WO2003066679A1 - Rhamnose binding protein - Google Patents

Rhamnose binding protein Download PDF

Info

Publication number
WO2003066679A1
WO2003066679A1 PCT/AU2003/000135 AU0300135W WO03066679A1 WO 2003066679 A1 WO2003066679 A1 WO 2003066679A1 AU 0300135 W AU0300135 W AU 0300135W WO 03066679 A1 WO03066679 A1 WO 03066679A1
Authority
WO
WIPO (PCT)
Prior art keywords
rbp
rhamnose
antibody
cancer
cell
Prior art date
Application number
PCT/AU2003/000135
Other languages
English (en)
French (fr)
Inventor
Richard J Lipscombe
Stephen John Carter
Michael Ruane
Original Assignee
Solbec Pharmaceuticals Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Solbec Pharmaceuticals Limited filed Critical Solbec Pharmaceuticals Limited
Priority to NZ534485A priority Critical patent/NZ534485A/en
Priority to AU2003202322A priority patent/AU2003202322B2/en
Priority to JP2003566050A priority patent/JP2005538931A/ja
Priority to CA002475066A priority patent/CA2475066A1/en
Priority to EP03700718A priority patent/EP1472287A4/en
Publication of WO2003066679A1 publication Critical patent/WO2003066679A1/en
Priority to HK06100115.4A priority patent/HK1080089A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • 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

Definitions

  • the present invention relates to an isolated rhamnose binding protein (RBP) that is over expressed in cancer cells relative to non-cancer cells.
  • RBP rhamnose binding protein
  • the present invention also relates to methods of diagnosing cancer by detecting RBP levels and to RBP agonists such as antibodies and methods of treating and diagnosing cancer using RBP agonists.
  • BEC® is a mixture of the triglycosides: solasonine and solamargine that has anti- cancer activity. Studies on the mode of action of BEC® indicate that the glycosides gain entry to cancer cells via a cell surface receptor and that the in vitro toxicity of BEC® to cancer cells is reduced by co-administration of rhamnose.
  • EEL endogenous endocytic ligand receptors
  • the present invention provides an isolated RBP with at least one of the following characteristics: (a) a molecular weight of approximately 65-70 kDa and more preferably 66- 69kDa;
  • the ability of the RBP to bind ligands such as rhamnose to a RBP bearing cell, such as a carcinoma render it useful in various methods. For example, it has been found that when the RBP binds a ligand, such as rhamnose, cell adhesion of the RBP bearing cells is inhibited.
  • the present invention also provides a method of inhibiting cell adhesion between RBP bearing cells comprising the step of contacting the RBP bearing cells with an effective amount of a RBP ligand.
  • the effective amount may be varied depending on the circumstances and may be determined by those skilled in the art. However, when the RBP ligand is rhamnose the effective amount may be approximately 70 picograms/cell.
  • the ligand may be internalised in the cell or remain on the cell surface. Whether or not a ligand is internalised after binding to a cell associated RBP of the present invention depends on a variety of factors such as the molecular weight, charge, structure and/or biological activity of the ligand.
  • the present invention also provides a method of delivering an agent to a RBP bearing cell comprising contacting an agent-ligand complex with the RBP bearing cell.
  • the agent may be delivered to the cell surface or inside the cell by selecting an appropriate ligand-agent complex. For example, by selecting an agent-complex of a certain molecular weight or structure it is possible to control the delivery of the agent to the cell surface or the inside of the cell. In this regard, it has been found that if the agent is above a certain threshold weight then it cannot be efficiently internalised in by the RBP bearing cell and will remain at the cell surface.
  • the present invention also provides a method of detecting a RBP bearing cell comprising the steps of: (i) contacting a cell or tissue sample with an agent adapted to selectively bind to RBP and (ii) detecting the RBP bearing cells.
  • the agent may be varied and includes antibodies and other ligands or agonists that are adapted to bind to RBP. Furthermore, to ease detection of the RBP bearing cells the agent may be adapted to be visualised.
  • the RBP of the present invention may be used to identify agents that bind to the RBP and thus can be used in assays for the RBP, as diagnostics to identify RBP bearing cells or to target therapeutic agents to cancer cells via the RBP.
  • the RBP may also form a component of a screening system for antagonists or agonists of agents that bind to the RBP.
  • Figure 1 depicts a PAGE gel containing fractions 1-9 eluted from a biotinylated rhamnose-ITC affinity column using 100mM free rhamnose and lane 10 contains standard molecular weight markers;
  • Figure 2A is a fluoro-image of proteins crosslinked to FRITC and analysed on a
  • Figure 2B is the total proteins from the gel depicted in Figure 2A stained with
  • Figure 3 is a graph used to calculate the molecular mass of the proteins in Figure
  • Figure 4 is an image of A2058 cells following incubation with 12 ⁇ fluorescein rhamnose-ITC at 37°C for 15min in HEPES buffered saline containing 2mM Ca 2+ and Mg 2+ ;
  • Figure 5 is a plot of the relationship between dose per cell at LD 50 and the Day 1 cell density for each cell line;
  • Figure 6 depicts the data in Figure 5 condensed and fitted to a single exponential function
  • Figure 7 is a plot of the relationship between dose per cell at LD 50 and the Day 1 cell density for two particular breast cancer lines;
  • Figure 8 is a comparison of the plots in Figures 6 and 7;
  • Figure 9 is table containing single point LD50 data from another 11 carcinomas
  • Figure 10 is a graphical representation of the data presented in the table in
  • Figure 11 illustrates the protective effects of rhamnose when co-administered with BEC® via a graph of % cell (A2058, 600 cells) survival v's concentration of
  • BEC® Figure 12: illustrates the protective effects of rhamnose when co-administered with BEC® via a graph of % cell (A2058, 5000 cells) survival v's concentration of
  • FIG. 13 illustrates a fluoro-image of A2058 proteins crosslinked to FRITC, solvent extracted and analysed on a 4-20% SDS polyacrylamide gel. From left, Lane 1 : Standards 83, 42.3, 32.2, 18.8kD; Lane 2 blank; Lanes 3-8: replicate flasks of cells + approx 5 ⁇ M FRITC + 100 ⁇ M carbonyl di-imidazole; and Figure 14 illustrates immunoprecipitation of FRITC -protein cross-linked complex analysed on a 4-20% SDS polyacrylamide gel.
  • Rhamnose binding protein RBP
  • the present invention is based on the isolation and identification of a cellular receptor of the lectin group that is more abundant on neoplastic (cancer) cells than non-cancer cells.
  • the receptor (“RBP”) is adapted to bind and internalise rhamnose and thus represents a valuable diagnostic and therapeutic tool.
  • the RBP and other polypeptides of the invention may be in a substantially isolated form. In this regard, it will be understood that they may be mixed with carriers or diluents that will not interfere with their intended purpose and still be regarded as substantially isolated.
  • a polypeptide of the invention may also be in a substantially purified form, in which case it will generally comprise the polypeptide in a preparation in which at least 90%, 95%, 98% or 99% of the protein in the preparation is a polypeptide of the invention.
  • the RBP of the present invention may be used in assays to identify compounds that interact with (e.g., bind to) it.
  • the compounds which may be screened in accordance with the invention include, but are not limited to peptides, antibodies and fragments thereof, and other organic compounds (e.g., peptidomimetics) that bind to the RBP and either mimic the activity triggered by the natural ligand - rhamnose (i.e., agonists) or inhibit the activity triggered by the natural ligand - rhamnose (i.e., antagonists).
  • organic compounds e.g., peptidomimetics
  • Other compounds that may be screened according to the present invention are peptides, antibodies or fragments thereof, and other organic compounds that mimic the extra cellular domain of the RBP (or a portion thereof) and bind to and "neutralize" natural ligand such as rhamnose.
  • Such compounds may include, but are not limited to, peptides such as, for example, soluble peptides, including but not limited to members of random peptide libraries; and combinatorial chemistry-derived molecular library made of D- and/or L- configuration amino acids, phosphopeptides including, but not limited to, members of random or partially degenerate, directed phosphopeptide libraries, antibodies (including, but not limited to, polyclonal, monoclonal, humanized, anti- idiotypic, chimeric or single chain antibodies, and FAb, F(ab').sub.2 and FAb expression library fragments, and epitope-binding fragments thereof), and small organic or inorganic molecules.
  • peptides such as, for example, soluble peptides, including but not limited to members of random peptide libraries; and combinatorial chemistry-derived molecular library made of D- and/or L- configuration amino acids, phosphopeptides including, but not limited to, members of random or partially degenerate, directed phospho
  • Computer modelling and searching technologies permit identification of compounds, or the improvement of already identified compounds, that can modulate RBP expression or activity. Having identified such a compound or composition, the active sites or regions are identified. Such active sites might typically be ligand binding sites, such as the interaction domains of rhamnose with RBP itself.
  • the active site can be identified using methods known in the art including, for example, from study of complexes of RBP with rhamnose. In this regard, chemical or X-ray crystallographic methods can be used to find the active site by finding where on the factor the complexed ligand is found. Next, the three dimensional geometric structure of the active site is determined. This can be done by known methods, including X-ray crystallography, which can determine a complete molecular structure. On the other hand, solid or liquid phase NMR can be used to determine certain intra-molecular distances.
  • candidate modulating compounds can be identified by searching databases containing compounds along with information on their molecular structure. Such a search seeks compounds having structures that match the determined active site structure and that interact with the groups defining the active site. Such a search can be manual, but is preferably computer assisted. These compounds found from this search are potential RBP modulating compounds. Alternatively, these methods can be used to identify improved modulating compounds from an already known modulating compound or ligand.
  • the composition of the known compound can be modified and the structural effects of modification can be determined using the experimental and computer modelling methods described above applied to the new composition. The altered structure is then compared to the active site structure of the compound to determine if an improved fit or interaction results. In this manner systematic variations in composition, such as by varying side groups, can be quickly evaluated to obtain modified modulating compounds or ligands of improved specificity or activity.
  • Compounds identified via assays such as those described herein may be useful, for example, in elaborating the biological function of the RBP and for treating cancer.
  • the compounds capable of binding RBP may also be used to identify and isolate RBP homologues.
  • the compounds may be used to screen various cell types such as cancer cell types to locate variants of the RBP that could be used to design specific therapeutic agents for treatment of related cancers.
  • In vitro systems may be designed to identify compounds capable of interacting with (e.g., binding to) RBP (including, but not limited to, the extra cellular domain of RBP). These compounds may be useful, for example, in modulating the activity of wild type and/or mutant RBP; elaborating the biological function of the RBP; screening for compounds that disrupt normal RBP interactions; or may in themselves disrupt such interactions.
  • the principle of the assays used to identify compounds that bind to the RBP involves preparing a reaction mixture of the RBP and the test compound under conditions and for a time sufficient to allow the two components to interact and bind, thus forming a complex which can be removed and/or detected in the reaction mixture.
  • the RBP species used can vary depending upon the goal of the screening assay.
  • the full length RBP, or a soluble truncated RBP e.g., in which the transmembrane or cellular domain is deleted from the molecule
  • a peptide corresponding to the extracellular domain or a fusion protein comprising the RBP extracellular domain fused to a protein or polypeptide that affords advantages in the assay system e.g., labelling, isolation of the resulting complex, etc.
  • the screening assays can be conducted in a variety of ways.
  • one method to conduct such an assay involves anchoring the RBP or fusion protein or the test substance onto a solid phase and detecting RBP/test compound complexes anchored on the solid phase at the end of the reaction.
  • the RBP may be anchored onto a solid surface, and the test compound, which is not anchored, may be labelled, either directly or indirectly.
  • microtiter plates may conveniently be utilized as the solid phase.
  • the anchored component may be immobilized by non-covalent or covalent attachments. Non-covalent attachment may be accomplished by simply coating the solid surface with a solution of the RBP or test compound and drying.
  • an immobilized antibody such as a monoclonal antibody, specific for the protein to be immobilized may be used to anchor the protein to the solid surface.
  • the nonimmobilized component is added to the coated surface containing the anchored component. After the reaction is complete, unreacted components are removed (e.g., by washing) under conditions such that any complexes formed will remain immobilized on the solid surface.
  • the detection of complexes anchored on the solid surface can be accomplished in a number of ways. Where the previously nonimmobilized component is pre-labelled, the detection of label immobilized on the surface indicates that complexes were formed.
  • an indirect label can be used to detect complexes anchored on the surface; e.g., using a labelled antibody specific for the previously nonimmobilized component (the antibody, in turn, may be directly labelled or indirectly labelled with a labelled anti-lg antibody).
  • a reaction can be conducted in a liquid phase, the reaction products separated from unreacted components, and complexes detected; e.g., using an immobilized antibody specific for RBP or the test compound to anchor any complexes formed in solution, and a labelled antibody specific for the other component of the possible complex to detect anchored complexes.
  • Cell-based assays can also be used to identify compounds that interact with RBP.
  • cell lines that naturally express RBP such as a cancer cell line selected from the group comprising: HT-29, LS174-T. AGS, 5637, A431 , 786-O, Hs578Bst, CCD 18Lu, HeLa 229, HepG2, JAM, N036, U87-MG, DV145, LNCaP and A2058, or cell lines (e.g., COS cells, CHO cells, fibroblasts, etc.) that have been genetically engineered to express RBP (e.g., by transfection or transduction of RBP DNA) can be used. Interaction of the test compound with, for example, the extracellular domain of RBP expressed by the host cell can be determined by comparison or competition with native rhamnose.
  • the RBP of the present invention and agonists thereof can be employed for the diagnostic and prognostic evaluation of cancer.
  • Such methods may, for example, utilize reagents such as the antibodies described herein.
  • reagents may be used, for example, to detect an over-abundance of RBP relative to normal cells.
  • the present invention provides a method for detecting cancer in a sample comprising the steps of: (i) detecting the level of RBP in the sample; and (ii) comparing it to the level of RBP in a sample from a non-cancer source.
  • the detection method of the present invention may be used to diagnose cancer in vitro.
  • the present invention provides a method of diagnosing cancer in a patient comprising the steps of: (i) detecting the level of RBP in a sample from the patient; and (ii) comparing it to the level of RBP in a sample from a non-cancer source.
  • the detection method may be used to diagnose cancer in vivo.
  • agents that are adapted to bind to RBP can be labelled and administered to a subject suspected of having cancer and later detected to perform the diagnosis.
  • the present invention also provides a method of diagnosing cancer in a patient comprising the steps of: (i) detecting the level and/or distribution of RBP in the patient; and (ii) analysing the distribution and/or levels of RBP to identify differences that are indicative of cancer.
  • the methods described herein may be performed, for example, by utilizing prepackaged diagnostic kits comprising at least one specific RBP antibody reagent described herein, which may be conveniently used, e.g., in clinical settings, to diagnose patients suspected of having cancer.
  • RBP antibodies and other agonists of RBP may be used as cancer diagnostics and prognostics, as described herein. Such diagnostic methods may be used to detect abnormalities in the level of RBP and may be performed in vivo or in vitro, such as, for example, on biopsy tissue.
  • antibodies directed to epitopes of the RBP can be used in vivo to detect the pattern and level of expression of the RBP in the body.
  • Such antibodies can be labelled, e.g., with a radio-opaque or other appropriate compound and injected into a subject in order to visualize binding to the RBP expressed in the body using methods such as X-rays, CAT-scans, or MRI.
  • Labelled antibody fragments e.g., the Fab or single chain antibody comprising the smallest portion of the antigen binding region may also be used for this purpose.
  • account must be taken on background signal or "noise" from non-cancer cells that also bear the RBP, albeit at lower levels. However, those skilled in the art are readily able to discern noise from actual signal in performing the diagnosis.
  • Immunoassays or fusion protein detection assays can also be used to diagnose or type cancer in biopsy or autopsy samples in vitro.
  • Agonists described herein including antibodies, or fragments of antibodies may also be used to quantitatively or qualitatively detect the presence of RBP or conserved variants or peptide fragments thereof. This can be accomplished, for example, by immunofluorescence techniques employing a fluorescently labelled antibody coupled with light microscopic, flow cytometric, or fluorimetric detection.
  • agonists such as antibodies (or fragments thereof) of the present invention may, additionally, be employed histologically, as in immunofluorescence, immunoelectron microscopy or non-immuno assays, for in situ detection of RBP or conserved variants or peptide fragments thereof.
  • In situ detection may be accomplished by removing a histological specimen from a patient, and applying thereto a labelled antibody or fusion protein of the present invention.
  • the antibody (or fragment) or fusion protein is preferably applied by overlaying the labelled antibody (or fragment) onto a biological sample.
  • Immunoassays and non-immunoassays for RBP or conserved variants or peptide fragments thereof will typically comprise incubating a sample, such as a biological fluid, a tissue extract, freshly harvested cells, or lysates of cells which have been incubated in cell culture, in the presence of a detectably labelled antibody capable of identifying RBP or conserved variants or peptide fragments thereof, and detecting the bound antibody by any of a number of techniques well-known in the art.
  • the biological sample may be brought in contact with and immobilized onto a solid phase support or carrier such as nitrocellulose, or other solid support that is capable of immobilizing cells, cell particles or soluble proteins.
  • the support may then be washed with suitable buffers followed by treatment with the detectably labelled RBP antibody or other agonist.
  • the solid phase support may then be washed with the buffer a second time to remove unbound antibody.
  • the amount of bound label on solid support may then be detected by conventional means.
  • solid phase support or carrier any support capable of binding an antigen or an antibody.
  • supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite.
  • the nature of the carrier can be either soluble to some extent or insoluble for the purposes of the present invention.
  • the support material may have virtually any possible structural configuration so long as the coupled molecule is capable of binding to an antigen or antibody.
  • the support configuration may be spherical, as in a bead, or cylindrical, as in the inside surface of a test tube, or the external surface of a rod.
  • the surface may be flat such as a sheet, test strip, etc.
  • Preferred supports include polystyrene beads. Those skilled in the art will know many other suitable carriers for binding antibody or antigen, or will be able to ascertain the same by use of routine experimentation.
  • one of the ways in which the antibody can be detectably labelled is by linking the same to an enzyme. This then renders the antibody suitable for use in an enzyme immunoassay (EIA).
  • EIA enzyme immunoassay
  • the enzyme that is bound to the antibody will react with an appropriate substrate, preferably a chromogenic substrate, in such a manner as to produce a chemical moiety that can be detected, for example, by spectrophotometric, fluorimetric or by visual means.
  • Enzymes which can be used to detectably label the antibody include, but are not limited to, malate dehydrogenase, staphylococcal nuclease, delta-5- steroid isomerase, yeast alcohol dehydrogenase, alphaglycerophosphate, dehydrogenase, those phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and acetylcholinesterase.
  • the detection can be accomplished by calorimetric methods that employ a chromogenic substrate for the enzyme. Detection may also be accomplished by visual comparison of the extent of enzymatic reaction of a substrate in comparison with similarly prepared standards.
  • Detection may also be accomplished using any of a variety of other immunoassays.
  • a radioimmunoassay RIA
  • the radioactive isotope can be detected by such means as the use of a gamma counter or a scintillation counter or by autoradiography.
  • fluorescent labelling compounds fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, o- phthaldehyde and fluorescamine.
  • Agonists such as antibodies can also be detectably labelled using fluorescence emitting metals such as 152 Eu, or others of the lanthanide series. These metals can be attached to the antibody using such metal chelating groups such as diethylenetriaminepentacetic acid (DTPA) or ethylenediaminetetraacetic acid (EDTA).
  • DTPA diethylenetriaminepentacetic acid
  • EDTA ethylenediaminetetraacetic acid
  • the antibody or other agonist can also be detectably labelled by coupling it to a chemiluminescent compound.
  • the presence of the chemiluminescent-tagged antibody is then determined by detecting the presence of luminescence that arises during the course of a chemical reaction.
  • particularly useful chemiluminescent labelling compounds are luminol, isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester.
  • a bioluminescent compound may be used to label the antibody or other agonist of the present invention.
  • Bioluminescence is a type of chemiluminescence found in biological systems in, which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence.
  • Important bioluminescent compounds for purposes of labelling are luciferin, luciferase and aequorin.
  • the ability of the agonists of the present invention bind to RBP and subsequently become internalised in the target cell renders them useful for preferentially delivering agents to cells with a higher load of RBP, such as cancer cells.
  • the agents linked to the agonists of the present invention may be any agent that is adapted to prevent cell growth or division or cause cell death such as, Doxorubicin, Daunorubicin, Vincristine, Vimblastine, Vindesine, Methothrexate, Cytarabine, Etopside, Cisplatin, Carboplatin, 5- Fluorouracil, Bleomycin, Epirubicin, Cyproterone, Irinotecan etc.
  • the agonists of the present invention may be used to treat cancer in a patient.
  • the present invention provides a method of treating cancer in a subject comprising administering a therapeutically effective amount of a RBP agonist anticancer conjugate to said subject.
  • the agonists of the present invention may also be used to treat BEC® overdose.
  • an agonist of the present invention may be administered to bind to the RBP of the present invention and prevent or at least reduce BEC® binding.
  • the present invention also comprises a method of treating BEC® overdose in a subject, the method comprising administering an effective amount of an RBP agonist to the subject.
  • Agonists for use in this aspect of the invention may be varied and include RBP antibodies, rhamnose or some other RBP ligand.
  • compositions comprising an agonist of the present invention and a pharmaceutically acceptable carrier.
  • the compositions will further comprise an agent adapted to cause cell death such as a glycoside.
  • Pharmaceutical compositions of proteineous drugs of this invention are particularly useful for parenteral administration, i.e., subcutaneously, intramuscularly or intravenously.
  • the compositions for parenteral administration may comprise a solution of the compounds of the invention or a cocktail thereof dissolved in an acceptable carrier, preferably an aqueous carrier, an emulsion or formulated as micelles in an appropriate carrier.
  • aqueous carriers may be employed, e.g., water, buffered water, 0.4% saline, 0.3% glycine, and the like. These solutions are preferably sterile and generally free of particulate matter. These solutions may be sterilized by conventional, well known sterilization techniques.
  • the compositions may further contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents.
  • concentration of the compounds of the invention in such pharmaceutical formulation can very widely, i.e., from less than about 0.1%, usually at or at least about 1% to as much as 15 or 20% by weight and will be selected primarily based on fluid volumes, viscosities, etc., according to the particular mode of administration selected.
  • a pharmaceutical composition of the invention for intramuscular injection could be prepared to contain 1 mL sterile buffered water, and 50 mg of a compound of the invention.
  • a pharmaceutical composition of the invention for intravenous infusion could be made up to contain 250 ml of sterile Ringer's solution, and 150 mg of a compound of the invention.
  • Actual methods for preparing parenterally administrable compositions are well known or will be apparent to those skilled in the art and are described in more detail in, for example, Remington's Pharmaceutical Science, 15th ed., Mack Publishing Company, Easton, Pa.
  • the compounds described herein can be lyophilized for storage and reconstituted in a suitable carrier prior to use. This technique has been shown to be effective with conventional proteins and art-known lyophilization and reconstitution techniques can be employed.
  • the agonist in situations where the agonist is non-proteineous, it may be administered alone or in combination with pharmaceutically acceptable carriers.
  • pharmaceutically acceptable carriers The proportion of which is determined by the solubility and chemical nature of the compound, chosen route of administration and standard pharmaceutical practice.
  • they may be administered orally in the form of tablets or capsules containing such excipients as starch, milk sugar, certain types of clay and so forth.
  • They may be administered sublingually in the form of troches or lozenges in which the active ingredient is mixed with fillers and binders, flavouring agents and dyes; and then dehydrated sufficiently to make it suitable for pressing into a solid form.
  • They may be administered orally in the form of solutions that may be injected parenterally, that is, intramuscularly, intravenously or subcutaneously.
  • parenteral administration they may be used in the form of a sterile solution containing other solutes, for example, enough saline or glucose to make the solution isotonic.
  • the physician or veterinarian will determine the dosage of the present therapeutic agents that will be most suitable and it will vary with the form of administration and the particular compound chosen, and furthermore, it will vary with the particular subject under treatment.
  • the physician will generally wish to initiate treatment with small dosages substantially less than the optimum dose of the compound and increase the dosage by small increments until the optimum effect under the circumstances is reached. It will generally be found that when the composition is administered orally, larger quantities of the active agent will be required to produce the same effect as a smaller quantity given parenterally.
  • the compounds are useful in the same manner as other serotonergic agents and the dosage level is of the same order of magnitude as is generally employed with these other therapeutic agents.
  • the therapeutic dosage will generally be from 1 to 1000 milligrams per day and higher although it may be administered in several different dosage units. Tablets containing from 5 to 100 mg. of active agent are particularly useful.
  • compositions of the present invention may be adapted for topical application to a patient.
  • Topical formulations may be produced by dissolving or combining the agonist of the present invention in an aqueous or nonaqueous carrier.
  • aqueous or nonaqueous carrier any liquid, cream, or gel, or similar substance that does not appreciably react with the agonist or any other of the active ingredients that may be introduced into the composition and which are non-irritating are suitable.
  • Appropriate non-sprayable viscous, semi-solid or solid forms can also be employed that include a carrier compatible with topical application and have a dynamic viscosity preferably greater than water.
  • Suitable formulations are well known to those skilled in the art and include, but are not limited to, solutions, suspensions, emulsions, creams, gels, ointments, powders, liniments, salves, aerosols, transdermal patches, etc, which are, if desired, sterilized or mixed with auxiliary agents, e.g., preservatives, stabilizers, emulsifiers, wetting agents, fragrances, colouring agents, odour controllers, thickeners such as natural gums etc.
  • Particularly preferred topical formulations include ointments, creams or gels.
  • Ointments generally are prepared using either (1) an oleaginous base, i.e., one consisting of fixed oils or hydrocarbons, such as white petroleum or mineral oil, or (2) an absorbent base, i.e., one consisting of an anhydrous substance or substances which can absorb water, for example anhydrous lanolin.
  • an oleaginous base i.e., one consisting of fixed oils or hydrocarbons, such as white petroleum or mineral oil
  • an absorbent base i.e., one consisting of an anhydrous substance or substances which can absorb water, for example anhydrous lanolin.
  • the active ingredient is added to an amount affording the desired concentration.
  • Creams are oil/water emulsions. They consist of an oil phase (internal phase), comprising typically fixed oils, hydrocarbons and the like, waxes, petroleum, mineral oil and the like and an aqueous phase (continuous phase), comprising water and any water-soluble substances, such as added salts.
  • the two phases are stabilised by use of an emulsifying agent, for example, a surface active agent, such as sodium lauryl sulfite; hydrophilic colloids, such as acacia colloidal clays, veegum and the like.
  • an emulsifying agent for example, a surface active agent, such as sodium lauryl sulfite; hydrophilic colloids, such as acacia colloidal clays, veegum and the like.
  • Gels comprise a base selected from an oleaginous base, water, or an emulsion- suspension base.
  • a gelling agent that forms a matrix in the base, increasing its viscosity.
  • examples of gelling agents are hydroxypropyl cellulose, acrylic acid polymers and the like.
  • the agonist is added to the formulation at the desired concentration at a point preceding addition of the gelling agent.
  • the amount of compound incorporated into a topical formulation is not critical; the concentration should be within a range sufficient to permit ready application of the formulation to the affected tissue area in an amount that will deliver the desired amount of agonist to the desired treatment site.
  • the customary amount of a topical formulation to be applied to an affected tissue will depend upon an affected tissue size and concentration of the agonist in the formulation.
  • compositions of the invention are administered to a subject afflicted with cancer in an amount sufficient to at least improve the condition of the patient and preferably cure the patient of cancer.
  • compositions of the invention can be carried out with dose levels and pattern being selected by the treating physician or veterinarian.
  • the composition of the invention should provide a quantity of the compounds of the invention sufficient to effectively treat the cancer in the subject.
  • Antibodies that specifically recognize one or more epitopes of RBP, or epitopes of conserved variants of RBP, or peptide fragments of the RBP are also encompassed by the invention.
  • Such antibodies include but are not limited to polyclonal antibodies, monoclonal antibodies (mAbs), humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab').sub.2 fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above.
  • the antibodies of the invention may be used, for example, in the detection of the RBP in a biological sample and may, therefore, be utilized as part of a diagnostic or prognostic technique whereby patients may be tested for abnormal amounts of RBP.
  • Such antibodies may also be utilized in conjunction with, for example, compound screening schemes, as described herein for evaluating the effect of test compounds on the ability of RBP to bind its ligand. Additionally, such antibodies may be used to inhibit RBP activity that may be useful in various studies on the dynamics of the binding between the RBP and its ligand.
  • host animals may be immunized by injection with the RBP or an immunogenic portion thereof such as one corresponding to a functional domain of the RBP, e.g. the extracellular domain.
  • Host animals may include but are not limited to rabbits, mice, and rats, to name but a few.
  • adjuvants may be used to increase the immunological response, depending on the host species, including but not limited to Freund's (complete and incomplete), mineral gels such as aluminium hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, dinitrophenol, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and Corynebacterium parvum.
  • BCG Bacille Calmette-Guerin
  • Monoclonal antibodies may be obtained by any technique that provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique of Kohler and Milstein, (1975, Nature 256:495-497; and U.S. Pat. No. 4,376,110), the human B-cell hybridoma technique (Kosbor et al., 1983, Immunology Today 4:72; Cole et al.,
  • Such antibodies may be of any immunoglobulin class including IgG, IgM, IgE, IgA, IgD and any subclass thereof.
  • the hybridoma producing the monoclonals of this invention may be cultivated in vitro or in vivo. Production of high titres of monoclonals in vivo makes this the presently preferred method of production.
  • a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine monoclonal and a human immunoglobulin constant region.
  • Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide.
  • Antibody fragments that recognize specific epitopes may be generated by known techniques.
  • such fragments include but are not limited to: the F(ab').sub.2 fragments which can be produced by pepsin digestion of the antibody molecule and the Fab fragments which can be generated by reducing the disulfide bridges of the F(ab').sub.2 fragments.
  • Fab expression libraries may be constructed (Huse et al., 1989, Science, 246:1275-1281) to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity.
  • Antibodies to the RBP can, in turn, be utilized to generate anti-idiotype antibodies that "mimic" the RBP, using techniques well known to those skilled in the art.
  • antibodies that bind to the RBP and competitively inhibit the binding of rhamnose to the RBP can be used to generate anti-idiotypes that "mimic" the extracellular domain of the RBP and therefore bind rhamnose.
  • Example 1 Isolation of a rhamnose binding protein using affinity chromatography
  • Biotin Rhamnose-ITC (BRITC) was formed by dissolving Rhamnose-ITC (Sigma R6881 ; RMM 297.3) in DMSO, diluting it to 1mg/ml in 10mM sodium bicarbonate # pH 9.1 and then adding biotin hydrazide (Sigma, RMM 258.3) at 1 :1 or 5:1 molar ratio and allowing the reaction to proceed at room temperature for 16h.
  • Streptavidin sepharose conjugated columns (Amersham 17-5112-01) or free resin (17-5113-01) with a theoretical capacity for biotin labelled rhamnose (BRITC) of 60 ⁇ g/ml was used.
  • An excess amount of BRITC was dissolved in phosphate buffered saline (PBS) and circulated over the pre-equilibrated column at a flow rate of 0.2ml/min for 30min. Successful coupling of the BRITC was monitored by HPLC analysis of the BRITC-PBS solution.
  • PBS phosphate buffered saline
  • MSS Multiple Surfactant Solution
  • MSS comprises 5 M urea, 2 M thiourea, 0.002 M n-tributyl phosphine, 0.5% pH 3- 10
  • Pharmalyte carrier ampholytes Pharmacia, Uppsala [only in 2-D preparations], 2% 3-([3-cholamidopropyl]-dimethylammonio)-1-propanesulfonate (CHAPS), 2% caprylyl sulfo-betaine, and 0.001% Orange G dye.
  • CHAPS 3-([3-cholamidopropyl]-dimethylammonio)-1-propanesulfonate
  • Tris-HCI 4-20% polyacrylamide gradient gels (Bio-Rad) were used with electrode buffer Tris/glycine, pH 8.3.
  • Sample loading solution Tris pH 6.8, 0.1 % SDS, glycerol, dithiothreitol, bromophenol blue marker.
  • Electrophoresis conditions 100V for 90min.
  • Fluorescein Rhamnose-ITC was formed by reacting Rhamnose-ITC with fluorescein amine (Sigma F1148, RMM 347.3) at a molar ratio of 1:10 in 10mM sodium bicarbonate pH 9.1.
  • A2058 cells prepared in 40ml culture flasks were incubated with FRITC (5 or 10 ⁇ M) as described above and washed once with HBS 2+ .
  • Carbonyl di-imidazole (Aldrich 115533) was dissolved at 1M in DMSO immediately prior to use. This stock solution was then diluted in HBS 2+ or DMSO to 100 ⁇ M-10mM and added to the cells at room temperature. After 15min the cross-linker was removed and the flasks stored on ice. Cells were then removed from the flasks by scraping and taken up into MSS lysis buffer. Protein fractions were subjected to SDS-PAGE and the gels visualised as set out above.
  • the strip was then run in the second dimension using 1-D PAGE according to method 4 above, except a 10% polyacrylamide gel was used.
  • the gel was analysed using a fluoroimager and silver stained.
  • Figure 2A depicts the proteins cross-linked to fluorescein that were visualised by fluorescence scanning and Figure 2B depicts the total protein stained with Coomassie brilliant blue.
  • the total protein stain indicates that there are approximately equal amounts of protein loaded in each lane.
  • two fluorescently labelled proteins are detectable that are not present in the CDI only lanes.
  • Calibration of the gel using the molecular mass markers ( Figure 3) gives masses of 22kD and 68kD for these proteins. However, these masses include one or more FRITC molecules and consequently the mass of the receptor is approximately 67kD.
  • Example 3 Staining of cells with a fluorescein tagged rhamnose probe (FRITC)
  • FRITC fluorescein tagged rhamnose probe
  • FRITC at concentrations from 3-25 ⁇ M was incubated with A2058 cells for 15 minutes at 37°C.
  • the cells following incubation were found to fluoresce confirming the presence of a rhamnose binding protein on the cells.
  • An image of the cells is depicted in Figure 4 and closer inspection of the stained cells indicates an increased concentration of staining in the cell nucleus, suggesting the rhamnose probe is also taken inside the nuclear membrane. It was found that the staining could be inhibited by co-incubation of the FRITC and cells with free rhamnose at 10mM concentration.
  • cytotoxicities for different cell lines needed to be conducted using different sized cell populations, it was considered prudent to determine the effect, if any, of cell number on the measured value of LD 50 .
  • Five cell lines, HT-29, LS174-T, 5637, A431 and MCF-7 were evaluated at four different seeding cell densities.
  • Hs578T and CCD 18Lu were evaluated at three seeding densities and Hs578Bst, both early and late passage cells, were evaluated at two seeding densities.
  • Figure 10 shows that the single data points for the other cell lines evaluated are plotted, with the exception of MIA PaCa-2, the breast cancer lines and the early passage normal breast fibroblasts, all fall on the same exponential curve of Figure 6.
  • A2058 cells at two different cell densities were treated with BEC® +/- rhamnose and cell survival was monitored after 4 days.
  • the treatments comprised: (i) BEC® only for 4 days (ii) BEC® only for 5 minutes (iii) BEC® and 5mM rhamnose for 4 days and (iv) BEC® and 5 mM rhamnose for 5 minutes.
  • Figures 11 and 12 indicate that rhamnose competition with BEC® uptake is more readily observed at the higher cell density (5000 cells). Under these conditions, where the amount of BEC® available to each cell is a major factor determining LD 50, the presence of rhamnose at the relatively high concentration of 5 mM affects the amount of BEC® taken up by the cells in both 5 minutes and 4 days from solutions in specific concentration ranges. Data in Figure 12 suggests that the rhamnose protective effect is more significant in the pulsed treatment experiment.
  • A2058 cells were grown to 80-90% confluency in 25ml culture flasks and then washed with 2x HEPES buffered saline containing 140mM NaCI, 2mM MgCI 2 , and
  • FRITC solution was added to the cells and the flask incubated for 15min at 37°C.
  • the FRITC was removed, the cells washed once with HBS 2+ , and freshly prepared carbonyl di-imidazole (100 ⁇ M in 1ml DMSO) was added immediately at room temperature. After a minimum of 15min the cross-linker was removed and the flasks stored on ice.
  • Cell extracts are removed from the flask and added to a fresh tube.
  • the protein is precipitated by adding 1ml of methanol (or acetone) and storing the sample over night at -80°C.
  • methanol or acetone
  • the tube was centrifuged for 30min at 13000rpm and 4°C.
  • the methanol was removed and the sample resuspended in 300 ⁇ l of MSS.
  • To this 1.2 ml of hexane was added and the sample again centrifuged for 30min at 13000rpm and 4 ° C.
  • the top layer was discarded and any white particulate matter on the surface of the aqueous layer was also removed. Samples were then analysed by SDS-PAGE.
  • 100 ⁇ l of protein extract (section 1 above) was diluted to 10ml with 50mM Tris-CI pH7 and 0.05% Tween 20.
  • 50 ⁇ l of protein A sepharose [Amersham] was added and incubated overnight at 4°C on a rotating wheel. After incubation, the sample was centrifuged at 2000rpm for 5min to pellet the sepharose. The supernatant was added to 10 ⁇ l of anti-FITC antibody [Sigma], 50 ⁇ l of protein A sepharose and incubated overnight at 4°C on a rotating wheel.
  • FRITC fluorescein labelled rhamnose probe
  • the fluorescent complex appears to be associated with the cell debris/DNA that is precipitated during the initial methanol precipitation.
  • the protein-FRITC complex was diluted into a low detergent, low salt buffer and incubated with an antibody directed against fluorescein. Any complexes formed were absorbed onto protein A, precipitated and analysed by SDS-PAGE. The experiments produced a feint protein band at approx 70kD that was detectable by Coomassie blue staining Figure 14).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Cell Biology (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Urology & Nephrology (AREA)
  • Physics & Mathematics (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hospice & Palliative Care (AREA)
  • Biotechnology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Microbiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Food Science & Technology (AREA)
  • Public Health (AREA)
  • Analytical Chemistry (AREA)
  • Oncology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
PCT/AU2003/000135 2002-02-07 2003-02-07 Rhamnose binding protein WO2003066679A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
NZ534485A NZ534485A (en) 2002-02-07 2003-02-07 Rhamnose binding protein
AU2003202322A AU2003202322B2 (en) 2002-02-07 2003-02-07 Rhamnose binding protein
JP2003566050A JP2005538931A (ja) 2002-02-07 2003-02-07 ラムノース結合タンパク質
CA002475066A CA2475066A1 (en) 2002-02-07 2003-02-07 Rhamnose binding protein
EP03700718A EP1472287A4 (en) 2002-02-07 2003-02-07 PROTEIN FIXING RHAMNOSE
HK06100115.4A HK1080089A1 (zh) 2002-02-07 2006-01-04 鼠李糖結合蛋白

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AUPS0364A AUPS036402A0 (en) 2002-02-07 2002-02-07 Rhamnose binding protein
AUPS0364 2002-02-07

Publications (1)

Publication Number Publication Date
WO2003066679A1 true WO2003066679A1 (en) 2003-08-14

Family

ID=3833965

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2003/000135 WO2003066679A1 (en) 2002-02-07 2003-02-07 Rhamnose binding protein

Country Status (8)

Country Link
EP (1) EP1472287A4 (ja)
JP (1) JP2005538931A (ja)
CN (1) CN1639191A (ja)
AU (1) AUPS036402A0 (ja)
CA (1) CA2475066A1 (ja)
HK (1) HK1080089A1 (ja)
NZ (1) NZ534485A (ja)
WO (1) WO2003066679A1 (ja)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009040726A2 (en) * 2007-09-24 2009-04-02 Universite De Geneve Rhamnose antagonists and use thereof
US7998943B2 (en) 2005-07-06 2011-08-16 Btg International Limited Core 2 GlcNAc-T inhibitors III
US8197794B2 (en) 2003-12-22 2012-06-12 Ms Therapeutics Limited Core 2 GlcNAc-T inhibitors
US8609633B2 (en) 2005-07-06 2013-12-17 Ms Therapeutics Limited Core 2 GlcNAc-T inhibitors

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
AMINI H.R. ET AL: "Cryptic domains of a 60 kDa heat shock protein of helicobacter pylori bound to bovine lactoferrin", FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY, vol. 16, no. 3-4, 31 December 1996 (1996-12-31), pages 247 - 255, XP002903367 *
DAIKHORA T. ET AL: "Comparative studies of the agglutination of tumor cells and erythrocytes by plecoglossus altivelis (Ayu fish) roe lectin", PHYSICO-CHEMICAL BIOLOGY, vol. 37, no. 1, 1993, pages 31 - 40, XP008040630 *
GRILLON C., MONSIGNY M., KIEDA C.: "Soluble human lymphocyte sugar binding proteins with immunosuppressive activity", IMMUNOLOGY LETTERS, vol. 28, no. 1, April 1991 (1991-04-01), pages 47 - 55, XP002903364 *
KOMORI Y. ET AL: "Primary structure and biological activity of snake venom lectin (APL) from agkistrodon p. piscivorus (eastern cottonmouth)", TOXICON, vol. 37, no. 7, July 1999 (1999-07-01), pages 1053 - 1064, XP002903366 *
NITTA K. ET AL: "Comparative studies of carbohydrate-binding proteins from xenopus laevis skin and eggs. Sugar-binding specificities and affinity purification", CHEMICAL AND PHARMACEUTICAL BULLETIN, vol. 38, no. 4, April 1990 (1990-04-01), pages 975 - 981, XP002990409 *
See also references of EP1472287A4 *
TATENO H. ET AL: "Isolation and characterization of rhamnose-binding lectins from eggs of steelhead trout (oncorhyncus mykiss) homologous to low density lipoprotein receptor superfamily", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 273, no. 30, 24 July 1998 (1998-07-24), pages 19190 - 19197, XP002903368 *
TOBIN J.F., SCHLEIF R.F.: "Purification and properties of RhaR, the positive regulator of the L-rhamnose operons of escherichia coli", JOURNAL OF MOLECULAR BIOLOGY, vol. 211, no. 1, 5 January 1990 (1990-01-05), pages 75 - 89, XP002903374 *
WEEBADDA W.K. ET AL: "Avian air sac and plasma proteins that bind surface polysaccharides of escherichia coli 02", COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY, PART B, BIOCHEMISTRY AND MOLECULAR BIOLOGY, vol. 130, no. 3, October 2001 (2001-10-01), pages 299 - 312, XP002903365 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8197794B2 (en) 2003-12-22 2012-06-12 Ms Therapeutics Limited Core 2 GlcNAc-T inhibitors
US7998943B2 (en) 2005-07-06 2011-08-16 Btg International Limited Core 2 GlcNAc-T inhibitors III
US8609633B2 (en) 2005-07-06 2013-12-17 Ms Therapeutics Limited Core 2 GlcNAc-T inhibitors
WO2009040726A2 (en) * 2007-09-24 2009-04-02 Universite De Geneve Rhamnose antagonists and use thereof
WO2009040726A3 (en) * 2007-09-24 2009-09-24 Universite De Geneve Rhamnose antagonists and use thereof

Also Published As

Publication number Publication date
EP1472287A4 (en) 2005-10-05
CN1639191A (zh) 2005-07-13
CA2475066A1 (en) 2003-08-14
NZ534485A (en) 2006-09-29
AUPS036402A0 (en) 2002-02-28
HK1080089A1 (zh) 2006-04-21
JP2005538931A (ja) 2005-12-22
EP1472287A1 (en) 2004-11-03

Similar Documents

Publication Publication Date Title
EP1126028B1 (en) Novel g protein-coupled receptor proteins, dnas thereof and ligands to the same
JP4367866B2 (ja) 肺癌用のタンパク質マーカーおよびその使用
Del Vecchio et al. In vivo detection of multidrug-resistant (MDR1) phenotype by technetium-99m sestamibi scan in untreated breast cancer patients
Bradley et al. Increased expression of the epidermal growth factor receptor on human colon carcinoma cells
Guillaume et al. Antibodies for the immunochemistry of the human β3‐adrenergic receptor
JP3284133B2 (ja) ヒト乳脂肪小球膜ムチン抗原の糖タンパク質を確認する単クローン性抗体
US7348152B2 (en) Rhamnose binding protein
US20080038248A1 (en) Proteins Ligands For Nkg2d And Ul16 Receptors And Uses Thereof
EP1472287A1 (en) Rhamnose binding protein
WO1998039653A9 (en) Methods for producing pure perlecan and other heparan sulfate proteoglycans
AU2003202322B2 (en) Rhamnose binding protein
JPH10512158A (ja) 膀胱の核マトリックスタンパク質ならびに細胞増殖性疾患の検出及び治療におけるその使用
US6403325B1 (en) Methods and compounds for treating alzheimer's disease
US5741651A (en) Assays for identifiying compounds that bind to the gastrin releasing peptide receptor
JP2003530109A (ja) エルビンをコードする遺伝子及びその診断及び治療的使用
US20080187935A1 (en) Homo and Heterodimer Proteins of the Abcg Family, Methods For Detection and Screening Modulators Thereof
ES2378417T3 (es) KCNB: una proteína del canal de potasio
US6245522B1 (en) Master molecular rheostat switch for cell signaling
JP2003528599A (ja) Gタンパク質共役レセプター
WO2002059608A2 (en) Compositions and methods related to chemokine (c motif) xc receptor 1 (ccxcr1)
WO2012133994A1 (ko) Pauf 및 그의 결합 파트너를 상호작용을 이용한 암 치료제의 스크리닝 방법
US20020127226A1 (en) Class of RPTPases: their structural domains and ligands
Eisenblätter Identification and characterisation of a new multidrug resistance protein at the blood brain barrier
WO2001074297A2 (en) Methods and compositions utilizing an alternative splice variant of the sigma-1 receptor
JPWO2001062915A1 (ja) 新規蛋白質及びそれをコードする遺伝子

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

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

WWE Wipo information: entry into national phase

Ref document number: 2003202322

Country of ref document: AU

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

Ref document number: 534485

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2475066

Country of ref document: CA

Ref document number: 1726/CHENP/2004

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2003566050

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2003700718

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 20038055139

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2003700718

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 2003202322

Country of ref document: AU