US20060258605A1 - Nucleolin-mediated cancer diagnostics and therapy - Google Patents

Nucleolin-mediated cancer diagnostics and therapy Download PDF

Info

Publication number
US20060258605A1
US20060258605A1 US11/193,042 US19304205A US2006258605A1 US 20060258605 A1 US20060258605 A1 US 20060258605A1 US 19304205 A US19304205 A US 19304205A US 2006258605 A1 US2006258605 A1 US 2006258605A1
Authority
US
United States
Prior art keywords
nucleolin
cell
antibody
angiogenesis
endostatin
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
US11/193,042
Other languages
English (en)
Inventor
Yongzhang Luo
Hubing Shi
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.)
Tsinghua University
Protgen Ltd
Original Assignee
Yongzhang Luo
Hubing Shi
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 Yongzhang Luo, Hubing Shi filed Critical Yongzhang Luo
Publication of US20060258605A1 publication Critical patent/US20060258605A1/en
Assigned to TSINGHUA UNIVERSITY, PROTGEN LTD. reassignment TSINGHUA UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LUO, YONGZHANG, SHI, HUBING
Priority to US12/412,065 priority Critical patent/US11434288B2/en
Priority to US17/858,710 priority patent/US20230174644A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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
    • G01N33/57492Immunoassay; 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 involving compounds localized on the membrane of tumor or cancer cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell

Definitions

  • the present invention relates to a novel method of identifying cancer subjects, in particular human patients, who are suitable candidates for anti-angiogenesis cancer therapy.
  • the present invention also relates to a novel approach in searching and screening for angiogenesis inhibitors, molecules which are believed to be effective in reducing the malignant growth of cells, particularly in those cancers which are angiogenesis-dependent.
  • the present invention discloses methods of screening for inhibitors of angiogenesis using the molecule nucleolin.
  • the present invention relates to screening for angiogenesis inhibitors which functions in a manner that is analogous to the protein endostatin.
  • the invention is based upon the discovery that nucleolin is a specific receptor for endostatin, and is involved in the signal transduction pathway of endostatin when it functions as an angiogenesis inhibitor.
  • the effectiveness of cancer therapy can vary greatly among targeted patients depending upon a variety of factors, both external and internal.
  • External factors include the different stages of the cancer at the time of treatment, where early detection is key to effective treatment and recovery, the relevant strength of the cancer therapy, such as surgery, chemotherapy, or radiation therapy.
  • Internal factors include the health of the immune system of the patient, where a strong system can sustain a longer and stronger regiment of treatment, and thus helping the patient recover faster.
  • personalized medicine The notion that different individuals may have different tolerance and susceptibility to the same cancer drug or therapy has inspired a variety of approaches in the effort to increase the efficacy of a particular cancer therapy. Thus, due to individual variations, a drug that is effective on one patient may not be so on another.
  • endostatin has been hailed as an effective cancer therapy because it works to kill cancer cells by inhibiting angiogenesis, a process which is required by the cancer cells in order to metastasize. Every increase in the tumor cell population must be preceded by an increase in new capillaries that converge upon the tumor. This phenomenon is nearly universal; most of the human solid tumors and hematopoietic malignancies are angiogenesis dependent. Additional advantages to antiangiogenic therapy include low toxicity, minimal drug resistance, and repeated cycles of antiangiogenic therapy may be followed by a prolonged tumor dormancy without further therapy. See Boehm et al., Antiangiogenic therapy of experimental cancer does not induce acquired drug resistance. Nature (1997) 390: 404-407.
  • endostatin therapy is being applied to cancer patients across the board without referencing to each patient's susceptibility of the therapy.
  • Numerous clinical trials have been conducted in hope of finding an effective drug for anti-angiogenesis. It would be a significant progress if patients can be selected using objective criteria so they can be treated more effectively with endostatin.
  • This invention discloses methods and diagnostic kits directed to this endeavor.
  • the present invention provides a kit for determining the susceptibility of a subject to endostatin cancer therapy, comprising a label that labels nucleolin and a usage instruction for performing a screening of a sample of said subject with said label such as that an amount of nucleolin present in the sample is determined.
  • the subject is a mammal. More preferably, the subject is a human.
  • the nucleolin being labeled is cell surface nucleolin.
  • the label comprises an antibody that specifically binds to nucleolin, preferably a polyclonal antibody, and more preferably, a monoclonal antibody.
  • the label comprises a nucleic acid molecule, or probe, preferably a DNA probe, and still preferably, an RNA probe.
  • the present invention further provides a method of determining the likelihood of success of endostatin cancer therapy in a subject, comprising screening a sample from said subject for the level of expression of nucleolin, and determining if said subject is susceptible to endostatin cancer therapy based on the amount of nucleolin expression.
  • the present invention provides methods of screening for angiogenesis inhibitors, in particular, molecules which function in a similarly manner as endostatin.
  • the invention provides methods of using Nucleolin, referred to herein as “NL”, as a target molecule, applying conventional methodologies, candidate molecules can be examined so as to uncover those molecules that bind specifically to NL, at the same time, exhibiting angiogenesis activities. Due to the fact that NL is a specific cell surface receptor for endostatin, the new molecules discovered in the manner described above should function in a similarly manner as endostatin.
  • the present invention also provides methods of enhancing the sensitivity of target endothelial cells to endostatin.
  • the methods herein provide introducing exogenous NL molecules into the target endothelial cells, such that the NL molecules are over-expressed as compared to their wild type levels.
  • these target cells are those that normally do not express a high level NL endogenously.
  • the methods further provide the introduction of NL to targeted endothelial cells, such that these modified cells can be effectively killed by endostatin due to endostatin's angiogenesis properties.
  • the present invention also provides for antibodies against NL molecules, which can be used to detect target cancer cells having a high level of surface NL and as such are good candidate for ES cancer therapy.
  • the present invention provides a method of producing an NL-specific angiogenesis inhibitor effective in inhibiting angiogenesis, comprising: applying an appropriate binding assay to a pool of candidate molecules, thereby obtaining a plurality of NL-specific molecules; testing each of the plurality of NL-specific molecules for its effectiveness of inhibiting angiogenesis using an anti-angiogenesis assay; and selecting the resulting NL-specific molecule which is effective in inhibiting angiogenesis as demonstrated by the anti-angiogenesis assay.
  • the present invention provides a method of selecting an angiogenesis inhibitor having the ability to inhibit endothelial proliferation when added to proliferating endothelial cells in vitro, comprising the steps of: using a pharmaceutically acceptable method to discover molecules that specifically interact with NL as the target molecule; testing the molecules thus derived from the previous step for their effectiveness in inhibiting endothelial cell proliferation or migration; and harvesting the molecule thus derived which are effective in inhibition of endothelial cell proliferation or migration, wherein the effectiveness of the anti-angiogenesis function of said molecule is compared to that of Endostatin.
  • the present invention provides a method of increasing the receptiveness of a target cell to an angiogenesis inhibitor, comprising: introducing exogenous NL into the target cells, thereby obtaining a plurality of modified target cells expressing exogenous NL, and measuring the killing rate of the modified target cells by endostatin.
  • the present invention provides a method of enhancing the anti-angiogenesis effect of an angiogenesis inhibitor on a target endothelial cell, comprising introducing into said target cell a pharmaceutically effective amount of exogenous NL molecule, said NL molecule being able to express in said target cell; and incubating said target cell with said angiogenesis inhibitor, thereby causing the inhibition of the growth of said target cell.
  • the present invention provides a method of increasing the efficacy of angiogenesis inhibitors on controlling the growth of a cancer in a patient having such cancer, comprising identifying the presence of the level of endogenous NL molecules in a sample of the cancer of said patient, using acceptable methods; and determining the likelihood of efficacy of angiogenesis inhibitor on said cancer patient using the level of expression of NL in said patient, a higher level of NL indicating a higher degree of success of such angiogenesis inhibitor treatment.
  • the angiogenesis inhibitor is endostatin.
  • the present invention provides a diagnostic kit for assaying the individual sensitivity of target cells towards angiogenesis inhibitors, comprising a molecule that specifically bind to an NL molecule; and a pharmaceutically acceptable carrier.
  • the molecule is a polyclonal antibody, and more preferably, the antibody is a monoclonal antibody.
  • the present invention provides a method of identifying target cancer cells which are susceptible to an anti-angiogenesis inhibitor treatment, comprising generating an anti-nucleolin antibody; using said anti-nucleolin antibody to screen a sample of said target cancer cells; and identifying target cancer cells susceptible to anti-angiogenesis inhibitor treatment as indicated by the specific interactions between the target cancer cells and the anti-nucleolin antibody.
  • the antibody is a polyclonal antibody, and more preferably, the antibody is a monoclonal antibody.
  • the present invention provides a diagnostic kit for determining the target cancer cells which are susceptible to anti-angiogenesis inhibitor treatment, comprising an antibody against nucleolin and a pharmaceutically acceptable carrier.
  • the present invention provides a diagnostic kit for determining and selecting cancer subjects which are susceptible to anti-angiogenesis inhibitor treatment, comprising a label that is capable of binding to nucleolin and indicating its presence, and a written instruction for performing a screening test of a sample of the cancer subject such that the level of presence of nucleolin is detected in the sample.
  • FIG. 1 shows that HMEC (human microvascular endothelial cells) is a sensitive cell line in response to ES in migration and proliferation.
  • HMEC human microvascular endothelial cells
  • a Cell migration assay were performed with HMECs in the presence of ES at concentrations as indicated, PBS serves as a control.
  • FIG. 2 shows that ES binds to cell surface NL.
  • a ES binding proteins isolated from the cell surface of HMECs were identified as NL and its fragment. The ES binding proteins were isolated from HMECs' plasma membrane using ES-Ni-NTA affinity beads as described in methods. The fractions eluted with 500 mM sodium chloride in PBS buffer were applied to SDS-PAGE (left panel) and immunoblotting using monoclonal antibodies against NL (right panel).
  • b ES binds NL specifically in vitro. Immunoprecipitation was carried out using recombinant NL and ES.
  • c Heparin interrupts the formation of ES-NL complex.
  • HMECs were incubated with ES and different concentrations of antibodies against NL for 30 min at RT, and then were washed for 3 times with PBS buffer. The cells were applied to SDS-PAGE and immunoblotting using antibodies against ES. ⁇ -actin was used as control.
  • C HMECs were incubated with ES (20 ⁇ g/ml) for different periods of time at 37° C. and 5% CO 2 .
  • FIG. 3 shows that nucleolin is a receptor for endostatin.
  • a Indicated concentrations of recombinant NL were tested to lift the inhibition of ES in HUVECs migration assay, PBS serves as a control.
  • b Recombinant NL itself has no effect on cell migration.
  • HUVECs migration assay was performed with indicated concentrations of ES, NL, and both of them, respectively, PBS is a control.
  • c HUVECs proliferation assay was performed with indicated concentrations of ES, NL, and Anti-NL, PBS as a control. The cell number was evaluated by MTT assay.
  • d Cell adhesion assay was performed with NL-deficient and control HMECs.
  • NL-deficient cells were obtained by suppressing the expression of NL with RNAi plasmid BS/U6/1356. Control cells were transfected with blank plasmid pBS/U6. ES was added with indicated concentrations. The number of cells was evaluated by MTT assay.
  • FIG. 4 shows that nucleolin mediates endostatin signal network.
  • a-f ES is internalized by HMECs. HMECs were incubated with or without (a) 10 ⁇ g/ml biotinylated ES for 0.5 h (b), 1 h (c), 2 h (d), 3 h (e), and 7 h (f), respectively. The internalized ES was stained with TRITC-labeled avidin. g, The internalized ES was blocked by incubating cell with Anti-NL. Scale bar, 25 ⁇ m. h, In nuclei, ES inhibits CK2-mediated NL phosphorylation. Phosphorylation assay was performed as described in Methods.
  • Phosphorylated NL was detected by SDS-PAGE and autoradiography.
  • the NL blots serve as loading controls.
  • i-k Colocalization of cell surface NL with integrin ⁇ 1 on HMEC surface.
  • Intact HMECs were stained by mouse Anti-NL and rabbit Anti-integrin ⁇ 1, and detected by indirect immunofluorescence using laser scanning confocal microscopy. Scale bar, 10 ⁇ m.
  • FIG. 5 shows the distribution of cell surface nucleolin depends on cell growth states.
  • a-h Distribution of cell surface NL on HMECs.
  • Cell surface NL of proliferating cells and quiescent cells was detected by indirect immunofluorescence using rabbit Anti-NL.
  • DAPI indicates all cells in the field.
  • Scale bar 20 ⁇ m.
  • Cell cycle positions of proliferating cells (g) and quiescent cells (h) were detected by flow cytometry, respectively. Relatively quiescent cells were obtained by serum starvation for 24 h.
  • i-l Distribution of cell surface NL in tumor-bearing nude mice. Immunohistochemistry was performed as described in Methods. The blood vessels of heart (i), kidney (j), lung (k), and tumor (l) were indicated by arrows. Endothelial cells, which contain cell surface NL, were stained brown. Scale bar, 50 ⁇ m.
  • FIG. 6 provides a working model for nucleolin-mediated endostatin signal network.
  • a big complex which is composed of cell surface NL, integrin (such as integrin ⁇ 5 ⁇ 1), and other proteins, is involved in the ES signal network.
  • NL binds to myosin, which links NL and actin filaments.
  • integrin binds indirectly to bundles of actin filaments via the intracellular anchor proteins talin, ⁇ -actinin, filamin, and vinculin. No direct interaction can be observed between NL and integrin.
  • ES binds to this complex competing with ECM, which leads to the decrease of cell adhesion and migration. This binding can also trigger integrin-mediated signal transduction.
  • this complex can mediate ES internalization, in which myosin serves as a transporter. Subsequently, ES may be released into cytoplasm. Some ES serve as an inhibitor of NL which can stabilize the mRNA of Bcl-2, and exert other functions. The remaining ES is transported into nuclei, where it inhibits CK2-mediated NL phosphorylation as well as other down stream events.
  • FIG. 7 shows the co-localization between ES and NL in vivo.
  • Biotinylated ES and antibodies against NL were injected intravenously into mice bearing B16-F10 tumors.
  • Biotinylated ES and purified rabbit IgG was injected intravenously as a control.
  • the distribution of the biotin-labeled ES and anti-NL in heart (a-c), liver (d-f), kidney (g-i), tumor (j-l), and control tumor (m-o) were detected with both TRITC-conjugated avidin and FITC-conjugated secondary antibodies.
  • Scale bar 50 ⁇ m.
  • FIG. 8 shows the kinetic binding sensorgrams depicting real-time interactions between ES and NL at indicated concentrations.
  • nucleolin acts as a specific receptor for ES and facilitates its function in antiangiogenesis.
  • NL is a ubiquitous, nonhistone protein, which was first isolated from nucleolus. It is very interesting that the amount of NL is correlated to cell proliferation, which is regulated by Granzyme A and self-cleaving activity. Nucleolin also undergoes self-cleavage, which is decreased when cells enter a proliferative stage, as well as being cleaved by Granzyme A, an esterase secreted by cytotoxic lymphocytes (Chen et al., J. Biol. Chem., 1991, 266, 7754-7758; Fang and Yeh, Exp. Cell. Res., 1993, 208, 48-53; Pasternack et al., J. Biol. Chem., 1991, 266, 14703-14708). The cleavage and concomitant degradation of the protein provides for post-translational regulation of nucleolin.
  • NL exerts a critical and fundamental effect on cell proliferation, including organization of nucleolar chromatin, packaging of pre-RNA, rDNA transcription, and ribosome assembly. These activities are regulated by certain protein kinases such as casein kinase 2 (CK2) and cdc2 which are under strict control of other cell cycle proteins. Moreover, NL also functions as a cell surface receptor, shuttling between cell surface, cytoplasm, and nucleus. As a receptor of many viruses and cytokines, NL triggers the internalization of ligands as soon as these ligands bind to it.
  • CK2 casein kinase 2
  • cdc2 cdc2
  • Nucleolin has been described by Orrick et al (1973) as a protein with molecular weight about 100-110 kDa, and mainly existing in the nucleus of the propagating cells. Nucleolin exhibits auto-degradation and shows two degraded bands about 70 and 50 kDa in Western blotting analysis. Nucleolin is highly phosphorylated and methylated, and can be ADP-ribosylated. Because synthesis of the nucleolin is positively correlated with increased rate of cell division, tumor cells and rapidly dividing cells have higher levels of nucleolin content. The sequence of NL was reported earlier in Srivastava, et al., Cloning and sequencing of the human nucleolin cDNA. FEBS Lett. 250 (1), 99-105 (1989)
  • Nucleolin also known as P92 and C23
  • P92 and C23 is the most abundant nucleolar phosphoprotein in actively growing cells (Srivastava et al., FEBS Lett., 1989, 250, 99-105; Srivastava et al., J. Biol. Chem., 1990, 265, 14922-14931). It has been described by several groups and shown to participate primarily in ribosome biogenesis (Ghisolfi. et al., Mol. Biol. Rep., 1990, 14, 113-114; Sipos and Olson, Biochem. Biophys. Res.
  • Nucleolin contributes to ribosome biosynthesis by transiently binding to the pre-ribosomes in the nucleolus via a ribonucleoprotein consensus sequence (Bugler et al., J. Biol. Chem., 1987, 262, 10922-10925; Ghisolfi-Nieto et al., J. Mol. Biol., 1996, 260, 34-53; Sapp et al., Eur. J. Biochem., 1989, 179, 541-548).
  • nucleolin can represent up to 5% of the total nucleolar protein (Lapeyre et al., Proc. Natl. Acad. Sci. U.S.A., 1987, 84, 1472-1476; Sapp et al., Eur. J. Biochem., 1989, 179, 541-548). However, it has also been shown to be involved in cytokinesis, nucleogenesis, cell proliferation and growth, transcriptional repression, replication, signal transduction and chromatin decondensation reviewed in (Tuteja and Tuteja, Crit. Rev. Biochem. Mol. Biol., 1998, 33, 407-436).
  • the multifunctionality of this protein arises from the presence of distinct structural and functional domains within the protein (Creancier et al., Mol. Biol. Cell., 1993, 4, 1239-1250; Sapp et al., Eur. J. Biochem., 1989, 179, 541-548). Three domains have been described within the nucleolin protein, the N-terminal domain, the central domain and the C-terminal domain. Contained in the N-terminal domain are sequences that show homology with the high-mobility group (HMG) and are responsible for interactions with chromatin (Erard et al., Eur. J. Biochem., 1988, 175, 525-530).
  • HMG high-mobility group
  • the central domain contains four RNA recognition motifs and binds specifically with the short stem loop of the 18S and 28S ribosomal RNA (Bugler et al., J. Biol. Chem., 1987, 262, 10922-10925) while the C-terminal domain contains regions that are capable of unstacking bases in RNA (Ghisolfi et al., Mol. Biol. Rep., 1990, 14, 113-114; Ghisolfi-Nieto et al., J. Mol. Biol., 1996, 260, 34-53).
  • Nucleolin contains a bipartite nuclear localization signal, spanning both the N-terminal and central regions of the protein, which facilitates transport into the nucleus where nucleolin accumulates due to interactions with other proteins (Schmidt-Zachmann and Nigg, J. Cell Sci., 1993, 105, 799-806).
  • nucleolin The domain structure of nucleolin has led the protein to be classified as an Ag-NOR protein (Active ribosomal gene located in the Nucleolar Organizer Region) otherwise known as markers of active ribosomal genes (Roussel et al., Exp. Cell. Res., 1992, 203, 259-269). It has been shown that transcription of ribosomal genes requires the presence of Ag-NOR proteins and the expression of Ag-NOR proteins has been associated with the prediction of tumor growth rate in cancers.
  • Ag-NOR protein Active ribosomal gene located in the Nucleolar Organizer Region
  • Nucleolin has also been purified as a matrix attachment region (MAR) binding protein from human erythroleukemia cells. In these studies, nucleolin was shown to participate in the anchoring of chromatin loops to the nuclear matrix (Dickinson and Kohwi-Shigematsu, Mol. Cell. Biol., 1995, 15, 456-465).
  • MAR matrix attachment region
  • Nucleolin is highly phosphorylated and has been shown to be a substrate for casein kinase II (Csermely et al., J. Biol. Chem., 1993, 268, 9747-9752; Schneider and Issinger, Biochem. Biophys. Res. Commun., 1988, 156, 1390-1397), Protein kinase C-.xi. (Zhou et al., J. Biol. Chem., 1997, 272, 31130-31137), and Cdc2 (Belenguer et al., Mol. Cell. Biol., 1990, 10, 3607-3618). Furthermore, the phosphorylation of nucleolin has been shown to regulate the subcellular localization of the protein.
  • Nucleolin also undergoes self-cleavage, which is decreased when cells enter a proliferative stage, as well as being cleaved by Granzyme A, an esterase secreted by cytotoxic lymphocytes (Chen et al., J. Biol. Chem., 1991, 266, 7754-7758; Fang and Yeh, Exp. Cell. Res., 1993, 208, 48-53; Pastemack et al., J. Biol. Chem., 1991, 266, 14703-14708). The cleavage and concomitant degradation of the protein provides for post-translational regulation of nucleolin.
  • Anti-nucleolin antibodies have been found in the sera of patients with systemic connective tissue diseases including systemic lupus erythromatosus (SLE) (Minota et al., J. Immunol., 1990, 144, 1263-1269; Minota et al., J. Immunol., 1991, 146, 2249-2252) and scleroderma-like chronic graft vs. host disease (Bell et al., Br. J. Dermatol., 1996, 134, 848-854).
  • SLE systemic lupus erythromatosus
  • the pharmacological modulation of nucleolin expression may therefore be an appropriate point of therapeutic intervention in pathological conditions.
  • nucleolin correlates with cell proliferation rate. Nucleolin levels are highest in tumors and moderate in other rapidly dividing cells. It can be used in studies of different cancer cell lines as useful marker for cell proliferation. Since nucleolin plays a vital role in tumor cell proliferation, the present invention provides a strategy of inhibition of nucleolin expression to suppress the growth rate of tumor cells.
  • Endostatin (referred to herein as “ES”) is a 20 kDa C-terminal globular domain of the collagen-like protein, collagen XVIII. It was originally isolated from the supernatant of a cultured murine hemangioendothelioma cell line for its ability to inhibit the proliferation of capillary endothelial cells. In animal tests, tumor dormancy was induced following repeated cycles of ES treatment without any drug resistance. Moreover, low toxicity of ES was observed in both animal tests and clinical trials. ES exhibits potent activities in inhibiting endothelial cell proliferation, migration, adhesion, survival, and in inducing cell apoptosis.
  • nucleolin is a specific receptor to which endostatin binds, and mediates endostatin's function as an angiogenesis inhibitor provides a basis to screen and generate additional small molecules which also can function as angiogenesis inhibitor, and yet possesses additional properties that are more readily adapted as compared to the protein endostatin.
  • endostatin in order for endostatin to function effectively as an angiogenesis inhibitor, a large quantity of endostatin must be produced and administered into the cancer subjects, such as a mammal or a human, in order to produce the desired anticancer effect.
  • the production of endostatin can be prohibitively expensive due to the demand of high dose in cancer therapy using endostatin.
  • angiogenesis inhibitors other than endostatin, which can be used effectively and economically produced in reducing cancer growth.
  • exogenous NL is introduced into target cells so that the cells will express higher than normal amount of surface NL.
  • modified target cells are more sensitive to the action of ES molecules due to the presence of high level of NL molecules, which as described above, act as the specific receptor for ES.
  • antibodies against NL can be used to screen those cancer cells or endothelial cells which express a high level of surface NL. Finding this particular group of patients is extremely beneficial to the effectiveness of angiogenesis-related cancer therapy, since patients with high expression of surface NL are ideal for the administration of ES in order to inhibit their tumor growth.
  • endostatin refers to a protein that is preferably 18 kDa to 21 kDa in size as determined by non-reduced and reduced gel electrophoresis, respectively.
  • the term endostatin also includes precursor forms of the 18 kDa to 20 kDa protein.
  • the term endostatin also includes fragments of the 18 kDa to 20 kDa protein and modified proteins and peptides that have a substantially similar amino acid sequence, and which are capable of inhibiting proliferation of endothelial cells.
  • endostatin includes shortened proteins or peptides wherein one or more amino acids is removed from either or both ends of endostatin, or from an internal region of the protein, yet the resulting molecule retains endothelial proliferation inhibiting activity.
  • endostatin also includes lengthened proteins or peptides wherein one or more amino acid is added to either or both ends of endostatin, or to an internal location in the protein, yet the resulting molecule retains endothelial proliferation inhibiting activity.
  • Such molecules for example with tyrosine added in the first position are useful for labeling such as radioiodination with 125 I iodine for use in assays. Labeling with other radioisotopes may be useful in providing a molecular tool for destroying the target cell containing endostatin receptors.
  • nucleolin refers to a protein that is preferably 100 kDa (The exact MW of 80 kDa without post-modification) in size as determined by reduced gel electrophoresis.
  • nucleolin also includes precursor forms of the 100 kDa protein.
  • nucleolin also includes fragments of 100 kDa protein and modified proteins and peptides that have a substantially similar amino acid sequence, and which are capable of inhibiting proliferation of endothelial cells.
  • silent substitutions of amino acids wherein the replacement of an amino acid with a structurally or chemically similar amino acid does not significantly alter the structure, conformation or activity of the protein, is well known in the art. Such silent substitutions are intended to fall within the scope of the appended claims.
  • nucleolin includes shortened proteins or peptides wherein one or more amino acids is removed from either or both ends of nucleolin, or from an internal region of the protein, yet the resulting molecule retains its specific binding activity for ES.
  • nucleolin also includes lengthened proteins or peptides wherein one or more amino acid is added to either or both ends of nucleolin, or to an internal location in the protein, yet the resulting molecule retains the specific ES related NL activity.
  • Such molecules for example with tyrosine added in the first position are useful for labeling such as radioiodination with 125 I iodine for use in assays.
  • NL-specific refers to the ability of NL binding to an angiogenesis inhibitor, and mediates its inhibition activity.
  • angiogenesis-dependent refers to those cancers and tumors whose growth or migration require angiogenesis, including those that would require for their growth in either volume or mass, or both, an increase in the number and density of the blood vessels supplying them with blood.
  • the term “subject” refers to any animal, such as a mammal, including but not limited to a human, a non-human primate, a rodent, a pig, a rabbit, and the like, which is to receive a particular treatment, or undergoing a particular procedure such as screening for the level of presence of a particular molecule.
  • sample is used in its broadest sense, including, but not limited to a biological sample and environmental sample. In one sense, it is meant to include a specimen or culture obtained from any source, as well as biological and environmental samples. Biological samples may be obtained from animals (including humans) and encompass fluids, solids, tissues, and gases. Biological samples include blood products, such as plasma, serum and the like. Environmental samples include environmental material such as surface matter, soil, water, minerals, crystals and industrial samples. Such examples are not to be construed as limiting the sample types encompassed by the present invention.
  • label encompasses chemical or biological molecules that are used in detecting the presence in a sample of a target molecule which is capable of binding to or otherwise interact with the label so as to indicate its presence in the sample, and the amount of the target molecule in the sample.
  • labels include, but not limited to, a nucleic acid probe such as a DNA probe, or RNA probe, an antibody, a radioisotope, a fluorescent dye, and the like.
  • usage instruction includes instructions in the kit for carrying out the procedure for detecting the presence of a target molecular such as nucleolin in the sample to be tested.
  • the usage instruction comprising the statement of intended use required by the U.S. Food and Drug Administration (FDA) in labeling in vitro diagnostic products. It would be apparent to one with ordinary skill in the art of medical diagnostic devices as to the format and content of these usage instructions as required by the FDA.
  • an appropriate binding assay for selecting specific NL-related angiogenesis inhibitor includes HPLC, immunoprecipitation, fluorescent-binding assay, capillary electrophoresis, and so forth.
  • an “anti-angiogenesis assay” is an experiment where a pool of candidate molecules are screened in order to discover the effectiveness of the candidate molecules in inhibiting angiogenesis.
  • various methods can be applied to carry out the present invention. For example, proteins and peptides derived from these and other sources, including manual or automated protein synthesis, may be quickly and easily tested for endothelial proliferation inhibiting activity using a biological activity assay such as the bovine capillary endothelial cell proliferation assay.
  • bioassays for inhibiting activity include the chick embryonic chorioallantoic membrane (CAM) assay, the mouse corneal assay, and the effect of administering isolated or synthesized proteins on implanted tumors.
  • the chick CAM assay is described by O'Reilly, et al. in “Angiogenic Regulation of Metastatic Growth”, Cell, vol. 79 (2), Oct. 21, 1994, pp. 315-328, which is hereby incorporated by reference in its entirety.
  • Additional anti-angiogenesis assays for screening for angiogenesis inhibitors can be found in Yu, et al., PNAS, Vol. 101, No. 21, pp 8005-8010 (2004), which is hereby incorporated by reference in its entirety.
  • proliferation and/or migration refers to two situations: 1) both proliferation and migration of endothelial cells are modulated; 2) either proliferation or migration, but not both, of endothelial cells are modulated.
  • link refers to connecting antibody to a cytotoxic agent such as a cytokine molecule, using conventional, well-known biological or chemical techniques such as cross-linking, and so forth.
  • the NL molecules can be used to generate polyclonal or monoclonal antibodies, which can in turn be used to both identify and quantify the level of NL in a particular target cells.
  • NL labeled with appropriate markers such as radioisotopes and fluorescent dye can be used to for the detection of endostatin in body fluids and tissues for the purpose of diagnosis or prognosis of angiogenesis-mediated diseases such as cancer.
  • the present invention also includes methods of treating or preventing angiogenic diseases and processes including, but not limited to, arthritis and tumors by increasing the efficacy of endostatin in those angiogenesis-dependent cancers in a patient.
  • methods such as flow cytometry as well as Enzyme-linked Immunosorbent Assay (ELISA) techniques are used for quantification of the NL peptide.
  • ELISA Enzyme-linked Immunosorbent Assay
  • RNA molecules of NL can be performed using standard molecular biological techniques such as DNA probe hybridization, PCR, etc. General references for methods that can be used to perform the various PCR and cloning procedures described herein can be found in Molecular Cloning: A Laboratory Manual (Sambrook et al., eds. Cold Spring Harbor Lab Publ. 1989, latest edition). Detection of the RNA molecules of NL can be performed using Northern blot analysis. Northern blot analysis involves the separation of RNA and hybridization of a complementary labeled probe. In certain embodiments, RNA (or corresponding cDNA) is detected by hybridization to an oligonucleotide probe.
  • the TaqMan assay (PE Biosystems, Foster City, Calif.; See e.g., U.S. Pat. Nos. 5,962,233 and 5,538,848, each of which is herein incorporated by reference) is utilized.
  • the assay is performed during a PCR reaction.
  • the TaqMan assay exploits the 5′-3′ exonuclease activity of the AMPLITAQ GOLD DNA polymerase.
  • a probe consisting of an oligonucleotide with a 5′-reporter dye (e.g., a fluorescent dye) and a 3′-quencher dye is included in the PCR reaction.
  • the 5′-3′ nucleolytic activity of the AMPLITAQ GOLD polymerase cleaves the probe between the reporter and the quencher dye.
  • the separation of the reporter dye from the quencher dye results in an increase of fluorescence.
  • the signal accumulates with each cycle of PCR and can be monitored with a fluorimeter.
  • RNA reverse-transcriptase PCR
  • RNA is enzymatically converted to complementary DNA or “cDNA” using a reverse transcriptase enzyme.
  • the cDNA is then used as a template for a PCR reaction.
  • PCR products can be detected by any suitable method, including but not limited to, gel electrophoresis and staining with a DNA specific stain or hybridization to a labeled probe.
  • the quantitative reverse transcriptase PCR with standardized mixtures of competitive templates method described in U.S. Pat. Nos. 5,639,606, 5,643,765, and 5,876,978 (each of which is herein incorporated by reference) is utilized.
  • Detection of the protein molecule of NL can be performed using techniques known in the art (e.g., radioimmunoassay, ELISA (enzyme-linked immunosorbant assay), “sandwich” immunoassays, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays (e.g., using colloidal gold, enzyme or radioisotope labels, for example), Western blots, precipitation reactions, agglutination assays (e.g., gel agglutination assays, hemagglutination assays, etc.), complement fixation assays, immunofluorescence assays, protein A assays, and immunoelectrophoresis assays, etc.
  • radioimmunoassay e.g., ELISA (enzyme-linked immunosorbant assay), “sandwich” immunoassays, immunoradiometric assays, gel diffusion precipitation
  • antibody binding is detected by detecting a label on the primary antibody.
  • the primary antibody is detected by detecting binding of a secondary antibody or reagent to the primary antibody.
  • the secondary antibody is labeled. Many methods are known in the art for detecting binding in an immunoassay and are within the scope of the present invention.
  • an automated detection assay is utilized.
  • Methods for the automation of immunoassays include those described in U.S. Pat. Nos. 5,885,530, 4,981,785, 6,159,750, and 5,358,691, each of which is herein incorporated by reference.
  • the analysis and presentation of results is also automated.
  • software that generates a prognosis based on the presence or absence of a series of proteins corresponding to cancer markers is utilized.
  • Antibodies specific for NL and NL analogs are made according to techniques and protocols well known in the art.
  • the antibodies may be either polyclonal or monoclonal.
  • the antibodies are utilized in well-known immunoassay formats, such as competitive and non-competitive immunoassays, including ELISA, sandwich immunoassays and radioimmunoassays (RIAs), to determine the presence or absence of the endothelial proliferation inhibitors of the present invention in body fluids.
  • body fluids include but are not limited to blood, serum, peritoneal fluid, pleural fluid, cerebrospinal fluid, uterine fluid, saliva, and mucus.
  • the present invention provides isolated antibodies that can be used in the diagnostic kits in the detection of NL.
  • the present invention provides monoclonal antibodies that specifically bind to NL.
  • An antibody against NL in the present invention may be any monoclonal or polyclonal antibody, as long as it can recognize the protein.
  • Antibodies can be produced by using NL or its analogue as the antigen using conventional antibody or antiserum preparation processes.
  • the present invention contemplates the use of both monoclonal and polyclonal antibodies. Any suitable method may be used to generate the antibodies used in the methods and compositions of the present invention, including but not limited to, those disclosed herein.
  • a monoclonal antibody protein, as such, or together with a suitable carrier or diluent is administered to an animal (e.g., a mammal) under conditions that permit the production of antibodies.
  • complete or incomplete Freund's adjuvant may be administered.
  • the protein is administered once every 2 weeks to 6 weeks, in total, about 2 times to about 10 times.
  • Animals suitable for use in such methods include, but are not limited to, primates, rabbits, dogs, guinea pigs, mice, rats, sheep, goats, etc.
  • an individual animal whose antibody titer has been confirmed e.g., a mouse
  • 2 days to 5 days after the final immunization, its spleen or lymph node is harvested and antibody-producing cells contained therein are fused with myeloma cells to prepare the desired monoclonal antibody producer hybridoma.
  • Measurement of the antibody titer in antiserum can be carried out, for example, by reacting the labeled protein, as described hereinafter with the antiserum and then measuring the activity of the labeling agent bound to the antibody.
  • the cell fusion can be carried out according to known methods, for example, the method described by Koehler and Milstein (Nature 256:495 [1975]).
  • a fusion promoter for example, Sendai virus (HVJ) or, preferably, polyethylene glycol (PEG), is used.
  • Polyclonal antibodies may be prepared by any known method or modifications of these methods including obtaining antibodies from patients. For example, a complex of an immunogen (an antigen against the protein) and a carrier protein is prepared and an animal is immunized by the complex according to the same manner as that described with respect to the above monoclonal antibody preparation. A material containing the antibody against is recovered from the immunized animal and the antibody is separated and purified.
  • an immunogen an antigen against the protein
  • a carrier protein is prepared and an animal is immunized by the complex according to the same manner as that described with respect to the above monoclonal antibody preparation.
  • a material containing the antibody against is recovered from the immunized animal and the antibody is separated and purified.
  • the present invention provides for a method of inhibiting the proliferation and/or migration of endothelial cells by producing a combination antibody containing an anti-NL antibody linked to a cytotoxic agent such as a chemokine, i.e. a Tumor Necrosis Factor-alpha, etc.
  • a combination antibody containing an anti-NL antibody linked to a cytotoxic agent such as a chemokine, i.e. a Tumor Necrosis Factor-alpha, etc.
  • a combination antibody When such a combination antibody is administered to a cell sample including an endothelial cells, the anti-NL antibody will direct the toxic agent to the endothelial cells, and thus bring the toxic agent such as Tumor Necrosis Factor-alpha to act upon the endothelial cells, and killing the cell growth.
  • First-generation immunotoxins were constructed by coupling toxins to MAb or antibody fragments using a heterobifunctional cross-linking agent. It was also discovered that genetic engineering could be used to replace the cell-binding domains of bacterial toxins with the Fv portions of antibodies or with growth factors.
  • cytokines are small protein molecules that are the core of communication between immune system cells, and even between these cells and cells belonging to other tissue types. They are actively secreted by immune cells as well as other cell types. Cytokines that are produced by immune cells form a subset known as lymphokines. Their action is often local, but sometimes can have effects on the whole body.
  • cytokines that have both stimulating and suppressing action on lymphocyte cells and immune response.
  • Some of the better known cytokines include: histamine, prostaglandin, TNF- ⁇ , IL-1, and IL-6.
  • kits for the detection and characterization of nucleolin in cancer diagnostics contain antibodies specific for NL, in addition to detection reagents and buffers.
  • the kits contain reagents specific for the detection of mRNA or cDNA (e.g., oligonucleotide probes or primers) of NL.
  • the kits contain all of the components necessary to perform a detection assay, including all controls, directions for performing assays, and any necessary software for analysis and presentation of results.
  • Kits containing labels such as antibodies again NL for measurement of NL are also contemplated as part of the present invention.
  • Antibody solution is prepared such that it can detect the presence of NL peptides in extracts of plasma, urine, tissues, and in cell culture media are further examined to establish easy to use kits for rapid, reliable, sensitive, and specific measurement and localization of endostatin.
  • assay kits include but are not limited to the following techniques; competitive and non-competitive assays, radioimmunoassay, bioluminescence and chemiluminescence assays, fluorometric assays, sandwich assays, immunoradiometric assays, dot blots, enzyme linked assays including ELISA, microtiter plates, antibody coated strips or dipsticks for rapid monitoring of urine or blood, and immunocytochemistry.
  • competitive and non-competitive assays radioimmunoassay, bioluminescence and chemiluminescence assays, fluorometric assays, sandwich assays, immunoradiometric assays, dot blots, enzyme linked assays including ELISA, microtiter plates, antibody coated strips or dipsticks for rapid monitoring of urine or blood, and immunocytochemistry.
  • a diagnostic kit in the present invention can be used for localization of endostatin in tissues and cells.
  • This NL immunohistochemistry kit provides instructions, NL molecules, preferably labeled and linked to a fluorescent molecule such as fluorescein isothiocyanate, or to some other reagent used to visualize the primary antiserum. Immunohistochemistry techniques are well known to those skilled in the art.
  • This NL immunohistochemistry kit permits localization of endostatin in tissue sections and cultured cells using both light and electron microscopy. It is used for both research and clinical purposes. For example, tumors are biopsied or collected and tissue sections cut with a microtome to examine sites of endostatin production. Such information is useful for diagnostic and possibly therapeutic purposes in the detection and treatment of cancer.
  • Nucleolin is an Endostatin-Binding Protein
  • ES-binding proteins directly from the membrane proteins of human microvascular endothelial cells (HMECs) with immobilized ES.
  • Nucleolin was identified as a critical member in the ES signaling network, and is the most interesting one in the network.
  • NL serves as a novel receptor for ES, and mediates the activity of ES in antiangiogenesis.
  • Endothelial cells (HMECs or HUVECs, 2 ⁇ 10 4 per well) were seeded into the upper chamber of TranswellTM filter (8 ⁇ m pores, Costar) with DMEM medium containing 0.5% FCS and 10 ng/ml VEGF (PeproTech EC).
  • ES from Protgen
  • NL and Anti-NL were added in both upper chamber and lower chamber at the beginning of the migration assay.
  • the endothelia cells were allowed to migrate for 6 h at 37° C. and 5% CO 2 . After fixed with ethanol and stained with Eosin, the cells migrated completely through the filter to the lower chamber were calculated as the average number of cells observed in five random high-power ( ⁇ 400) fields per well in triplicate wells.
  • Endothelia cells (HMECs or HUVECs, 1 ⁇ 10 3 per well) were seeded in a 96-well plate with DMEM medium containing 0.5% FCS and 10 ng/ml bFGF. ES with indicated concentrations and other reagents were added at the beginning of the proliferation assay with a final volume of 200 ⁇ l.
  • the endothelial cells were allowed to proliferate for 48 h at 37° C. and 5% CO 2 . After 48 h incubation, the medium was replaced with DMEM medium without phenol red and 0.5 mg/ml MTT with a final volume of 100 ⁇ l. The cells were incubated further for 4 h at 37° C. and 5% CO 2 . Subsequently, the cells were lysed with iso-propanol containing 0.05 M hydrochloride, and the absorbance at 570 nm was measured.
  • HMECs were incubated with ES (20 ⁇ g/ml) for 1 h at 37° C. and 5% CO 2 . Without permeabilized, the HMECs were stained with indicated antibodies. FITC-linked goat anti-mouse IgG and TRITC-linked goat anti-rabbit IgG were used as the secondary antibodies. Confocal fluorescence imaging was performed on an Olympus Fluoview laser scanning confocal imaging system (Olympus Inc.). Images were captured using multiple photomultiplier tubes regulated by Fluoview 2.0 software (Olympus).
  • the cDNA of NL was obtained from HMECs by RNA isolation and reverse transcription system (Promega).
  • the sequence of NL fusing with a polyhistidine (His) 6 tag was subcloned into pPIC9K (Invitrogen).
  • This plasmid was linearized with restriction enzyme Sal I (Promega) and electrotransformed into pichia strain GS115.
  • a stable transformant was selected using G418 (Invitrogen) and then grown in a 30° C. shaking flask for 3 days in BMMY medium as described by the manufacturer. The supernatant was obtained and NL was purified with Ni-NTA nickel ion-affinity columns (Qiagen).
  • New Zealand White rabbits were immunized with 50 ⁇ g of recombinant NL prepared by pPIC9K-GS115 pichia expression system as described previously.
  • the initial subcutaneous injection was administered in Freund's complete adjuvant, and a booster dose of 50 ⁇ g was given intramuscularly on day 14 in Freund's incomplete adjuvant.
  • Boosts of 50 ⁇ g of NL were given subcutaneously without any adjuvant at 4, 10, 16, and 22 weeks.
  • Cells to be tested were serum-starved for 30 min, and then seeded in a 96-well plate which is coated with endostatin (20 ⁇ g/ml) and polylysine (50 ⁇ g/ml). The plate was incubated for 1 h at 37° C. and 5% CO 2 . Unbounded cells were removed by washing with fresh medium gently. The remained cells were stained with crystal violet (0.1% in ddH 2 O) for 25 min at RT. The plated was washed with tape water, and the remained crystal violet was solubilized with 0.5% Triton X-100 (diluted in ddH 2 O). The absorbance at 570 nm was measured.
  • HMEC cell line was chosen that meets the criteria as a good model for antiangiogenesis assay.
  • ES inhibited the migration of HMECs stimulated by VEGF in a dose dependent manner and the half-maximal inhibitory concentration (IC50) was observed at 4 ⁇ g/ml ( FIG. 1 a ).
  • IC50 half-maximal inhibitory concentration
  • HMECs may contain potential receptor(s) for ES.
  • ES can specifically bind HMECs under physiological conditions (see FIG. 2 d below).
  • Recombinant ES was pre-loaded on to Ni-NTA beads via its N-terminal His-tag.
  • Crude fraction of plasma membrane of HMECs was prepared and treated with 1% Triton X-100 to release proteins from plasma membrane as described by Marshak et al.
  • the prepared plasma membrane proteins were applied to the ES-Ni-NTA beads. Unbound proteins were removed from ES-Ni-NTA beads with PBS buffer.
  • IP immunoprecipitation
  • ES and total endogenous NL form a complex in living cells.
  • co-localization between ES and NL on cell surface of HMECs was also observed by laser scanning confocal immunofluorescence microscopy ( FIG. 2 f - i ).
  • ES specifically binds to NL both in vitro and in vivo, which indicates that NL is a potential receptor for ES.
  • Nucleolin is a Novel Receptor for Endostatin
  • NL is a receptor for ES, it should mediate the activities of ES in antiangiogenesis such as inhibiting the migration, proliferation, and adhesion of endothelial cells.
  • competitive cell migration and proliferation assays were therefore performed with ES, recombinant NL, and polyclonal antibodies against NL, respectively. Since NL was isolated from HMECs as a potential receptor for ES in antiangiogenesis, whether or not NL plays similar roles in other widely accepted endothelial cells should be demonstrated.
  • human umbilical vein endothelial cell (HUVEC), isolated directly from umbilical veins, were subjected to competitive cell migration and proliferation assays.
  • RNA interference RNA interference
  • the amount of internalized ES reached the maximum at about 2 h of incubation ( FIG. 4 d ). By 3 h, the nucleus-accumulated ES began to disappear ( FIG. 4 e ). By 7 h, little ES could be found in nuclei ( FIG. 4 f ). There was a lag phase in the fluorescence assay ( FIG. 4 b - d ) comparing with IP ( FIG. 2 e ) for the amount of internalized ES to reach their maximals, probably the degradation rate of ES is faster than that of biotinylated ES.
  • Myosins constitute a huge superfamily, which is involved in membrane dynamics and actin organization at cell cortex, thus affecting cell migration, adhesion, and endocytosis.
  • two-head class-V myosins can serve as transporters for vesicles, organelles, and mRNA particles along actin filaments.
  • cell surface NL can bind ES and transport ES into cell nuclei ( FIG. 4 a - f ). Since myosin is an intracellular protein, it must bind to the intracellular domain of cell surface NL. We speculate that this NL-myosin complex may serve as a transporter during the process of ES internalization. Similar process was also reported by Shibata et al.
  • midkine a growth factor in neural cells
  • NL a growth factor in neural cells
  • antibodies against the N-terminus of NL can be internalized by Hep-2 cells via cell surface NL. Therefore, internalization appears to be a general phenomenon and is inevitable when a ligand binds to cell surface NL. The difference is that ES can inhibit cell proliferation whereas antibodies against NL cannot ( FIG. 3 c ).
  • phosphorylation of NL is stimulated by bFGF rather than VEGF ( FIG. 4 h ), which may provide an explanation for previous reports that bFGF can stimulate proliferation of endothelial cells, whereas VEGF can stimulate migration of endothelial cells.
  • bFGF can stimulate proliferation of endothelial cells
  • VEGF can stimulate migration of endothelial cells.
  • Our observations demonstrate that ES inhibits bFGF-stimulated phosphorylation of NL which is mediated by CK2, consequently, inhibits cell survival and proliferation.
  • NL is a novel receptor for ES in inhibiting cell migration, proliferation, and adhesion. Since integrin was also reported to be a receptor for ES, it is interesting to test whether there is an interaction between NL and integrin.
  • integrin ⁇ 5 ⁇ 1 was reported to be the receptor for ES by Rehn et al. and Sudhakar et al., therefore, colocalization between cell surface NL and integrin ⁇ 5 ⁇ 1 was performed by indirect immunofluorescence using the mouse anti-integrin ⁇ 1 and the rabbit anti-NL. Certain overlap between cell surface NL and integrin ⁇ 1 was observed by laser scanning confocal microscopy ( FIG.
  • ES may interfere with cell motility and adhesion mediated by NL-myosin complex. It is plausible that cell surface NL and integrin ⁇ 5 ⁇ 1 along with other proteins such as myosin form a huge complex which together serves as an integrated receptor for ES (see FIG. 6 ).
  • the Distribution of NL Provides a Basis for the Low Toxicity of Endostatin
  • Recombinant nucleolin modulates the function of endostatin in cell migration assay
  • the cDNA of NL was obtained from human microvascular endothelial cells (HMECs) using the SV total RNA isolation system and reverse transcription system (Promega) following the manufacture's protocol.
  • the sequence of NL fusing with polyhistidine (His) 6 tag was amplified by PCR and then subcloned into pPIC9K (Invitrogen). Following the manufacture's protocol, this plasmid was linearized with restriction enzyme Sal I (Promega) and electrotransformed into yeast strain GS115. A stable transformant was selected using G418 and then grown in a 30° C.
  • BMMY medium (10 g/L yeast extract; 20 g/L peptone; 100 mmol/L potassium phosphate, pH 6.0; 13.4 g/L yeast nitrogen base; 40 mg/L biotin; and daily additions of methanol to the medium to yield a final concentration of 0.5%).
  • the supernatant was obtained and the NL was purified on Ni-NTA nickel ion-affinity columns (Qiagen).
  • Qiagen Ni-NTA nickel ion-affinity columns
  • Polyclonal antibodies against nucleolin were prepared with this protein.
  • Human umbilical vein endothelial cell (2 ⁇ 10 4 per well) were seeded into the upper chamber of Transwell filter (8 ⁇ m pores, Costar) with DMEM medium, 0.5% FCS, and 10 ng/ml bFGF (PeproTech EC).
  • ES (5 ⁇ g/ml, Protgen) and rh-nucleolin (20 ⁇ g/ml), or antibodies against nucleolin (20 ⁇ g/ml) were added at the beginning of the migration assay.
  • the PBS was added into this migration assay system as a control.
  • the same DMEM medium and reagents were added to the lower chamber.
  • the endothelia cells were allowed to migrate for 6 h at 37° C. and 5% CO 2 .
  • the cDNA of NL was obtained from human microvascular endothelial cells (HMECs) using the SV total RNA isolation system and reverse transcription system (Promega) following the manufacture's protocol.
  • the sequence of NL fusing with polyhistidine (His) 6 tag was amplified by PCR and then subcloned into pPIC9K (Invitrogen). Following the manufacture's protocol, this plasmid was linearized with restriction enzyme Sal I (Promega) and electrotransformed into yeast strain GS115. A stable transformant was selected using G418 and then grown in a 30° C.
  • BMMY medium (10 g/L yeast extract; 20 g/L peptone; 100 mmol/L potassium phosphate, pH 6.0; 13.4 g/L yeast nitrogen base; 40 mg/L biotin; and daily additions of methanol to the medium to yield a final concentration of 0.5%).
  • the supernatant was obtained and the NL was purified on Ni-NTA nickel ion-affinity columns (Qiagen).
  • Qiagen Ni-NTA nickel ion-affinity columns
  • Polyclonal antibodies against nucleolin were prepared with this protein.
  • Hela cells were inoculated in the subcutaneous space of nude mice. Starting from the next day, antibodies against NL were injected slowly into the subcutaneous dorsal of mice at a site remote from the inoculated tumors every 3 days. After the seventh injection, the nude mice were killed, and the tumors were weighed and three measurements of the tumor were taken. The results of these animal tests show that the tumor growth was accelerated significantly when the cell surface nucleolin was blocked with antibodies against nucleolin. These results also demonstrated that nucleolin play a key role in regulating the tumor growth and angiogenesis.
  • the cDNA of NL is obtained from human microvascular endothelial cells (HMECs) using the SV total RNA isolation system and reverse transcription system (Promega) following the manufacture's protocol.
  • the sequence of NL fusing with polyhistidine (His) 6 tag is amplified by PCR and then subcloned into pPIC9K (Invitrogen). Following the manufacture's protocol, this plasmid is linearized with restriction enzyme Sal I (Promega) and electrotransformed into yeast strain GS115. A stable transformant is selected using G418 and then grown in a 30° C.
  • BMMY medium (10 g/L yeast extract; 20 g/L peptone; 100 mmol/L potassium phosphate, pH 6.0; 13.4 g/L yeast nitrogen base; 40 mg/L biotin; and daily additions of methanol to the medium to yield a final concentration of 0.5%).
  • the supernatant is obtained and the NL was purified on Ni-NTA nickel ion-affinity columns (Qiagen).
  • Qiagen Ni-NTA nickel ion-affinity columns
  • This nucleolin is fixed on the Ni-NTA nickel ion-affinity columns (Qiagen) via its fusion peptide at the N-terminus.
  • This affinity bead can be used to screen the nucleolin-binding proteins in a high-throughput manner.
  • the nucleolin-binding proteins can be identified by PFM with MALDI-TOF.
  • the bioactivities of these nucleolin-binding proteins in antiangiogenesis are detected with cellular experiment such as cell migration assay and cell proliferation assay as described above.
  • Binding kinetics was determined by SPR using a BIAcore 2000TM (Amersham Pharmacia Biotech) biosensor system.
  • Purified ES was diluted to 100 ⁇ g/ml in 20 mM sodium acetate, pH 6.5, and covalently immobilized on the research CM5 sensor chips using the amine coupling kit (1-ethyl-3-(dimethylaminopropyl)carbodiimide, (N-hydroxysuccinimide) according to the manufacturer.
  • ES 100 ⁇ g/ml
  • sodium acetate pH 6.5
  • the unreacted moieties on the surface were blocked with ethanolamine (pH 8.5; BIAcore AB).
  • the SPR analysis between ES and NL was performed at 25° C. with a 20 ⁇ l volume of NL (62.5, 125, 250, and 500 nM) in a flow buffer HBS (10 mM Hepes, 150 mM NaCl, 3.4 mM EDTA, and 0.005% surfactant P20, pH 7.4; BIAcore AB) at a flow rate of 10 ⁇ l/min.
  • a flow buffer HBS 10 mM Hepes, 150 mM NaCl, 3.4 mM EDTA, and 0.005% surfactant P20, pH 7.4; BIAcore AB
  • Time course of NL, free in flow buffer, were continuously recorded as resonance units (RU).
  • the surface was regenerated by several 10 ⁇ l pulses of 100 mM NaOH (or 100 mM HCl) flowing at 10 ⁇ l/min.
  • the primary data were analyzed using the BIAevaluation 3.1 Software (BIAcore AB), applying a Langmuir binding model (stoichiometry of 1:1) to calculate k a (association rate constant M ⁇ 1 s ⁇ 1 ), k b (dissociation rate constant, M ⁇ 1 s ⁇ 1 ), and K D (equilibrium constant) for the interaction of ES with NL.
  • the binding affinity between ES and NL was determined by real-time surface plasmon resonance (SPR), which is a rapid and sensitive method for evaluating the affinities involved in bimolecular interaction.
  • SPR surface plasmon resonance
  • the equilibrium constant (K D ) for the interaction of ES with NL is derived to be 2.32 ⁇ 10 ⁇ 8 M from these curves.
  • Exponential growing B16/F10 mouse melanoma cells (2 ⁇ 10 6 cells in 200 ⁇ l of PBS) were inoculated in the subcutaneous space of 2-mo-old Balb/c mice. The animals were used for experiments of co-localization between ES and NL in vivo 8 days after the implantation.
  • the biotinylated ES (40 ⁇ g) and polyclonal rabbit antibodies against NL (200 ⁇ g) were injected i.v., respectively.
  • the biotinylated ES (40 ⁇ g) and purified rabbit IgG was intravenously injected as a control.
  • the mice were anesthetized 1 hour after the injection, perfused through the heart with 20 ml PBS, and killed.
  • mice Some tissues and tumor of the mice were fixed and sectioned. The sections were detected with both TRITC-conjugated avidin (Pierce) and FITC-conjugated secondary antibodies, and were observed under the Olympus Fluoview laser scanning confocal imaging system (Olympus Inc.).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Biomedical Technology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Hospice & Palliative Care (AREA)
  • Food Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Peptides Or Proteins (AREA)
US11/193,042 2005-05-12 2005-07-29 Nucleolin-mediated cancer diagnostics and therapy Abandoned US20060258605A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/412,065 US11434288B2 (en) 2005-05-12 2009-03-26 Nucleolin-mediated cancer diagnostics and therapy
US17/858,710 US20230174644A1 (en) 2005-05-12 2022-07-06 Nucleolin-mediated cancer diagnostics and therapy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN200510011707.3 2005-05-12
CN2005100117073A CN1862258B (zh) 2005-05-12 2005-05-12 核仁素辅助的癌症诊断与治疗方法

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/412,065 Division US11434288B2 (en) 2005-05-12 2009-03-26 Nucleolin-mediated cancer diagnostics and therapy

Publications (1)

Publication Number Publication Date
US20060258605A1 true US20060258605A1 (en) 2006-11-16

Family

ID=37389730

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/193,042 Abandoned US20060258605A1 (en) 2005-05-12 2005-07-29 Nucleolin-mediated cancer diagnostics and therapy
US12/412,065 Active 2025-09-06 US11434288B2 (en) 2005-05-12 2009-03-26 Nucleolin-mediated cancer diagnostics and therapy
US17/858,710 Pending US20230174644A1 (en) 2005-05-12 2022-07-06 Nucleolin-mediated cancer diagnostics and therapy

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/412,065 Active 2025-09-06 US11434288B2 (en) 2005-05-12 2009-03-26 Nucleolin-mediated cancer diagnostics and therapy
US17/858,710 Pending US20230174644A1 (en) 2005-05-12 2022-07-06 Nucleolin-mediated cancer diagnostics and therapy

Country Status (11)

Country Link
US (3) US20060258605A1 (ja)
EP (2) EP2375251B1 (ja)
JP (3) JP4742142B2 (ja)
CN (2) CN1862258B (ja)
AT (1) ATE516499T1 (ja)
AU (2) AU2006246197B2 (ja)
CA (1) CA2607832A1 (ja)
DK (1) DK1912066T3 (ja)
ES (2) ES2369383T3 (ja)
HK (2) HK1114903A1 (ja)
WO (1) WO2006119706A1 (ja)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090017009A1 (en) * 2002-04-08 2009-01-15 Bates Paula J Method for the diagnosis and prognosis of malignant diseases
WO2009088837A2 (en) * 2007-12-31 2009-07-16 The University Of Louisville Research Foundation, Inc. Methods and products to target, capture and characterize stem cells
WO2011062997A2 (en) * 2009-11-17 2011-05-26 Musc Foundation For Research Development Human monoclonal antibodies to human nucleolin
US9452219B2 (en) 2011-06-02 2016-09-27 University Of Louisville Research Foundation, Inc. Anti-nucleolin agent-conjugated nanoparticles
WO2017011411A1 (en) * 2015-07-10 2017-01-19 Ohio State Innovation Foundation Methods and compositions relating to anti-nucleolin recombinant immunoagents
WO2017181049A1 (en) * 2016-04-15 2017-10-19 Charlestonpharma, Llc Anti-nucleolin agents
US10857237B2 (en) 2015-05-05 2020-12-08 University Of Louisville Research Foundation, Inc. Anti-nucleolin agent-conjugated nanoparticles as radio-sensitizers and MRI and/or X-ray contrast agents
CN116754768A (zh) * 2023-05-30 2023-09-15 中山大学附属第五医院 Ddx24在维持内皮细胞的核仁稳态中的应用

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1862258B (zh) 2005-05-12 2012-05-30 清华大学 核仁素辅助的癌症诊断与治疗方法
WO2011123945A1 (en) * 2010-04-06 2011-10-13 The University Of British Columbia Compostions, methods and uses for treating respiratory syncytial virus infection
US20130023479A1 (en) * 2011-07-22 2013-01-24 Protgen Ltd. Use of nucleolin as a biomarker for lymphangiogenesis in cancer prognosis and therapy
CN106405096A (zh) * 2015-07-31 2017-02-15 复旦大学附属华山医院 核仁素在制备胃癌诊断试剂和治疗药物中的应用
CN106405084A (zh) * 2015-07-31 2017-02-15 复旦大学附属华山医院 核仁素在制备肠癌诊断试剂和治疗药物中的应用

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030194754A1 (en) * 2002-04-08 2003-10-16 Miller Donald M. Method for the diagnosis and prognosis of malignant diseases

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4981785A (en) 1988-06-06 1991-01-01 Ventrex Laboratories, Inc. Apparatus and method for performing immunoassays
US5376313A (en) 1992-03-27 1994-12-27 Abbott Laboratories Injection molding a plastic assay cuvette having low birefringence
US5643765A (en) 1993-04-06 1997-07-01 University Of Rochester Method for quantitative measurement of gene expression using multiplex competitive reverse transcriptase-polymerase chain reaction
US5876978A (en) 1993-04-06 1999-03-02 Medical College Of Ohio Method for quantitative measurement of gene expression using multiplex competitive reverse transcriptase-polymerase chain reaction
US5639606A (en) 1993-04-06 1997-06-17 The University Of Rochester Method for quantitative measurement of gene expression using multiplex competitive reverse transcriptase-polymerase chain reaction
US5538848A (en) 1994-11-16 1996-07-23 Applied Biosystems Division, Perkin-Elmer Corp. Method for detecting nucleic acid amplification using self-quenching fluorescence probe
JP2000502450A (ja) 1995-12-22 2000-02-29 アボツト・ラボラトリーズ 蛍光偏光免疫アッセイによる診断法
US5885529A (en) 1996-06-28 1999-03-23 Dpc Cirrus, Inc. Automated immunoassay analyzer
US6201104B1 (en) 1998-12-04 2001-03-13 Entremed, Inc. Angiogenesis—inhibiting protein binding peptides and proteins and methods of use
CN1266064A (zh) * 1999-03-03 2000-09-13 华西医科大学 血管生成抑制因子—内皮抑素及其制备方法
JP2003180368A (ja) 2001-12-19 2003-07-02 Pharma Design Inc 癌患者に対する放射線治療の有効性の予測方法
US7544767B2 (en) * 2002-04-05 2009-06-09 Burnham Institute For Medical Research HMGN2 peptides and related molecules that selectively home to tumor blood vessels and tumor cells
WO2003087124A2 (en) * 2002-04-05 2003-10-23 The Burnham Institute Hmgn2 peptides and related melecules that selectively home to tumor blood vessels and tumor cells
US7357928B2 (en) * 2002-04-08 2008-04-15 University Of Louisville Research Foundation, Inc. Method for the diagnosis and prognosis of malignant diseases
GB0209893D0 (en) * 2002-04-30 2002-06-05 Molmed Spa Conjugate
US7078485B2 (en) 2002-12-05 2006-07-18 Yantai Medgenn Ltd. N-terminal modified recombinant human endostatin and its production
US7470667B2 (en) * 2002-12-05 2008-12-30 Medgenn (Hong Kong) Ltd Methods of treating cancer using a modified endostatin protein
JP2007525167A (ja) * 2003-04-01 2007-09-06 ジェンザイム・コーポレーション 乳房内皮細胞発現パターン
JP2007503844A (ja) * 2003-05-20 2007-03-01 ファイザー・プロダクツ・インク 血管新生が関与する障害を診断しかつ治療するための方法及び組成物
CN1862258B (zh) 2005-05-12 2012-05-30 清华大学 核仁素辅助的癌症诊断与治疗方法

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030194754A1 (en) * 2002-04-08 2003-10-16 Miller Donald M. Method for the diagnosis and prognosis of malignant diseases

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8029784B2 (en) 2002-04-08 2011-10-04 University Of Louisville Research Foundation, Inc. Method for the diagnosis and prognosis of malignant diseases
US8586717B2 (en) 2002-04-08 2013-11-19 University Of Louisville Research Foundation, Inc Method for the diagnosis and prognosis of malignant diseases
US20090017009A1 (en) * 2002-04-08 2009-01-15 Bates Paula J Method for the diagnosis and prognosis of malignant diseases
WO2009088837A2 (en) * 2007-12-31 2009-07-16 The University Of Louisville Research Foundation, Inc. Methods and products to target, capture and characterize stem cells
US20090226914A1 (en) * 2007-12-31 2009-09-10 Bates Paula J Methods and products to target, capture and characterize stem cells
WO2009088837A3 (en) * 2007-12-31 2009-11-05 The University Of Louisville Research Foundation, Inc. Methods and products to target, capture and characterize stem cells
JP2013511260A (ja) * 2009-11-17 2013-04-04 エムユーエスシー ファウンデーション フォー リサーチ ディベロップメント ヒトヌクレオリンに対するヒトモノクローナル抗体
CN102770529A (zh) * 2009-11-17 2012-11-07 Musc研究发展基金会 针对人核仁素的人单克隆抗体
WO2011062997A3 (en) * 2009-11-17 2011-07-14 Musc Foundation For Research Development Human monoclonal antibodies to human nucleolin
US20130115674A1 (en) * 2009-11-17 2013-05-09 Musc Foundation For Research Development Human Monoclonal Antibodies to Human Nucleolin
WO2011062997A2 (en) * 2009-11-17 2011-05-26 Musc Foundation For Research Development Human monoclonal antibodies to human nucleolin
US9260517B2 (en) * 2009-11-17 2016-02-16 Musc Foundation For Research Development Human monoclonal antibodies to human nucleolin
US10385128B2 (en) 2009-11-17 2019-08-20 Musc Foundation For Research Development Nucleolin antibodies
US9452219B2 (en) 2011-06-02 2016-09-27 University Of Louisville Research Foundation, Inc. Anti-nucleolin agent-conjugated nanoparticles
US11344633B2 (en) 2011-06-02 2022-05-31 University Of Louisville Research Foundation, Inc Anti-nucleolin agent-conjugated nanoparticles
US10857237B2 (en) 2015-05-05 2020-12-08 University Of Louisville Research Foundation, Inc. Anti-nucleolin agent-conjugated nanoparticles as radio-sensitizers and MRI and/or X-ray contrast agents
WO2017011411A1 (en) * 2015-07-10 2017-01-19 Ohio State Innovation Foundation Methods and compositions relating to anti-nucleolin recombinant immunoagents
WO2017181049A1 (en) * 2016-04-15 2017-10-19 Charlestonpharma, Llc Anti-nucleolin agents
CN116754768A (zh) * 2023-05-30 2023-09-15 中山大学附属第五医院 Ddx24在维持内皮细胞的核仁稳态中的应用

Also Published As

Publication number Publication date
EP2375251B1 (en) 2015-04-08
HK1162667A1 (en) 2012-09-21
US20230174644A1 (en) 2023-06-08
CN102539734A (zh) 2012-07-04
JP4742142B2 (ja) 2011-08-10
CN1862258B (zh) 2012-05-30
AU2010241351A1 (en) 2010-12-02
CN102539734B (zh) 2016-02-03
EP2375251A1 (en) 2011-10-12
JP2008541066A (ja) 2008-11-20
ES2369383T3 (es) 2011-11-30
ES2538344T3 (es) 2015-06-19
CN1862258A (zh) 2006-11-15
EP1912066A1 (en) 2008-04-16
DK1912066T3 (da) 2011-10-24
ATE516499T1 (de) 2011-07-15
US11434288B2 (en) 2022-09-06
AU2006246197A1 (en) 2006-11-16
US20090191224A1 (en) 2009-07-30
AU2010241351B2 (en) 2013-04-04
EP1912066A4 (en) 2008-10-15
WO2006119706A1 (fr) 2006-11-16
EP1912066B1 (en) 2011-07-13
CA2607832A1 (en) 2006-11-16
AU2006246197B2 (en) 2010-08-19
JP5502652B2 (ja) 2014-05-28
JP2011013225A (ja) 2011-01-20
JP2014032203A (ja) 2014-02-20
JP5758962B2 (ja) 2015-08-05
HK1114903A1 (en) 2008-11-14

Similar Documents

Publication Publication Date Title
US20230174644A1 (en) Nucleolin-mediated cancer diagnostics and therapy
US11203641B2 (en) Screening for anti-cancer compounds using netrin-1 activity
US10151754B2 (en) Cell surface prostate cancer antigen for diagnosis
AU2005219322B2 (en) Pharmaceutical Composition Comprising CXCR3 Inhibitor
US8440415B2 (en) BORIS isoforms and methods of detecting and treating disease
KR101374758B1 (ko) 항-사이토케라틴 8/18 복합체 자가면역항체를 포함하는 암 진단 마커 및 이의 항원을 포함하는 암 진단용 조성물
EP3917571A1 (en) Cnx/erp57 inhibitor for use in the treatment or prevention of cancer
US20100173314A1 (en) Diagnostic methods using tenascin-w compositions
KR101808763B1 (ko) 방사선 피폭에 의한 심장 손상 예측용 바이오마커 및 그 예측방법
US8062859B2 (en) Method for screening anticancer substances, set or kit for implementing said method
US11981729B2 (en) Method

Legal Events

Date Code Title Description
AS Assignment

Owner name: PROTGEN LTD., CHINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LUO, YONGZHANG;SHI, HUBING;REEL/FRAME:020535/0153

Effective date: 20080219

Owner name: TSINGHUA UNIVERSITY, CHINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LUO, YONGZHANG;SHI, HUBING;REEL/FRAME:020535/0153

Effective date: 20080219

STCB Information on status: application discontinuation

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