uses thereof
TW200530269A (en) 2003-12-12 2005-09-16 Chugai Pharmaceutical Co Ltd Anti-Mpl antibodies
US20080261870A1 (en) 2004-01-13 2008-10-23 Trackman Philip C Use of the Pro-Peptide Domain of Lysyl Oxidase as a Therapeutic Agent
EP1706139A4 (en) * 2004-01-23 2009-06-17 Massachusetts Eye & Ear Infirm Lysyl oxidase-like 1 (loxl1) and elastogenesis
CN101088089A (en) 2004-02-23 2007-12-12 鹿特丹伊拉斯姆斯大学医疗中心 Classification, diagnosis and prognosis of acute myeloid leukemia by gene expression profiling
AU2005217623A1 (en) 2004-02-24 2005-09-09 Attenuon Llp Inhibition of superoxide dismutase by tetrathiomolybdate: identification of new anti-angiogenic and antitumor agents
EP1756317A4 (en) 2004-04-01 2008-05-28 Sequenom Inc Methods for identifying risk of osteoarthritis and treatments thereof
CA2583208C (en) * 2004-10-15 2015-08-25 Seattle Genetics, Inc. Anti-cd70 antibody and its use for the treatment and prevention of cancer and immune disorders
US20060134172A1 (en) * 2004-12-21 2006-06-22 Alcon, Inc. Agents which regulate, inhibit, or modulate the activity and/or expression of lysyl oxidase (LOX) and LOX-like proteases as a unique means to both lower intraocular pressure and treat glaucomatous retinopathies/optic neuropathies
JP4171061B2 (en) 2005-02-09 2008-10-22 アーキュール,インコーポレーテッド Maleimide derivatives, pharmaceutical compositions and their use for the treatment of cancer
EP2314614B1 (en) 2005-02-28 2015-11-25 Sangamo BioSciences, Inc. Anti-angiogenic methods and compositions
US20060216722A1 (en) * 2005-03-25 2006-09-28 Christer Betsholtz Glomerular expression profiling
US9234897B2 (en) * 2005-03-31 2016-01-12 Two Cells Co., Ltd Method for distinguishing mesenchymal stem cell using molecular marker and use thereof
US20060223730A1 (en) * 2005-04-04 2006-10-05 Hl Distribution Company Calcium supplements
US20060246492A1 (en) 2005-04-05 2006-11-02 The General Hospital Corporation Method for predicting responsiveness to drugs
WO2006128740A2 (en) 2005-06-02 2006-12-07 Centelion Anti-vascular methods and therapies employing lysyl oxidase inhibitors
US20070225242A1 (en) * 2005-06-21 2007-09-27 The Board Of Trustees Of The Leland Stanford Junior University Method and composition for treating and preventing tumor metastasis in vivo
US20070021365A1 (en) 2005-06-21 2007-01-25 The Board Of Trustees Of The Leland Stanford Junior University Inhibition of Lysyl oxidase for treating tumor growth and diagnostics relating thereto
US7846445B2 (en) 2005-09-27 2010-12-07 Amunix Operating, Inc. Methods for production of unstructured recombinant polymers and uses thereof
US8158107B2 (en) 2005-09-30 2012-04-17 National Jewish Health Genes and proteins associated with angiogenesis and uses thereof
GB0521139D0 (en) 2005-10-18 2005-11-23 Univ Sheffield Therapeutic agent
US20090035348A1 (en) * 2005-11-22 2009-02-05 Z & Z Medical Holdings, Inc. Dissolution of arterial plaque
US8445198B2 (en) * 2005-12-01 2013-05-21 Medical Prognosis Institute Methods, kits and devices for identifying biomarkers of treatment response and use thereof to predict treatment efficacy
GB0524991D0 (en) 2005-12-08 2006-01-18 Ge Healthcare Ltd Novel imaging agents for fibrosis
JP5183484B2 (en) 2005-12-09 2013-04-17 ユセベ ファルマ ソシエテ アノニム Antibody molecule having specificity for human IL-6
US20090275633A1 (en) 2006-04-13 2009-11-05 Oncomethylome Sciences Sa Novel Tumour Suppressor
US20100092476A1 (en) 2006-11-17 2010-04-15 Hanash Samir M Pancreatic cancer biomarkers
US20100074897A1 (en) 2006-12-01 2010-03-25 University Of Utah Research Foundation Methods and Compositions related to HIF-1 alpha
US8077896B2 (en) * 2006-12-12 2011-12-13 Sound Services, Llc Laser inclinometer audio direction
KR101443214B1 (en) * 2007-01-09 2014-09-24 삼성전자주식회사 A composition, kit and microarray for diagnosing the risk of lung cancer recurrence in a patient after lung cancer treatment or a lung cancer patient
GB0708002D0 (en) 2007-04-25 2007-06-06 Univ Sheffield Antibodies
WO2008138578A2 (en) 2007-05-11 2008-11-20 Medical Prognosis Institute Methods, kits, and devices for identifying biomarkers of treatment response and use thereof to predict treatment efficacy
CA2692171C (en) 2007-06-22 2019-10-22 Randolph Watnick Methods and uses thereof of prosaposin
IL184627A0 (en) 2007-07-15 2008-12-29 Technion Res & Dev Foundation Agents for diagnosing and modulating metastasis and fibrosis as well as inflammation in a mammalian tissue
EP2537529B1 (en) 2007-08-02 2018-10-17 Gilead Biologics, Inc. Loxl2 inhibitory antibodies and uses thereof
US8815946B2 (en) 2008-01-25 2014-08-26 University of Pittsburgh—of the Commonwealth System of Higher Education Inhibition of proliferation and fibrotic response of activated corneal stromal cells
US9107935B2 (en) 2009-01-06 2015-08-18 Gilead Biologics, Inc. Chemotherapeutic methods and compositions
CA2751438A1 (en) 2009-02-06 2010-08-12 Arresto Biosciences, Inc. Methods and compositions for treatment of neovascularization
MX2012002271A (en) 2009-08-21 2012-07-20 Gilead Biologics Inc Therapeutic methods and compositions.
SG178845A1 (en) 2009-08-21 2012-04-27 Gilead Biologics Inc Catalytic domains from lysyl oxidase and loxl2
KR20120089274A (en) 2009-08-21 2012-08-09 길리아드 바이오로직스, 인크. In vivo screening assays
US20110044907A1 (en) 2009-08-21 2011-02-24 Derek Marshall In vivo screening assays
JP2013502228A (en) 2009-08-21 2013-01-24 ギリアド バイオロジクス,インク. In vitro screening assay
CA2775877A1 (en) 2009-09-29 2011-04-07 Gilead Biologics, Inc. Methods and compositions for treatment of ocular fibrosis
SG183174A1 (en) 2010-02-04 2012-09-27 Gilead Biologics Inc Antibodies that bind to lysyl oxidase-like 2 (loxl2) and methods of use therefor
US20140186340A1 (en) 2011-04-08 2014-07-03 Gilead Biologics, Inc. Methods and Compositions for Normalization of Tumor Vasculature by Inhibition of LOXL2
EP2714744A1 (en) 2011-06-01 2014-04-09 Gilead Biologics, Inc. Lysyl oxidase-like 2 assay and methods of use thereof

Patent Citations (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3850752A (en) * 1970-11-10 1974-11-26 Akzona Inc Process for the demonstration and determination of low molecular compounds and of proteins capable of binding these compounds specifically
US3839153A (en) * 1970-12-28 1974-10-01 Akzona Inc Process for the detection and determination of specific binding proteins and their corresponding bindable substances
US3791932A (en) * 1971-02-10 1974-02-12 Akzona Inc Process for the demonstration and determination of reaction components having specific binding affinity for each other
US3901654A (en) * 1971-06-21 1975-08-26 Biological Developments Receptor assays of biologically active compounds employing biologically specific receptors
US3853987A (en) * 1971-09-01 1974-12-10 W Dreyer Immunological reagent and radioimmuno assay
US3867517A (en) * 1971-12-21 1975-02-18 Abbott Lab Direct radioimmunoassay for antigens and their antibodies
US3879262A (en) * 1972-05-11 1975-04-22 Akzona Inc Detection and determination of haptens
US3850578A (en) * 1973-03-12 1974-11-26 H Mcconnell Process for assaying for biologically active molecules
US3935074A (en) * 1973-12-17 1976-01-27 Syva Company Antibody steric hindrance immunoassay with two antibodies
US3996345A (en) * 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4034074A (en) * 1974-09-19 1977-07-05 The Board Of Trustees Of Leland Stanford Junior University Universal reagent 2-site immunoradiometric assay using labelled anti (IgG)
US3984533A (en) * 1975-11-13 1976-10-05 General Electric Company Electrophoretic method of detecting antigen-antibody reaction
US4098876A (en) * 1976-10-26 1978-07-04 Corning Glass Works Reverse sandwich immunoassay
US4485088A (en) * 1982-03-26 1984-11-27 Bio-Products, Inc. Method of treatment of fibrotic lesions by topical administration of lathyrogenic drugs
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5011771A (en) * 1984-04-12 1991-04-30 The General Hospital Corporation Multiepitopic immunometric assay
US4666828A (en) * 1984-08-15 1987-05-19 The General Hospital Corporation Test for Huntington's disease
US4683202A (en) * 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4683202B1 (en) * 1985-03-28 1990-11-27 Cetus Corp
US4801531A (en) * 1985-04-17 1989-01-31 Biotechnology Research Partners, Ltd. Apo AI/CIII genomic polymorphisms predictive of atherosclerosis
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5021404A (en) * 1988-04-20 1991-06-04 The Children's Medical Center Corporation Angiostatic collagen modulators
US5545807A (en) * 1988-10-12 1996-08-13 The Babraham Institute Production of antibodies from transgenic animals
US5272057A (en) * 1988-10-14 1993-12-21 Georgetown University Method of detecting a predisposition to cancer by the use of restriction fragment length polymorphism of the gene for human poly (ADP-ribose) polymerase
US5192659A (en) * 1989-08-25 1993-03-09 Genetype Ag Intron sequence analysis method for detection of adjacent and remote locus alleles as haplotypes
US5633425A (en) * 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5545806A (en) * 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5625126A (en) * 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5661016A (en) * 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US20020128218A1 (en) * 1991-03-19 2002-09-12 Emanuele R. Martin Therapeutic delivery compositions and methods of use thereof
US20020123476A1 (en) * 1991-03-19 2002-09-05 Emanuele R. Martin Therapeutic delivery compositions and methods of use thereof
US20030096980A1 (en) * 1991-11-26 2003-05-22 Brian Froehler Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
US5281521A (en) * 1992-07-20 1994-01-25 The Trustees Of The University Of Pennsylvania Modified avidin-biotin technique
US5721138A (en) * 1992-12-15 1998-02-24 Sandford University Apolipoprotein(A) promoter and regulatory sequence constructs and methods of use
US6326174B1 (en) * 1994-12-02 2001-12-04 The Scripps Research Institute Enzymatic DNA molecules
US6316416B1 (en) * 1995-03-07 2001-11-13 Steven R. Patierno Uteroglobin therapy for epithelial cell cancer
US5879219A (en) * 1997-01-17 1999-03-09 Penjuke; Daniel Balloon inflation and illumination device
US6303318B1 (en) * 1997-03-19 2001-10-16 The Board Of Trustees Of The University Of Arkansas Methods for the early diagnosis of ovarian cancer
US6300092B1 (en) * 1999-01-27 2001-10-09 Millennium Pharmaceuticals Inc. Methods of use of a novel lysyl oxidase-related protein
US20030017068A1 (en) * 2000-01-20 2003-01-23 Ignacio Larrain Valve arrangement
US20030099213A1 (en) * 2001-11-29 2003-05-29 Gui-Jung Lee Wireless radio data protective device for private/public network wireless packet data services and authentication method according to internet connection request of mobile terminals receiving the services

Cited By (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050164968A1 (en) * 2001-05-18 2005-07-28 Sirna Therapeutics, Inc. RNA interference mediated inhibition of ADAM33 gene expression using short interfering nucleic acid (siNA)
US20040014113A1 (en) * 2002-05-31 2004-01-22 The Regents Of The University Of California Method for efficient RNA interference in mammalian cells
WO2003102214A3 (en) * 2002-05-31 2005-02-17 Univ California Method for efficient rna interference in mammalian cells
US8119610B2 (en) 2002-05-31 2012-02-21 The Regents Of The University Of California Method for efficient RNA interference in mammalian cells
US8815823B2 (en) 2002-11-27 2014-08-26 Technion Research & Development Foundation Ltd. Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
US20060127402A1 (en) * 2002-11-27 2006-06-15 Technion Research & Development Foundation Ltd. Pharmaceutical compositions and methods useful for modulating angiogenesis, inhibiting metastasis and tumor fibrosis, and assessing the malignancy of colon cancer tumors
US8163494B2 (en) 2002-11-27 2012-04-24 Technion Research & Development Foundation Ltd. Method for assessing metastatic properties of breast cancer
US8168180B2 (en) 2002-11-27 2012-05-01 Technion Research & Development Foundation Ltd. Methods and compositions for modulating angiogenesis
US20030232400A1 (en) * 2002-12-20 2003-12-18 Susan Radka Methods of screening subjects for expression of soluble receptors of vascular endothelial growth factor (VEGF) for use in managing treatment and determining prognostic outcome
US20150218563A1 (en) * 2004-07-01 2015-08-06 Dan Littman Compositions and methods for modulation of rorgammat functions
WO2006128740A3 (en) * 2005-06-02 2007-04-12 Centelion Anti-vascular methods and therapies employing lysyl oxidase inhibitors
WO2006128740A2 (en) * 2005-06-02 2006-12-07 Centelion Anti-vascular methods and therapies employing lysyl oxidase inhibitors
US20070225242A1 (en) * 2005-06-21 2007-09-27 The Board Of Trustees Of The Leland Stanford Junior University Method and composition for treating and preventing tumor metastasis in vivo
US20070021365A1 (en) * 2005-06-21 2007-01-25 The Board Of Trustees Of The Leland Stanford Junior University Inhibition of Lysyl oxidase for treating tumor growth and diagnostics relating thereto
EP2010224A2 (en) * 2006-04-27 2009-01-07 The Board of Trustees of The Leland Stanford Junior University Method and composition for treating and preventing tumor metastasis in vivo
EP2010224A4 (en) * 2006-04-27 2010-10-27 Univ Leland Stanford Junior Method and composition for treating and preventing tumor metastasis in vivo
US8389709B2 (en) 2007-07-15 2013-03-05 Technion Research & Development Foundation Ltd. Compositions and methods for treating tumors, fibrosis, and pulmonary alveolar proteinosis
US9617347B2 (en) 2007-07-15 2017-04-11 Technion Research & Development Foundation Limited Compositions and methods for treating tumors, fibrosis, and pulmonary alveolar proteinosis
US9255271B2 (en) 2007-07-15 2016-02-09 Technion Research & Development Foundation Ltd Compositions and methods for treating tumors, fibrosis, and pulmonary alveolar proteinosis
US20100317721A1 (en) * 2007-07-15 2010-12-16 Technion Research & Development Foundation Ltd. Compositions and Methods for Treating Tumors, Fibrosis, and Pulmonary Alveolar Proteinosis
US8815824B2 (en) 2007-07-15 2014-08-26 Technion Research & Development Foundation Ltd. Compositions and methods for treating tumors, fibrosis, and pulmonary alveolar proteinosis
US8461303B2 (en) 2007-08-02 2013-06-11 Gilead Biologics, Inc. LOX and LOXL2 inhibitors and uses thereof
US10494443B2 (en) 2007-08-02 2019-12-03 Gilead Biologics, Inc. LOX and LOXL2 inhibitors and uses thereof
US8658167B2 (en) 2007-08-02 2014-02-25 Gilead Biologics, Inc. Methods and compositions for treatment and diagnosis of fibrosis, tumor invasion, angiogenesis, and metastasis
US9176139B2 (en) 2007-08-02 2015-11-03 Gilead Biologics, Inc. LOX and LOXL2 inhibitors and uses thereof
US8679485B2 (en) 2007-08-02 2014-03-25 Gilead Biologics, Inc. Methods and compositions for treatment and diagnosis of fibrosis, tumor invasion, angiogenesis, and metastasis
US20090053224A1 (en) * 2007-08-02 2009-02-26 Arresto Biosciences Lox and loxl2 inhibitors and uses thereof
US9107935B2 (en) 2009-01-06 2015-08-18 Gilead Biologics, Inc. Chemotherapeutic methods and compositions
US20100209415A1 (en) * 2009-01-06 2010-08-19 Victoria Smith Chemotherapeutic methods and compositions
US9289447B2 (en) 2009-01-06 2016-03-22 Gilead Biologics, Inc. Chemotherapeutic methods and compositions
US20100203062A1 (en) * 2009-02-06 2010-08-12 Ingeborg Stalmans Methods and Compositions for Treatment of Neovascularization
US8512990B2 (en) 2009-08-21 2013-08-20 Gilead Biologics, Inc. Catalytic domains from lysyl oxidase and LOXL2
US20110076739A1 (en) * 2009-08-21 2011-03-31 Mccauley Scott Catalytic domains from lysyl oxidase and loxl2
US8927700B2 (en) 2009-08-21 2015-01-06 Gilead Biologics, Inc. Catalytic domains from lysyl oxidase and LOXL2
US20110044981A1 (en) * 2009-08-21 2011-02-24 Spangler Rhyannon Methods and compositions for treatment of pulmonary fibrotic disorders
US20110044907A1 (en) * 2009-08-21 2011-02-24 Derek Marshall In vivo screening assays
WO2011022667A3 (en) * 2009-08-21 2011-04-14 Arresto Biosciences, Inc Catalytic domains from lysyl oxidase and loxl2
US20110076272A1 (en) * 2009-08-21 2011-03-31 Victoria Smith Therapeutic methods and compositions
US20110076285A1 (en) * 2009-09-29 2011-03-31 Ingeborg Stalmans Methods and Compositions For Treatment of Ocular Fibrosis
US20110200606A1 (en) * 2010-02-04 2011-08-18 Mccauley Scott Alan Antibodies that Bind to Lysyl Oxidase-Like 2 (LOXL2) and Methods of Use Therefor
WO2011097513A1 (en) * 2010-02-04 2011-08-11 Gilead Biologics, Inc Antibodies that bind to lysyl oxidase-like 2 (loxl2) and methods of use therefor
US8680246B2 (en) 2010-02-04 2014-03-25 Gilead Biologics, Inc. Antibodies that bind to lysyl oxidase-like 2 (LOXL2)
EP3388078A1 (en) * 2015-03-27 2018-10-17 Immatics Biotechnologies GmbH Novel peptides and combination of peptides for use in immunotherapy against various tumors (seq id 23 - mmp-002)
US10501522B2 (en) 2015-03-27 2019-12-10 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against various tumors
US10934338B2 (en) 2015-03-27 2021-03-02 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against various tumors
US11407810B2 (en) 2015-03-27 2022-08-09 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against various tumors
US11434273B2 (en) 2015-03-27 2022-09-06 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against various tumors
US11466072B2 (en) 2015-03-27 2022-10-11 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against various tumors
US11702460B2 (en) 2015-03-27 2023-07-18 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against various tumors
US11873329B2 (en) 2015-03-27 2024-01-16 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against various tumors
US11897934B2 (en) 2015-03-27 2024-02-13 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against various tumors
CN111118142A (en) * 2020-01-17 2020-05-08 上海市同济医院 Serum lysyl oxidase as a hierarchical diagnostic biomarker for renal interstitial fibrosis

Also Published As

Publication number Publication date
EP2062919A2 (en) 2009-05-27
US20060127402A1 (en) 2006-06-15
CY1117943T1 (en) 2017-05-17
DK2062919T3 (en) 2016-05-30
AU2003286391A1 (en) 2004-06-18
EP2062919A3 (en) 2009-07-01
US20120202206A1 (en) 2012-08-09
US20140302499A1 (en) 2014-10-09
EP1572100A2 (en) 2005-09-14
WO2004047720A3 (en) 2006-02-16
HK1131165A1 (en) 2010-01-15
US20120165398A1 (en) 2012-06-28
EP1572100A4 (en) 2008-04-02
US8815823B2 (en) 2014-08-26
US8163494B2 (en) 2012-04-24
US20190040470A1 (en) 2019-02-07
ES2584847T3 (en) 2016-09-29
EP2062918A2 (en) 2009-05-27
US8168180B2 (en) 2012-05-01
US20100119515A1 (en) 2010-05-13
EP2062919B1 (en) 2016-05-04
WO2004047720A2 (en) 2004-06-10
PT2062919E (en) 2016-06-08
AU2003286391A8 (en) 2004-06-18
EP2062918A3 (en) 2009-07-01

Similar Documents

Publication Publication Date Title
US20030114410A1 (en) Pharmaceutical compositions and methods useful for modulating angiogenesis and inhibiting metastasis and tumor fibrosis
DK2527441T3 (en) Compositions and uses for the treatment of pulmonary alveolar proteinosis
EP2359854B1 (en) Pharmaceutical compositions and methods useful for modulating angiogenesis
US20030216345A1 (en) Histone deacetylase inhibitor and use thereof
AU2014201635B2 (en) Compositions and methods for treating tumors, fibrosis, and pulmonary alveolar proteinosis

Legal Events

Date Code Title Description
AS Assignment

Owner name: TECHNION RESEARCH AND DEVELOPMENT FOUNDATION LTD.,

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:NEUFELD, GERA;AKIRI, GAL;VADASZ, ZAHAVA;AND OTHERS;REEL/FRAME:013547/0606;SIGNING DATES FROM 20021103 TO 20021114

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION