WO1993020229A1 - ANTIBODIES TO ALPHAvBETA3 INTEGRIN - Google Patents

ANTIBODIES TO ALPHAvBETA3 INTEGRIN Download PDF

Info

Publication number
WO1993020229A1
WO1993020229A1 PCT/US1993/002987 US9302987W WO9320229A1 WO 1993020229 A1 WO1993020229 A1 WO 1993020229A1 US 9302987 W US9302987 W US 9302987W WO 9320229 A1 WO9320229 A1 WO 9320229A1
Authority
WO
WIPO (PCT)
Prior art keywords
monoclonal antibody
binding
avβ3
antibody composition
cells
Prior art date
Application number
PCT/US1993/002987
Other languages
French (fr)
Inventor
Kyung Jin Kim
Michael A. HORTON
Sarah C. Bodary
Anan Chuntharapai
Original Assignee
Genentech, Inc.
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 Genentech, Inc. filed Critical Genentech, Inc.
Priority to EP93908677A priority Critical patent/EP0633945B1/en
Priority to US08/307,844 priority patent/US5578704A/en
Priority to JP51764493A priority patent/JP3353209B2/en
Priority to AU39413/93A priority patent/AU680411B2/en
Priority to CA002132091A priority patent/CA2132091C/en
Priority to DE69322860T priority patent/DE69322860T2/en
Publication of WO1993020229A1 publication Critical patent/WO1993020229A1/en

Links

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
    • C07K16/2839Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily
    • C07K16/2848Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily against integrin beta3-subunit-containing molecules, e.g. CD41, CD51, CD61
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S530/00Chemistry: natural resins or derivatives; peptides or proteins; lignins or reaction products thereof
    • Y10S530/867Chemistry: natural resins or derivatives; peptides or proteins; lignins or reaction products thereof involving immunoglobulin or antibody produced via recombinant dna technology

Definitions

  • This application relates to hybrid cell lines (lymphocyte hybridomas) for the production of monoclonal antibodies to ⁇ v ?3 i ⁇ tegrin, to such homogeneous antibodies, and to the use of such antibodies for diagnostic and therapeutic purposes.
  • av ⁇ 3 is a member of the integrin supergene family of cell-surface glycoprotein receptors that promote cellular adhesion. Each cell has a specific repertoire of receptors that define its adhesive capabilities.
  • the integrins are expressed as heterodimers of noncovalentl ⁇ associated a and ⁇ subunits. According to the nomenclature proposed by Hynes, R.O. [Cell 48. 875-886 (1987)1, the integrins can be divided into families each with a common ?-subunit and a set of variable ⁇ -subunits known to associate with the common .-subunit.
  • ⁇ v stands for a vitronectin receptor ⁇ -chain
  • integrin receptors have been shown to interact with proteins via a tripeptide sequence, Arg-Gly-Asp (or RGD using the single letter amino acid code), originally defined from studies of the cell binding domains of fibronectin [Ruoslahti, E. and Pierschbachter, M.D., Ceil 44 5170518 (1986); Ruoslahti, E. and Pierschbachter, M.D.. Science 238.
  • av ⁇ 3 also referred to as vitronectin receptor or VNR
  • VNR vitronectin receptor
  • av ⁇ 3 is a member of the 03 integrin subfamily and is expressed on a variety of cells, including endothelial, melanoma, smooth muscle cells and, along with another integrin ⁇ 2 ?1 ( VLA-2) (the receptor for Type I collagen and laminin), on the surface of osteoclasts [Horton, M.A. and Davies, J., J. Bone Min. Res.
  • ⁇ v ⁇ 3 mediates cell adhesion to vitronectin, fibrinogen, fibronectin, thrombospondin, osteopontin, bone sialo protein II and von Willebrand factor.
  • Osteoclasts are the main type of bone cells involved in the resorption of bone tissues.
  • the resorption process involves the proliferation and chemotaxis of developing osteoclasts to the skeleton from hematopoietic sites migration of mature cells to sites of subsequent resorption, attachment of osteoclasts to bone substrate and the eventual formation of the polarized, functional mature end cells which are directly involved in bone resorption.
  • the ⁇ v ⁇ 3 integrin mediates adhesion of osteoclasts to RGD sequence-containing bone matrix proteins.
  • Antibodies to ⁇ v ⁇ 3 are expected to be valuable diagnostic and therapeutic tools in studying the biological role and the structural/functional relationships of this integrin with its various liga ⁇ ds.
  • monoclonal antibodies (Mabs) detecting unique epitopes on osteoclasts would be of great value in understanding of the development of osteoclasts.
  • neutralizing Mabs specific for ⁇ v/73 that inhibit the osteoclast binding to the bone matrix proteins have great potential as therapeutic agents useful in the treatment of conditions associated with excessive bone resorption.
  • monoclonal antibodies known in the art that bind to various epitopes on av ⁇ 3. immunizing with osteoclasts from osteoclastomas (giant cell tumors of bone), Horton, M.A.
  • the present invention is based on successful research involving the production and extensive characterization of monoclonal antibodies to ⁇ v ⁇ 3 integrin. Accordingly, the present invention is directed to monoclonal antibodies, and derivatives thereof, which are capable of recognizing unique epitopes on ⁇ v ⁇ 3 and/or which exhibit high affinity for ⁇ v ⁇ 3. The invention is specifically directed to monoclonal antibodies recognizing unique epitopes on the ⁇ v ⁇ 3 complex or the ⁇ 3 portion thereof. The invention is further directed to monoclonal antibodies effectively inhibiting the binding to vitronectin and fibrinogen of av ⁇ 3 expressing cells.
  • the invention is directed to monoclonal antibodies specifically binding av ⁇ 3 on osteoclasts but not other ⁇ v ?3 on other cells (e.g. melanoma cells C32R, M-21 , HA-A, HA-L and HT-144 and human umbilical vein endothelial cells).
  • melanoma cells C32R, M-21 , HA-A, HA-L and HT-144 and human umbilical vein endothelial cells e.g. melanoma cells C32R, M-21 , HA-A, HA-L and HT-144 and human umbilical vein endothelial cells.
  • the invention concerns an anti- ⁇ v 3 monoclonal antibody that is capable of: (1 ) inhibiting the binding of av ⁇ 3 expressing cells to fibrinogen, (2) binding osteoclasts, and (3) binding to substantially the same epitope recognized by any one of a monoclonal antibody selected from the group consisting of 10C4.1.3, 9G2.1.3 and 9D4.9.1 or which has an affinity for ⁇ v ⁇ 3 which is about equal to or greater than that of the foregoing three antibodies.
  • the invention concerns isolated nucleic acid encoding such antibodies, and hybridoma or recombinant cells producing such antibodies.
  • the invention concerns the therapeutic or diagnostic use of such antibodies.
  • the monoclonal antibodies of the invention are useful as therapeutic agents, either by themselves or in conjunction with (chemo)therapeutic agents, to treat diseases or conditions that are characterized by excessive bone resorption and/or to inhibit tumor growth.
  • the monoclonal antibodies of the invention also are useful in diagnostic and analytical assays for determining the presence of av ⁇ 3 on cells, cell typing and in histochemical tissue staining.
  • Fig. 1 is a schematic of the methods for generation of anti- ⁇ v 3 antibodies.
  • Fig. 2 depicts an immunoprecipitation of av ⁇ 3 components using the antibodies of this invention, two positive controls (23C6 and 13C2) and an IgG negative control.
  • Fig. 3A-F is a flow cytometry comparison of Mab 23C6 av ⁇ 3 epitopes compared with the epitopes of several Mabs of this invention.
  • Fig. 3A shows staining of ⁇ v ⁇ 3 transformed cells with fluorescent labeled 23C6 alone
  • Fig. 3B depicts staining with fluorescent labeled 23C6 in competition with unlabeied 23C6, and
  • Figs. 3C, 3D, 3E and 3F illustrate staining with 4
  • Fig. 4A-B illustrates the ability of the Mabs to inhibit binding of av ⁇ 3 transformed 293 cells to fibrinogen (Fig. 4A) or vitronectin (Fig. 4B).
  • 23C6 could inhibit cell binding to fibrinogen, as could the other Mabs.
  • 9D4.9.1 demonstrated substantially higher affinity than did any of the other Mabs tested.
  • Figs. 5A and 5B respectively, depict the inhibition by various Mabs of soluble av ⁇ 3 binding to fibrinogen and vitronectin. The results largely parallel those shown in Fig.4A-B.
  • FIG. 6A-B shows the immunoperoxidase histochemical staining of human osteoclasts fmultinucleated cells) from giant cell tumor of bone.
  • 6A Mab 10C4.1.3.
  • 6B IgG control antibody. Pictures were taken at 330 X magnification.
  • monoclonal antibody refers to a substantially homogeneous population of antibodies, i.e., the individual antibodies comprising the population are identical in specificity and affinity except for possible naturally occurring mutations that may be present in minor amounts. Note that a monoclonal antibody composition may contain more than one monoclonal antibody.
  • the monoclonal antibodies included within the scope of the invention include hybrid and recombinant antibodies (e.g. "humanized” antibodies) regardless of species of origin or immunoglobulin class or subclass designation, as well as antibody fragments (e.g., Fab, F(ab') 2 , and Fv), so long as they have the novel and unobvious characteristics of the antibodies described herein, in preferred embodiments being antibodies that are capable of binding to substantially the same epitope as one recognized by monoclonal antibody 10C4.1.3, 9G2.1.3 or 9D4.9.1 and/or have affinity for that epitope which is greater than or equal to the affinity of 23C6 or 9D4.9.1.
  • the modifier "monoclonal” indicates the character of the antibody as a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies of the invention may be made using the hybridoma method first described by Kohler & Milstein, Nature 256:495 (1975), or may be made by recombinant DNA methods. For example, see Cabilly, e
  • a mouse or other appropriate host animal is immunized with av ⁇ 3 integrin by subcutaneous, i ⁇ traperitoneal, or intramuscular routes to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization.
  • lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell.
  • a suitable fusing agent such as polyethylene glycol
  • lymph node cells (rather than spleen or other tissue) as fusion partners was believed to be instrumental.
  • the hybridoma cells thus prepared are seeded and grown in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
  • HAT medium hypoxanthine, aminopterin, and thymidine
  • Preferred myeloma cells are those that fuse efficiently, support stable high level expression of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium.
  • preferred myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-1 1 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, California USA, and SP-2 cells available from the American Type Culture Collection, Rockville, Maryland USA, or P3X63Ag8U.1 murine myeloma cells [Yelton et al., Curr. Top. Microbiol. Immunol. 81. 1 (1978)].
  • Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies. Kozbor, J. Immunol. 133:3001 (1984). Brodeur. etal.. Monoclonal Antibody Production Techniques and Applications, DD.51 - 63 (Marcel Dekker, Inc., New York, 1987).
  • the hybridomas were prepared from lymph node fusions.
  • Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the individual chains or, preferably the ⁇ v ⁇ 3 complex.
  • the binding specificity is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA), or by FACS sorting.
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • FACS sorting FACS sorting.
  • the monoclonal antibodies of the invention are those that bind to soluble or cell bound ⁇ v ⁇ 3 and which are neutralizing, as explained infra.
  • the specificity of binding ⁇ v ⁇ 3 on various cell types is determined, with the objective being the identification of antibodies that do not bind to any other integrin than ⁇ v ⁇ 3 and, preferably, are able to discriminate between ⁇ v ⁇ 3 on melanoma tumor cells, endothelial cells and osteoclasts, i.e., are substantially specific for any one of such cell types.
  • Substantial specificity in general means that the antibody is specific for the candidate cell type at least to the degree of discrimination shown by Mab 10C4.1.3 for osteoclasts compared to any one of cell lines C32R, M-21 , HA-A, HA-L, HT-63 or MG-63.
  • the screen optionally is narrowed to detect antibodies that bind to substantially the same epitope recognized by antibodies 10C4.1.3, 9G2.1.3 or 9D4.9.1 (as determined by competition assays of the sort described infra for 23C6, except that the 3 Mabs of this invention will be employed as the labelled competitive agent to determine epitope binding of the candidate).
  • “same epitope” does not mean the exact amin ⁇ acid or carbohydrate to which any of the three benchmark antibodies bind, as may be determined for example by epitope mapping using alanine scanned variants of ⁇ v53.
  • “Same epitope” means the av ⁇ 3 domain which is blocked by the binding to av ⁇ 3 of one of the native benchmark antibodies in intact form.
  • “same epitope” includes the av ⁇ 3 domain residues or carbohydrate that structurally interacts or binds to the benchmark CDRs.
  • the monoclonal antibody will have an affinity which is greater than that of 23C6 and preferably is equal or greater than that of 9D4.9.1 , as determined, for example, by the Scatchard analysis of Munson & Pollard, Anal. Biochem. 10Z:220 (1980).
  • neutralizing antibody refers to a monoclonal antibody that is capable of substantially inhibiting or eliminating a biological activity of ⁇ v ⁇ 3.
  • a neutralizing antibody will inhibit binding of av ⁇ 3 to a cell matrix ligand such as vitronectin or fibrinogen to a degree equal to or greater than Mab 23C6, and preferably equal to or greater than Mabs 9D4.9.1 , 10C4.1.3 or 9G2.1.3.
  • hybridoma cells After hybridoma cells are identified that produce neutralizing antibodies of the desired specificity and affinity, the clones typically are subcloned by limiting dilution procedures and grown by standard methods. Goding, Monoclonal Antibodies: Principles and Practice, pp.59- 104 (Academic Press, 1986). Suitable culture media for this purpose include, for example, Dulbecco's Modified Eagle's Medium or RPMI-1640 medium. In addition, the hybridoma cells may be grown in vivo as ascites tumors in an animal.
  • the monoclonal antibodies secreted by the subduc are suitably separated from the culture medium, ascites fluid, or serum by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose-, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • DNA encoding the monoclonal antibodies of the invention is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies).
  • the hybridoma cells of the invention serve as a preferred source of such DNA.
  • the DNA is ligated into expression or cloning vectors, which are then
  • transformant cells •transfected into host cells such as simian COS cells, Chinese Hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein.
  • the transformant cells are cultured to obtain the synthesis of monoclonal antibodies in the recombinant host cell culture.
  • the DNA optionally is modified in order to change the character of the immunoglobulin produced by its expression.
  • Immunoglobulin variants are well known.
  • chimeric antibodies are made by substituting the coding sequence for human heavy and light chain constant domains in place of the homologous murine sequences, Cabilly et al. OP cit. or Morrison, et aL, Proc. Nat. Acad. Sci. 81:6851 (1984).
  • the Fc domain chosen is any of IgA, IgD, IgE, lgG-1 , -2, -3 or -4, or IgM.
  • the Fc domain optionally is capable of effector functions such as complement binding.
  • Humanized forms of the murine antibodies are made by substituting the complementarity determining regions of the mouse antibody into a human framework domain, e.g., see PCT Pub. No. W092/22653, published 23 December 1992.
  • selected murine framework residues also are substituted into the human recipient immunoglobulin.
  • Fusions of the immunoglobulins of this invention and cytotoxic moieties are made, for example, by ligating to the immunoglobulin coding sequence all or part of the coding sequence for a cytotoxic non-immunogiobulin polypeptide.
  • non-immunoglobuiin polypeptides include polypeptide toxins such as ricin, diphtheria toxin, or Pseudomonas exotoxin.
  • the conjugates can be prepared by in vitro methods.
  • immunotoxins may be constructed using a disulfide exchange reaction or by forming a thioether bond between the immunoglobulin and the toxin polypeptide.
  • suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate.
  • other fusions are readily produced by similar recombinant methods.
  • Suitable fusion partners for the immunoglobulin of this invention include viral sequences, cellular receptors such as the T-cell receptor, cytokines such as TNF, interferons, or interleukins, and other biologically or immunologically active polypeptides.
  • non-immunoglobulin fusion polypeptides are substituted for the constant domains of an antibody of the invention.
  • they are substituted for the variable domains of one antigen-combining site of an antibody of the invention.
  • Substitution of the Fr or CDRs of an antibody having specificity for a non ⁇ v/93 antigen will create a chimeric bivalent antibody comprising one antigen-combining site having specificity for ⁇ v ?3 and another antigen-combining site having specificity for a different antigen.
  • the light chain is deleted and the Fv of the heavy chain is substituted with the desired polypeptide.
  • antibodies are termed bivalent or polyvalent, depending upon the number of immunoglobulin "arms" possessed by the Fc domain employed (IgMs will be polyvalent).
  • the antibody also is rendered multivalent by recombination of antibodies having more than one specificity.
  • the antibody in some embodiments is capable of binding ⁇ v ⁇ as described elsewhere herein but is also capable of binding a T-cell determinant such as CD3, CD4, CD8, CD 8, CD1 1 a, CD1 1 b or CD1 1 c.
  • T-cell determinant such as CD3, CD4, CD8, CD 8, CD1 1 a, CD1 1 b or CD1 1 c.
  • the multispecific, multivalent antibodies are made by cotransforming a cell with DNA encoding the heavy and light chains of both antibodies and the proportion of expressed antibodies having the desired structure recovered by immunoaffinity chromatography or the like.
  • Such antibodies are made from monovalent antibodies which are recombined in vitro in conventional fashion.
  • Monovalent antibodies also are made by techniques that are conventional per se. Recombinant expression of light chain and a modified heavy chain is suitable. The heavy chain is truncated generally at any point in the Fc region so as to prevent heavy chain crosslinking. Alternatively, the relevant cysteines are substituted with another residue or deleted so as to prevent crosslinking. In vitro methods also are used to produce monovalent antibodies, e.g., Fab fragments are prepared by enzymatic cleavage of intact antibody.
  • the antibodies of the invention typically will be labeled with a detectable moiety.
  • the detectable moiety can be any one which is capable of producing, either directly or indirectly, a detectable signal.
  • the detectable moiety may be a radioisotope, such as 3 H, 14 C, 3 P, 3 ⁇ S, or 12B I, a fluorescent or chemiluminescent compound, such as fluorescein isothiocyanate, rhodamine, or luciferin; radioactive isotopic labels, such as, e.g., 126 l, 32 P, 1 C, technicium, or 3 H, or an enzyme, such as alkaline phosphatase, beta- galactosidase or horseradish peroxidase.
  • any method known in the art for separately conjugating the antibody to the detectable moiety may be employed, including those methods described by Hunter, e$ al.. Nature 144:945 (1962); David, ej ah. Biochemistry 13:1014 (1974); Pain, e
  • the antibodies of the present invention may be employed in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprec ⁇ pitation assays. Zola. Monoclonal Antibodies: A Manual of Techniques, pp.147- 158 (CRC Press, Inc., 1987).
  • ком ⁇ онентs rely on the ability of a labeled standard (which may be ⁇ v 73 or an immunologically reactive portion thereof) to compete with the test sample analyte ( ⁇ v ?3) for binding with a limited amount of antibody.
  • the amount of av ⁇ 3 in the test sample is inversely proportional to the amount of standard that becomes bound to the antibodies.
  • the antibodies generally are insolubilized before or after the competition, so that the standard and analyte that are bound to the antibodies may conveniently be separated from the standard and analyte which remain unbound.
  • Sandwich assays involve the use of two antibodies, each capable of binding to a different immunoge ⁇ ic portion, or epitope, of the protein to be detected.
  • the test sample analyte is bound by a first antibody which is immobilized on a solid support, and thereafter a second antibody binds to the analyte, thus forming an insoluble three part complex.
  • the second antibody may itself be labeled with a detectable moiety (direct sandwich assays) or may be measured using an anti-immunoglobulin antibody that is labeled with a detectable moiety (indirect sandwich assay).
  • sandwich assay is an ELISA assay, in which case the detectable moiety is an enzyme.
  • the antibodies of the invention also are useful for ]n vivo imaging, wherein an antibody labeled with a detectable moiety such as a radio-opaque agent or radioisotope is administered to a host, preferably into the bloodstream, and the presence and location of the labeled antibody in the host is assayed.
  • a detectable moiety such as a radio-opaque agent or radioisotope
  • This imaging technique is useful in the staging and treatment of neoplasms or bone disorders.
  • the antibody may be labeled with any moiety that is detectable in a host, whether by nuclear magnetic resonance, radiology, or other detection means known in the art.
  • the neutralizing antibodies of the invention are especially useful in therapeutic applications, to prevent or treat unwanted bone resorption, or tumor cell growth or metastasis.
  • Mabs of the 10C4.1.3 type are not useful for treating or in vivo imaging of tumors of the same type described in Table 2 infra since they do not bind to ⁇ v ⁇ 3 found on such cells. Instead these Mabs are especially useful of treating conditions of bone resorption or degradation, for example as found in osteoporosis or resulting from PTHrP over- expression by some tumors.
  • the antibodies of the invention are administered to a mammal, preferably a human, in a pharmaceutically acceptable dosage form. They are administered intravenously as a bolus or by continuous infusion over a period of time, by intramuscular, subcutaneous, intra-articular, intrasynovial, intrathecal, oral, topical, or inhalation routes.
  • the antibody possesses the suitable activity it is also suitably administered by intratumoral, peritumoral, intralesional, or perilesional routes, to exert local as well as systemic therapeutic effects.
  • Such dosage forms encompass pharmaceutically acceptable carriers that are inherently nontoxic and nontherapeutic.
  • pharmaceutically acceptable carriers include ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffers such as phosphate or glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts, or electrolytes such as protamine sulfate, sodium chloride, metal salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulosic polymers, and polyethylene glycol.
  • Carriers for topical or gel-based forms of antibody include polysaccharides such as sodium carboxymethylcelluiose or methylcellul ⁇ se, polyvinylpyrrolidone, polyacrylates, polyoxyethylene-polyoxypropylene-block polymers, polyethylene glycol, and wood wax alcohols.
  • Conventional depot forms include, for example, microcapsules, nano-capsules, liposomes, plasters, -sublingual tablets, and polymer matrices such as polylactide:polyglycolide copolymers.
  • the antibody When present in an aqueous dosage form, rather than being lyophilized, the antibody typically will be formulated at a concentration of about 0.1 mg/ml to 100 mg/ml, although wide variation outside of these ranges is permitted.
  • the appropriate dosage of antibody will depend on the type of disease to be treated, as defined above, the severity and course of the disease, whether the antibodies are administered for preventive or therapeutic purposes, the course of previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician.
  • the antibody is suitably administered to the patient at one time or over a series of treatments.
  • 0.015 to 15 mg of antibody/Kg of patient weight is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • the treatment is repeated until a desired suppression of disease symptoms occurs.
  • other dosage regimens may be useful and are not excluded herefrom.
  • the effectiveness of the antibody in preventing or treating disease may be improved by administering the antibody serially or in combination with another agent that is effective for the same clinical objective, such as another antibody directed against a different epitope than the principal antibody, or one or more conventional therapeutic agents known for the intended therapeutic indication, e.g. prevention or treatment of conditions associated with excessive bone resorption such as, osteoporosis or inhibition of tumor cell growth or metastasis.
  • Another agent that is effective for the same clinical objective such as another antibody directed against a different epitope than the principal antibody, or one or more conventional therapeutic agents known for the intended therapeutic indication, e.g. prevention or treatment of conditions associated with excessive bone resorption such as, osteoporosis or inhibition of tumor cell growth or metastasis.
  • the antibodies of the invention also are useful as affinity purification agents. In this process, the antibodies against ⁇ v/53 are immobilized on a suitable support, such a Sephadex resin or filter paper, using methods well known in the art.
  • the immobilized antibody then is contacted with a sample containing the av ⁇ 3 to be purified, and thereafter the support is washed with a suitable solvent that will remove substantially all the material in the sample except the ⁇ v/73, which is bound to the immobilized antibody. Finally, the support is washed with another suitable solvent, such as glycine buffer, pH 5.0, that will release the ⁇ v/73 from the antibody.
  • a suitable solvent such as glycine buffer, pH 5.0
  • mice were immunized with ⁇ v ⁇ 3 integrin purified from 293- 5D cell line expressing av ⁇ 3 complex which were generated by transfecting 293 cells (ATCC CRL1573) with DNAs prepared from PMNCV vector expressing av o ⁇ ⁇ 3 plus DNA coding for Neomycin resistance gene.
  • av ⁇ 3 was purified from an NP 40 cell lysate of 293-15D cells by using a lentil lectin column. The purity of the ⁇ v ?3 prepared was then confirmed by isoelectrophoresis.
  • mice were immunized into foot pads once with 5 ⁇ g of av ⁇ 3 emulsified in MPL/TDM adjuvant (Ribi Immunochem. Research Inc., Hamilton, MT) and then subsequently six times with 5 mg of ⁇ v ?3 immersified in M PL/TDM adjuvant at 2 week intervals.
  • lymph node cells from these mice were fused with P3X63Ag8U.1 myeloma cells (Yelton, et al., Curr. Top. Microbiol. Immunol. 81 : 1 , 1978) using 35% polyethylene glycol as described (Yarmush et al, Proc. Nat. Acad. Sci.
  • Hybridoma cell lines were selected for anti- ⁇ v ?3 antibody production by their ability to bind soluble ⁇ v ⁇ 3 by ELISA and to bind cell lines expressing various integrins by Flow microfluorometry analysis using FACSCAN (Becton Dickinson FACS systems. Mountain View, CA). Isotypes (Table 1 ) of these positive Mabs were determined by ELISA using isotype specific alkaline phosphatase- conjugated goat anti-mouse immunoglobulin (Harlow and Lane, Antibodies: A Laboratory Manual, p.597, Cold Spring Harbor Laboratory, 1988).
  • 293-15D transfectants grown F12/DMEM medium with 10% FCS were harvested by treatment with EDTA and biotinylated by using NHS-LC-Biotin.
  • Cells (5 X 106 cells/ml) were incubated with 1 /sg/ml of NIH-LS-Biotin for 1 hour at room temperature. The unbound biotin was then removed by washing in 0.05 mM tris buffer containing 0.5 M NaCI m 1 mm. CaCI2 and 1 m N MgCI2 (cell wash buffer). Cells were lysed by treatment with 1 % NP-40 and cell debris were removed by microcentrifugation for 10 min. The supernatant was used for the immunoprecipitation.
  • Transfected cells and tumor cells grown in F12/DMEM medium (1:1 o/o mixture) containing 10% FCS, glutamine and antibiotics were washed three times in the cell sorter buffer (CSB, PBS containing 1 % FCS and 0.01 % NaN3) by centrifugation at 1 ,000 rpm for 5 min and resuspended to be 4x10 ⁇ cells/ml in CSB. Twenty five ⁇ of cells were added into a 96 well U-bottom plate and incubated with 100 ⁇ of antibodies for 30 min on ice.
  • CSB cell sorter buffer
  • Table 2 depicts the determination of the portion of av ⁇ 3 recognized by these Mabs by FACS analysis of transfected cells expressing different integrins as well as by ELISA using soluble integrin proteins.
  • Mab 9G2.1.3 bound strongly to 293-15D expressing ⁇ v 73 and very weakly to 293-CLB expressing llb-llla, but not 293-52B expressing av ⁇ 3.
  • Mab 10C4.1.3 only bound to 293-15D but not others. Therefore, it was concluded that these two Mabs (9G2.1.3 and 10C4.1.3) recognized av ⁇ 3.
  • Mab 9D4.9.1 strongly bound to both 293-15D as well as 293-CLB but not 293-52B. Thus, the Mab 9D4.9.1 was concluded to the bind ⁇ 3 portion of av ⁇ Z.
  • Table 2 also shows that Mab 9C9.1 1.1 1 bound to osteoclasts, human endothelial cells and various melanoma cells. This staining pattern was similar to that of 23C6. In contrast, Mab 10C4.1.3 recognized only osteoclasts, suggesting a surprising and very narrow specificity.
  • the staining pattern of Mabs 9D4.9.1 and 9G2.1.3 for human melanoma tumor cells, glioma cells and normal endothelial cells were similar to that of Mab 23C6: these Mabs recognized various human melanoma cells, and osteoclasts strongly, and human osteosarcoma MG-63 cells and human endothelial cells weakly.
  • EXAMPLE 3 Determination of epitopes by competitive Immu ⁇ oflmorescence staining av ⁇ 3 transfected 293-15D cells (1X10 B cells/1 OQ ⁇ l) were incubated with 100 //I of the first purified Mab for 30 min on ice, washed twice and incubated with the second Mab, FITC conjugated Mab 23C6 for 30 min. At the end of the incubation, cells were washed twice in CSB and resuspended in 0.5 ml of CBS and the level of FITC conjugated Mab 23C6 binding on 293-15D cells was examined by flow microfluorometr ⁇ (FACSCAN). The results are depicted in Fig. 3. Panels are as follows:
  • FL-23C6 (shaded area represents unstained cells) FL-23C6 FL-23C6 FL-23C6
  • Mab 9G2.1 .3 at high concentration, has some inhibitory effect on the F- 23C6 binding but could not completely block the F-23C6 binding.
  • Mab 9G2.1.3 recognizes a different epitope from the one recognized by F-23C6, but these epitopes appear to be closely orientated. Since it has reported that LM 609 (Cheresh, et al., J. Biol. Chem.. 262:17703-1771 1 [1987]) recognizes the same epitope as 23C6 it is concluded that our Mabs recognized different epitopes from the one recognized by Mab LM 609. EXAMPLE 4
  • Microplates (NUNC, Breakapart C8 Maxi Sorp) were coated with 100 //l/well of 10 ⁇ glml of fibrinogen or vitronectin overnight at 4°C. After being washed three times in PBS, plates were blocked with 1 % BSA in PBS for 1 hr and then were incubated with various concentrations of Mabs for 30 min followed by the addition of 100 ml of 61 Cr labeled 293- 15D cells. Plates were centrifuged at 600 rpm for 2 min and incubated for 90 min at 37°C.
  • E1 Cr labeled 293-15D transfectants expressing av ⁇ 3 were prepared as follows. Cells were grown in F12/DMEM medium containing 10% FCS, 0.1 % glucose and 2mM glutamine for 40 hr, were harvested by treatment with 10 mM EDTA in PBS for 2 min, washed twice in PBS and resuspended to be 5 x 10 7 cells/ml in culture medium without FCS.
  • the top panel of Figure 4 shows the binding of 61 Cr-293 15 D to fibrinogen coated wells in the presence of various concentration of Mabs. All three Mabs inhibit the binding of 61 Cr-293 15 D to fibrinogen very effectively. The strongest inhibition was shown with Mab 9D4.9.1.
  • the bottom panel shows the binding of 61 Cr-293 15 D to vitronectin coated wells. Under the conditions tested, 9D4.9.1 and 10C4.1 .3 could inhibit this interaction but 9G2.1 .3 and 23C6 showed a very weak inhibition, if any. In general, it was harder to inhibit the interaction between ⁇ v ⁇ 3 transfected cells to vitronectin than the interaction between av ⁇ 3 transfected cells to fibrinogen.
  • Microtiter plates were coated with 100 .l/well of 10 ⁇ g/ml of purified fibrinogen or vitronectin, overnight at 4°C. After washing three times in PBS, the plates were blocked with
  • EXAMPLE 7 Histochemical staining of frozen sections of human tissues and bone imprints Frozen sections of human osteoclastoma tumors and bone imprints from human fetal limb bones (14 weeks gestation), newborn rabbit and rat bone, embryonic chick bone and adult red deer antler, and from the following tissues of adult human origin (liver, kidney, pancreas, colon, ileum, heart, lung, thymus, tonsil, spleen, placenta, skin, uterine cervix, umbilical cord, breast carcinoma, malignant melanoma, smears of peripheral blood and bone marrow ononuclear cells) were prepared as described [Horton, M.A. etal., Cancer Res. 45.
  • the bound peroxidase was developed in 0.1 mg/ml diaminobenzidine tetrahydrochloride containing 0.07% H 2 0 2 (Organon Teknina Corp. Durham, NC) in pBS and counter stained with 0.5% methyl green for 5 minutes. The slides were then dehydrated in graded alcohols, then cleared in xylene and mounted in permountant (Fisher Scientific Co., San Francisco, CA) for microscopy.
  • Figure 6 shows clear membrane staining of multinucleated osteoclasts from human bone imprints and frozen sections of giant cell tumor of bone using Mab 10C4.1.3; a control Mab did not show staining.
  • Mabs 9D4.9.1 and 9G2.1.3 showed a similar staining pattern to Mab 10C4.1.3. None of the Mabs recognized osteoclast vitronectin receptor in conventional formalin-fixed and paraffin-embedded tissue sections.
  • Bone imprints were stained with mAbs followed by F-goat anti-mouse IgG.
  • the level of fluorescence staining was examined by fluorescent microscopy and graded as weak ( +), moderate (+ +) and strong (+ + +) or absent (-).
  • Mab 9G2.1.3 recognizes osteoclasts from rabbit, chicken, deer in addition to human; this distribution is similar to that seen with 23C6 (Horton etal., 1985, oo. cit.). In contrast, Mabs 9D4.9.1 and 10C4.1.3 only recognized human osteoclasts; to date, no ⁇ v ⁇ 3 complex- specific Mabs have shown such species selectivity.
  • Mab 9D4.9.1 stained platelets (and megakaryocytes in bone marrow) intensely in all tissues studied.
  • vascular endotheiium was stained, variably and weakly, in all tissues.
  • Mab 9G2.1 .3 also stained vascular endotheiium, but failed to react with platelets and megakaryocytes.
  • Mab 10C4.1.3 failed to stain or gave a much weaker reaction in tissues recognized by Mab 9G2.1.3. For example, Mab 10C4.1.3 did not stain intestinal smooth muscle or placenta.
  • Mabs 9D4.9.1 and 9G2.1.3 bound strongly to various human melanoma cells and less to MG-63 human osteosarcoma cells and HUVEC.
  • Mab 10C4.1.3 bound weakly to only one of the human melanoma cell lines, M-21.

Abstract

The invention relates to monoclonal antibodies to the avβ3 integrin receptor known to be expressed in large amounts on the surface of osteoclasts and accordingly, associated with bone resorption. The disclosed monoclonal antibodies are believed to recognize unique epitopes on avβ3 and are useful in the treatment of conditions associated with excessive bone resorption and/or in the inhibition of tumor cell growth.

Description

ANTIBODIES TO ALPHAvBETA3 INTEGRIN ar
Field of the Invention
This application relates to hybrid cell lines (lymphocyte hybridomas) for the production of monoclonal antibodies to σv ?3 iπtegrin, to such homogeneous antibodies, and to the use of such antibodies for diagnostic and therapeutic purposes.
Background of the Invention avβ3 is a member of the integrin supergene family of cell-surface glycoprotein receptors that promote cellular adhesion. Each cell has a specific repertoire of receptors that define its adhesive capabilities. The integrins are expressed as heterodimers of noncovalentlγ associated a and β subunits. According to the nomenclature proposed by Hynes, R.O. [Cell 48. 875-886 (1987)1, the integrins can be divided into families each with a common ?-subunit and a set of variable σ-subunits known to associate with the common .-subunit. The different a chains are denoted by the original cell type, by a subscript used by the original discoverer, or, as in the case of the σv ?3 receptor, by the nature of the ligand (i.e. σv stands for a vitronectin receptor σ-chain). Many, but not all, integrin receptors have been shown to interact with proteins via a tripeptide sequence, Arg-Gly-Asp (or RGD using the single letter amino acid code), originally defined from studies of the cell binding domains of fibronectin [Ruoslahti, E. and Pierschbachter, M.D., Ceil 44 5170518 (1986); Ruoslahti, E. and Pierschbachter, M.D.. Science 238. 491-497 (1987)1. avβ3 (also referred to as vitronectin receptor or VNR) is a member of the 03 integrin subfamily and is expressed on a variety of cells, including endothelial, melanoma, smooth muscle cells and, along with another integrin σ2 ?1 ( VLA-2) (the receptor for Type I collagen and laminin), on the surface of osteoclasts [Horton, M.A. and Davies, J., J. Bone Min. Res.
4, 803-808 (1989); Davies, J. et al., J. Cell. Biol. 109. 1817-1826(1989); Horton, M„ Int.
J. EXD. Pathol. 71 741 -759 (1990)]. αvβ3 mediates cell adhesion to vitronectin, fibrinogen, fibronectin, thrombospondin, osteopontin, bone sialo protein II and von Willebrand factor.
Osteoclasts are the main type of bone cells involved in the resorption of bone tissues. The resorption process involves the proliferation and chemotaxis of developing osteoclasts to the skeleton from hematopoietic sites migration of mature cells to sites of subsequent resorption, attachment of osteoclasts to bone substrate and the eventual formation of the polarized, functional mature end cells which are directly involved in bone resorption. The σvβ3 integrin mediates adhesion of osteoclasts to RGD sequence-containing bone matrix proteins.
Antibodies to σvβ3 are expected to be valuable diagnostic and therapeutic tools in studying the biological role and the structural/functional relationships of this integrin with its various ligaήds. In particular, monoclonal antibodies (Mabs) detecting unique epitopes on osteoclasts would be of great value in understanding of the development of osteoclasts. Even more importantly, neutralizing Mabs specific for σv/73 that inhibit the osteoclast binding to the bone matrix proteins have great potential as therapeutic agents useful in the treatment of conditions associated with excessive bone resorption. There are several monoclonal antibodies known in the art that bind to various epitopes on avβ3. immunizing with osteoclasts from osteoclastomas (giant cell tumors of bone), Horton, M.A. eta/, f Cancer Res. 45. 5663-5669 (1985)1 produced eleven mouse hybridomas secreting monoclonal antibodies which bind to osteoclasts in normal human fetal bone and a variety of neopiastic and non-neoplastic bone lesions. One of these, designated 23C6, was subsequently shown to bind the σvβ3 complex, and was demonstrated to be able to disrupt osteoclast function [Horton, M.A. et al., Exp. Cell. Res. 195 368-375 (1991 )1. Another monoclonal antibody, LM609 (produced in hybridoma LM609 ATCC HB 9537) disclosed in PCT Application Publication No. WO 89/05155 (published 15 June 1989) and Cheresh et al. J. Biol. Chem. 262:17703-1771 1 (1987) was also found to bind the αvβ3 complex and, due to its ability to inhibit the binding of ECr molecules present on the surface of tumor cells and blood vessel forming endothelial cells to vitronectin, f ibrinogen and von Willebraπd factor, was proposed for therapeutic use as tumor growth inhibitor. Monoclonal antibody 3C2 (Horton, M.A. et al., Cancer Res. 1985, Supra) was shown to bind the σv portion of the αvβ3 molecule, whereas several other monoclonal antibodies were reported to recognize the β3 portion [Nesbitt, S. etal., Epitope Analysis of the Vitronectin Receptor (CD51 ), In "Leukocyte Typing IV" White Cell Differentiation Antigens, napp, W. etal. (eds.) 1991 , p. 10371. The specific monoclonal antibodies variously reported in the art were shown to also bind to endothelial cells and various melanoma cell lines.
There is a need for high affinity monoclonal antibodies to the σvβ3 integrin that are capable of effective inhibition of the binding of αvβ3.expressing cells to αvβ3 ligands, such as vitronectin and fibronectin.
It would be further desirable to provide monoclonal antibodies to αvβ3 that bind osteoclasts and optionally other cells known to express αvβ3.
It would be particularly desirable to provide monoclonal antibodies that are effective inhibitors of σvβ3 binding to its ligands and which specifically bind osteoclasts without binding to other cells known to express αvβ3, i.e., which are more specific for the target integrin on osteoclasts.
Summary of the Invention The present invention is based on successful research involving the production and extensive characterization of monoclonal antibodies to αvβ3 integrin. Accordingly, the present invention is directed to monoclonal antibodies, and derivatives thereof, which are capable of recognizing unique epitopes on αvβ3 and/or which exhibit high affinity for αvβ3. The invention is specifically directed to monoclonal antibodies recognizing unique epitopes on the σvβ3 complex or the β3 portion thereof. The invention is further directed to monoclonal antibodies effectively inhibiting the binding to vitronectin and fibrinogen of avβ3 expressing cells. In a particularly important aspect, the invention is directed to monoclonal antibodies specifically binding avβ3 on osteoclasts but not other σv ?3 on other cells (e.g. melanoma cells C32R, M-21 , HA-A, HA-L and HT-144 and human umbilical vein endothelial cells).
In one aspect, the invention concerns an anti-σv 3 monoclonal antibody that is capable of: (1 ) inhibiting the binding of avβ3 expressing cells to fibrinogen, (2) binding osteoclasts, and (3) binding to substantially the same epitope recognized by any one of a monoclonal antibody selected from the group consisting of 10C4.1.3, 9G2.1.3 and 9D4.9.1 or which has an affinity for σvβ3 which is about equal to or greater than that of the foregoing three antibodies.
In another aspect, the invention concerns isolated nucleic acid encoding such antibodies, and hybridoma or recombinant cells producing such antibodies. In a further aspect, the invention concerns the therapeutic or diagnostic use of such antibodies. The monoclonal antibodies of the invention are useful as therapeutic agents, either by themselves or in conjunction with (chemo)therapeutic agents, to treat diseases or conditions that are characterized by excessive bone resorption and/or to inhibit tumor growth. The monoclonal antibodies of the invention also are useful in diagnostic and analytical assays for determining the presence of avβ3 on cells, cell typing and in histochemical tissue staining. These and further aspects will be apparent from the following detailed description.
Brief Description of the Drawings
Fig. 1 is a schematic of the methods for generation of anti-σv 3 antibodies. Fig. 2 depicts an immunoprecipitation of avβ3 components using the antibodies of this invention, two positive controls (23C6 and 13C2) and an IgG negative control.
Fig. 3A-F is a flow cytometry comparison of Mab 23C6 avβ3 epitopes compared with the epitopes of several Mabs of this invention. Fig. 3A shows staining of σvβ3 transformed cells with fluorescent labeled 23C6 alone, Fig. 3B depicts staining with fluorescent labeled 23C6 in competition with unlabeied 23C6, and Figs. 3C, 3D, 3E and 3F illustrate staining with 4
Mabs of this invention in competition with labeled 23C6.
Fig. 4A-B illustrates the ability of the Mabs to inhibit binding of avβ3 transformed 293 cells to fibrinogen (Fig. 4A) or vitronectin (Fig. 4B). Interestingly, 23C6 could inhibit cell binding to fibrinogen, as could the other Mabs. However, 9D4.9.1 demonstrated substantially higher affinity than did any of the other Mabs tested. As shown in the lower panel, only Mabs 10C4.1.3 and 9D4.9.1 were able to substantially inhibit cell binding to vitronectin, and again the latter exhibited higher affinity than the other Mabs tested. Figs. 5A and 5B, respectively, depict the inhibition by various Mabs of soluble avβ3 binding to fibrinogen and vitronectin. The results largely parallel those shown in Fig.4A-B.
Fig. 6A-B shows the immunoperoxidase histochemical staining of human osteoclasts fmultinucleated cells) from giant cell tumor of bone. 6A: Mab 10C4.1.3. 6B: IgG control antibody. Pictures were taken at 330 X magnification.
Detailed Description of the Invention A. Definitions and General Methods
The term "monoclonal antibody" as used herein refers to a substantially homogeneous population of antibodies, i.e., the individual antibodies comprising the population are identical in specificity and affinity except for possible naturally occurring mutations that may be present in minor amounts. Note that a monoclonal antibody composition may contain more than one monoclonal antibody.
The monoclonal antibodies included within the scope of the invention include hybrid and recombinant antibodies (e.g. "humanized" antibodies) regardless of species of origin or immunoglobulin class or subclass designation, as well as antibody fragments (e.g., Fab, F(ab')2, and Fv), so long as they have the novel and unobvious characteristics of the antibodies described herein, in preferred embodiments being antibodies that are capable of binding to substantially the same epitope as one recognized by monoclonal antibody 10C4.1.3, 9G2.1.3 or 9D4.9.1 and/or have affinity for that epitope which is greater than or equal to the affinity of 23C6 or 9D4.9.1.
Thus, the modifier "monoclonal" indicates the character of the antibody as a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies of the invention may be made using the hybridoma method first described by Kohler & Milstein, Nature 256:495 (1975), or may be made by recombinant DNA methods. For example, see Cabilly, e| aL, U.S. Pat. No. 4,816,567; or Mage & Lamoyi, in Monoclonal Antibody Production Techniques and Applications, pp.79-97 (Marcel Dekker, Inc., New York, 1987). In the hybridoma method, a mouse or other appropriate host animal is immunized with avβ3 integrin by subcutaneous, iπtraperitoneal, or intramuscular routes to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization. Alternatively, lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell. Goding, Monoclonal Antibodies: Principles and Practice, pp.59-103 (Academic Press, 1986). immunization with the extracellular domain of avβ3 (truncated av/23 not containing its transmembrane or cytσplasmic domains) as shown in the examples produced a suφrisingly large population of anti-αv ?3 antibodies and is believed in part to be responsible for the unique specificities and high affinities of several Mabs so identified. In addition, use of lymph node cells (rather than spleen or other tissue) as fusion partners was believed to be instrumental.
The hybridoma cells thus prepared are seeded and grown in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells. For example, if the parental myeloma cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells. Preferred myeloma cells are those that fuse efficiently, support stable high level expression of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. Among these, preferred myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-1 1 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, California USA, and SP-2 cells available from the American Type Culture Collection, Rockville, Maryland USA, or P3X63Ag8U.1 murine myeloma cells [Yelton et al., Curr. Top. Microbiol. Immunol. 81. 1 (1978)]. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies. Kozbor, J. Immunol. 133:3001 (1984). Brodeur. etal.. Monoclonal Antibody Production Techniques and Applications, DD.51 - 63 (Marcel Dekker, Inc., New York, 1987). As noted, the hybridomas were prepared from lymph node fusions.
Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the individual chains or, preferably the σvβ3 complex. Preferably, the binding specificity is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA), or by FACS sorting. The monoclonal antibodies of the invention are those that bind to soluble or cell bound αvβ3 and which are neutralizing, as explained infra. Then the specificity of binding αvβ3 on various cell types is determined, with the objective being the identification of antibodies that do not bind to any other integrin than αvβ3 and, preferably, are able to discriminate between αvβ3 on melanoma tumor cells, endothelial cells and osteoclasts, i.e., are substantially specific for any one of such cell types. Substantial specificity in general means that the antibody is specific for the candidate cell type at least to the degree of discrimination shown by Mab 10C4.1.3 for osteoclasts compared to any one of cell lines C32R, M-21 , HA-A, HA-L, HT-63 or MG-63. Obviously this may be expressed in terms of the quantity of antibody that binds or in other conventional measures. Finally, the screen optionally is narrowed to detect antibodies that bind to substantially the same epitope recognized by antibodies 10C4.1.3, 9G2.1.3 or 9D4.9.1 (as determined by competition assays of the sort described infra for 23C6, except that the 3 Mabs of this invention will be employed as the labelled competitive agent to determine epitope binding of the candidate). It should be kept in mind that "same epitope" does not mean the exact aminσ acid or carbohydrate to which any of the three benchmark antibodies bind, as may be determined for example by epitope mapping using alanine scanned variants of σv53. "Same epitope" means the avβ3 domain which is blocked by the binding to avβ3 of one of the native benchmark antibodies in intact form. Of course, "same epitope" includes the avβ3 domain residues or carbohydrate that structurally interacts or binds to the benchmark CDRs. in a preferred embodiment of the invention, the monoclonal antibody will have an affinity which is greater than that of 23C6 and preferably is equal or greater than that of 9D4.9.1 , as determined, for example, by the Scatchard analysis of Munson & Pollard, Anal. Biochem. 10Z:220 (1980).
The term "neutralizing antibody" as used herein refers to a monoclonal antibody that is capable of substantially inhibiting or eliminating a biological activity of σvβ3. Typically a neutralizing antibody will inhibit binding of avβ3 to a cell matrix ligand such as vitronectin or fibrinogen to a degree equal to or greater than Mab 23C6, and preferably equal to or greater than Mabs 9D4.9.1 , 10C4.1.3 or 9G2.1.3.
After hybridoma cells are identified that produce neutralizing antibodies of the desired specificity and affinity, the clones typically are subcloned by limiting dilution procedures and grown by standard methods. Goding, Monoclonal Antibodies: Principles and Practice, pp.59- 104 (Academic Press, 1986). Suitable culture media for this purpose include, for example, Dulbecco's Modified Eagle's Medium or RPMI-1640 medium. In addition, the hybridoma cells may be grown in vivo as ascites tumors in an animal.
The monoclonal antibodies secreted by the subciones are suitably separated from the culture medium, ascites fluid, or serum by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose-, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
DNA encoding the monoclonal antibodies of the invention is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). The hybridoma cells of the invention serve as a preferred source of such DNA.
Once isolated, the DNA is ligated into expression or cloning vectors, which are then
•transfected into host cells such as simian COS cells, Chinese Hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein. The transformant cells are cultured to obtain the synthesis of monoclonal antibodies in the recombinant host cell culture.
The DNA optionally is modified in order to change the character of the immunoglobulin produced by its expression. Immunoglobulin variants are well known. For example, chimeric antibodies are made by substituting the coding sequence for human heavy and light chain constant domains in place of the homologous murine sequences, Cabilly et al. OP cit. or Morrison, et aL, Proc. Nat. Acad. Sci. 81:6851 (1984). In addition, the Fc domain chosen is any of IgA, IgD, IgE, lgG-1 , -2, -3 or -4, or IgM. The Fc domain optionally is capable of effector functions such as complement binding. Humanized forms of the murine antibodies are made by substituting the complementarity determining regions of the mouse antibody into a human framework domain, e.g., see PCT Pub. No. W092/22653, published 23 December 1992. In some embodiments, selected murine framework residues also are substituted into the human recipient immunoglobulin. Fusions of the immunoglobulins of this invention and cytotoxic moieties are made, for example, by ligating to the immunoglobulin coding sequence all or part of the coding sequence for a cytotoxic non-immunogiobulin polypeptide. Such non-immunoglobuiin polypeptides include polypeptide toxins such as ricin, diphtheria toxin, or Pseudomonas exotoxin.
Also, the conjugates can be prepared by in vitro methods. For example, immunotoxins may be constructed using a disulfide exchange reaction or by forming a thioether bond between the immunoglobulin and the toxin polypeptide. Examples of suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate. In addition, other fusions are readily produced by similar recombinant methods. Suitable fusion partners for the immunoglobulin of this invention include viral sequences, cellular receptors such as the T-cell receptor, cytokines such as TNF, interferons, or interleukins, and other biologically or immunologically active polypeptides. Typically such non-immunoglobulin fusion polypeptides are substituted for the constant domains of an antibody of the invention. Alternatively, they are substituted for the variable domains of one antigen-combining site of an antibody of the invention. Substitution of the Fr or CDRs of an antibody having specificity for a non σv/93 antigen will create a chimeric bivalent antibody comprising one antigen-combining site having specificity for σv ?3 and another antigen-combining site having specificity for a different antigen. In such embodiments, the light chain is deleted and the Fv of the heavy chain is substituted with the desired polypeptide. These antibodies are termed bivalent or polyvalent, depending upon the number of immunoglobulin "arms" possessed by the Fc domain employed (IgMs will be polyvalent). Aside from the nonimmunoglobulins mentioned above, the antibody also is rendered multivalent by recombination of antibodies having more than one specificity. For instance, the antibody in some embodiments is capable of binding σvβ as described elsewhere herein but is also capable of binding a T-cell determinant such as CD3, CD4, CD8, CD 8, CD1 1 a, CD1 1 b or CD1 1 c. These other antibodies are well known. The multispecific, multivalent antibodies are made by cotransforming a cell with DNA encoding the heavy and light chains of both antibodies and the proportion of expressed antibodies having the desired structure recovered by immunoaffinity chromatography or the like. Alternatively, such antibodies are made from monovalent antibodies which are recombined in vitro in conventional fashion.
Monovalent antibodies also are made by techniques that are conventional per se. Recombinant expression of light chain and a modified heavy chain is suitable. The heavy chain is truncated generally at any point in the Fc region so as to prevent heavy chain crosslinking. Alternatively, the relevant cysteines are substituted with another residue or deleted so as to prevent crosslinking. In vitro methods also are used to produce monovalent antibodies, e.g., Fab fragments are prepared by enzymatic cleavage of intact antibody.
For diagnostic applications, the antibodies of the invention typically will be labeled with a detectable moiety. The detectable moiety can be any one which is capable of producing, either directly or indirectly, a detectable signal. For example, the detectable moiety may be a radioisotope, such as 3H, 14C, 3 P, S, or 12BI, a fluorescent or chemiluminescent compound, such as fluorescein isothiocyanate, rhodamine, or luciferin; radioactive isotopic labels, such as, e.g., 126l, 32P, 1 C, technicium, or 3H, or an enzyme, such as alkaline phosphatase, beta- galactosidase or horseradish peroxidase.
Any method known in the art for separately conjugating the antibody to the detectable moiety may be employed, including those methods described by Hunter, e$ al.. Nature 144:945 (1962); David, ej ah. Biochemistry 13:1014 (1974); Pain, e| al., J. Immunol. Meth. 40:219 (1981); and Nygren, J. Histochem. and Cytochem. 30:407 (1982). The antibodies of the present invention may be employed in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprecϊpitation assays. Zola. Monoclonal Antibodies: A Manual of Techniques, pp.147- 158 (CRC Press, Inc., 1987).
Competitive binding assays rely on the ability of a labeled standard (which may be σv 73 or an immunologically reactive portion thereof) to compete with the test sample analyte (σv ?3) for binding with a limited amount of antibody. The amount of avβ3 in the test sample is inversely proportional to the amount of standard that becomes bound to the antibodies. To facilitate determining the amount of standard that becomes bound, the antibodies generally are insolubilized before or after the competition, so that the standard and analyte that are bound to the antibodies may conveniently be separated from the standard and analyte which remain unbound.
Sandwich assays involve the use of two antibodies, each capable of binding to a different immunogeπic portion, or epitope, of the protein to be detected. In a sandwich assay, the test sample analyte is bound by a first antibody which is immobilized on a solid support, and thereafter a second antibody binds to the analyte, thus forming an insoluble three part complex. David & Greene, U.S. Pat No. 4,376,110. The second antibody may itself be labeled with a detectable moiety (direct sandwich assays) or may be measured using an anti-immunoglobulin antibody that is labeled with a detectable moiety (indirect sandwich assay). For example, one type of sandwich assay is an ELISA assay, in which case the detectable moiety is an enzyme.
The antibodies of the invention also are useful for ]n vivo imaging, wherein an antibody labeled with a detectable moiety such as a radio-opaque agent or radioisotope is administered to a host, preferably into the bloodstream, and the presence and location of the labeled antibody in the host is assayed. This imaging technique is useful in the staging and treatment of neoplasms or bone disorders. The antibody may be labeled with any moiety that is detectable in a host, whether by nuclear magnetic resonance, radiology, or other detection means known in the art. The neutralizing antibodies of the invention are especially useful in therapeutic applications, to prevent or treat unwanted bone resorption, or tumor cell growth or metastasis. Obviously, Mabs of the 10C4.1.3 type are not useful for treating or in vivo imaging of tumors of the same type described in Table 2 infra since they do not bind to σvβ3 found on such cells. Instead these Mabs are especially useful of treating conditions of bone resorption or degradation, for example as found in osteoporosis or resulting from PTHrP over- expression by some tumors.
For therapeutic applications, the antibodies of the invention are administered to a mammal, preferably a human, in a pharmaceutically acceptable dosage form. They are administered intravenously as a bolus or by continuous infusion over a period of time, by intramuscular, subcutaneous, intra-articular, intrasynovial, intrathecal, oral, topical, or inhalation routes. When the antibody possesses the suitable activity it is also suitably administered by intratumoral, peritumoral, intralesional, or perilesional routes, to exert local as well as systemic therapeutic effects.
Such dosage forms encompass pharmaceutically acceptable carriers that are inherently nontoxic and nontherapeutic. Examples of such carriers include ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffers such as phosphate or glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts, or electrolytes such as protamine sulfate, sodium chloride, metal salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulosic polymers, and polyethylene glycol. Carriers for topical or gel-based forms of antibody include polysaccharides such as sodium carboxymethylcelluiose or methylcellulσse, polyvinylpyrrolidone, polyacrylates, polyoxyethylene-polyoxypropylene-block polymers, polyethylene glycol, and wood wax alcohols. Conventional depot forms include, for example, microcapsules, nano-capsules, liposomes, plasters, -sublingual tablets, and polymer matrices such as polylactide:polyglycolide copolymers. When present in an aqueous dosage form, rather than being lyophilized, the antibody typically will be formulated at a concentration of about 0.1 mg/ml to 100 mg/ml, although wide variation outside of these ranges is permitted. For the prevention or treatment of disease, the appropriate dosage of antibody will depend on the type of disease to be treated, as defined above, the severity and course of the disease, whether the antibodies are administered for preventive or therapeutic purposes, the course of previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician. The antibody is suitably administered to the patient at one time or over a series of treatments.
Depending on the type and severity of the disease, about 0.015 to 15 mg of antibody/Kg of patient weight is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. For repeated administrations over several days or longer, depending on the condition, the treatment is repeated until a desired suppression of disease symptoms occurs. However, other dosage regimens may be useful and are not excluded herefrom.
According to another embodiment of the invention, the effectiveness of the antibody in preventing or treating disease may be improved by administering the antibody serially or in combination with another agent that is effective for the same clinical objective, such as another antibody directed against a different epitope than the principal antibody, or one or more conventional therapeutic agents known for the intended therapeutic indication, e.g. prevention or treatment of conditions associated with excessive bone resorption such as, osteoporosis or inhibition of tumor cell growth or metastasis. The antibodies of the invention also are useful as affinity purification agents. In this process, the antibodies against σv/53 are immobilized on a suitable support, such a Sephadex resin or filter paper, using methods well known in the art. The immobilized antibody then is contacted with a sample containing the avβ3 to be purified, and thereafter the support is washed with a suitable solvent that will remove substantially all the material in the sample except the αv/73, which is bound to the immobilized antibody. Finally, the support is washed with another suitable solvent, such as glycine buffer, pH 5.0, that will release the σv/73 from the antibody.
The following examples are offered by way of illustration only and are not intended to limit the invention in any manner. EXAMPLE 1
A. Generation of Mabs specific for human αvg3
To produce Mabs specific for avβ3 integrin, Balb/c mice were immunized with σvβ3 integrin purified from 293- 5D cell line expressing avβ3 complex which were generated by transfecting 293 cells (ATCC CRL1573) with DNAs prepared from PMNCV vector expressing av oτ β3 plus DNA coding for Neomycin resistance gene. avβ3 was purified from an NP 40 cell lysate of 293-15D cells by using a lentil lectin column. The purity of the σv ?3 prepared was then confirmed by isoelectrophoresis. Mice were immunized into foot pads once with 5 μg of avβ3 emulsified in MPL/TDM adjuvant (Ribi Immunochem. Research Inc., Hamilton, MT) and then subsequently six times with 5 mg of σv ?3 immersified in M PL/TDM adjuvant at 2 week intervals. Three days after the last immunization, lymph node cells from these mice were fused with P3X63Ag8U.1 myeloma cells (Yelton, et al., Curr. Top. Microbiol. Immunol. 81 : 1 , 1978) using 35% polyethylene glycol as described (Yarmush et al, Proc. Nat. Acad. Sci. 77:2899, 980). The remainder of the process is depicted in Fig. 1 . Hybridoma cell lines were selected for anti-σv ?3 antibody production by their ability to bind soluble σvβ3 by ELISA and to bind cell lines expressing various integrins by Flow microfluorometry analysis using FACSCAN (Becton Dickinson FACS systems. Mountain View, CA). Isotypes (Table 1 ) of these positive Mabs were determined by ELISA using isotype specific alkaline phosphatase- conjugated goat anti-mouse immunoglobulin (Harlow and Lane, Antibodies: A Laboratory Manual, p.597, Cold Spring Harbor Laboratory, 1988).
Table I Characteristics of Mabs
Figure imgf000013_0001
Positive hybridoma cell lines were subcloned twice by limiting dilution technique. B. Immune precipitation of crv/?3 complex with Mabs
293-15D transfectants grown F12/DMEM medium with 10% FCS were harvested by treatment with EDTA and biotinylated by using NHS-LC-Biotin. Cells (5 X 106 cells/ml) were incubated with 1 /sg/ml of NIH-LS-Biotin for 1 hour at room temperature. The unbound biotin was then removed by washing in 0.05 mM tris buffer containing 0.5 M NaCI m 1 mm. CaCI2 and 1 m N MgCI2 (cell wash buffer). Cells were lysed by treatment with 1 % NP-40 and cell debris were removed by microcentrifugation for 10 min. The supernatant was used for the immunoprecipitation. Fifty μ\ of Protein-G in 0.05 M tris buffer containing 0.5 M NaCI and 0.1 % Tween-20 (IP wash buffer) were incubated with 100 /.I of Mabs (100μg/ml) for 30 min at room temperature. After washing twice in IP wash buffer, nonspecific binding sites on Protein-G were blocked with 1 % BSA for 1 hr. at room temperature, washed twice and incubated with the supernatant containing biotinylated membrane proteins for 1 hr at room temperature. The complex was washed six times, reduced in SDS PAGE-sample buffer containing 2-ME by boiling and analyzed by electrophoresis using 12% SDS polyacrylamide gel. Fig. 2 depicts the results. EXAMPLE 2 Cell culture and Immunofluorescence stainino of various tissues
Transfected cells and tumor cells grown in F12/DMEM medium (1:1 o/o mixture) containing 10% FCS, glutamine and antibiotics were washed three times in the cell sorter buffer (CSB, PBS containing 1 % FCS and 0.01 % NaN3) by centrifugation at 1 ,000 rpm for 5 min and resuspended to be 4x10β cells/ml in CSB. Twenty five μ\ of cells were added into a 96 well U-bottom plate and incubated with 100 μ\ of antibodies for 30 min on ice. At the end of the incubation, cells were washed twice in CSB and the Mab bound onto cells were detected by incubating cells with FITC conjugated goat anti-mouse Ig antibodies for 30 min on ice. Cells were washed twice in CSB, resuspended in 0.5 ml of CBS, and analyzed by Flow mϊcrofluorometry as described (Loken, et al. Ann. N.Y. Acad. Sci. 254:163-; Miller, et al.. Rev. Sci. Iπstrum. 49:1 137-9, 1978). The results are shown in Table 2.
Table 2 Flow microfluorometrv analysis of mabs with various cells
3
Figure imgf000015_0001
FACS Results Graded: - (Mean FL Channel 0-10), + (Mean FL Channel 10-100), + + (Mean FL Channel 100-1000),
+ + + (Mean FL Channel > 1 ,000) Tissue Frozen Section (Osteoclastoma), tissue section results graded: + (weak), + + (moderate), + + + (strong).
Table 2 depicts the determination of the portion of avβ3 recognized by these Mabs by FACS analysis of transfected cells expressing different integrins as well as by ELISA using soluble integrin proteins. Mab 9G2.1.3 bound strongly to 293-15D expressing σv 73 and very weakly to 293-CLB expressing llb-llla, but not 293-52B expressing avβ3. Mab 10C4.1.3 only bound to 293-15D but not others. Therefore, it was concluded that these two Mabs (9G2.1.3 and 10C4.1.3) recognized avβ3. in contrast, Mab 9D4.9.1 strongly bound to both 293-15D as well as 293-CLB but not 293-52B. Thus, the Mab 9D4.9.1 was concluded to the bind β3 portion of avβZ.
Table 2 also shows that Mab 9C9.1 1.1 1 bound to osteoclasts, human endothelial cells and various melanoma cells. This staining pattern was similar to that of 23C6. In contrast, Mab 10C4.1.3 recognized only osteoclasts, suggesting a surprising and very narrow specificity. The staining pattern of Mabs 9D4.9.1 and 9G2.1.3 for human melanoma tumor cells, glioma cells and normal endothelial cells were similar to that of Mab 23C6: these Mabs recognized various human melanoma cells, and osteoclasts strongly, and human osteosarcoma MG-63 cells and human endothelial cells weakly. In contrast Mab 10C4.1.3 showed a strong binding to human osteoclasts and a weak binding to one of the human myeloma cells, M-21 , but no binding to other cells. These results suggest that 10C4.1.3 recognizes an epitope unique to human osteoclasts.
EXAMPLE 3 Determination of epitopes by competitive Immuπoflmorescence staining avβ3 transfected 293-15D cells (1X10B cells/1 OQμl) were incubated with 100 //I of the first purified Mab for 30 min on ice, washed twice and incubated with the second Mab, FITC conjugated Mab 23C6 for 30 min. At the end of the incubation, cells were washed twice in CSB and resuspended in 0.5 ml of CBS and the level of FITC conjugated Mab 23C6 binding on 293-15D cells was examined by flow microfluorometrγ (FACSCAN). The results are depicted in Fig. 3. Panels are as follows:
FL-23C6 (shaded area represents unstained cells) FL-23C6 FL-23C6 FL-23C6
FL-23C6
Figure imgf000016_0001
FL-23C6
Several Mabs which recognize the σ-chain of avβ3 designated as CD51 , or/9-chain of vitronectin receptors, CD61 , have been described (Nesbitt et al, 1991 , in "Leukocyte typing IV, p1037). More recent study (Horton, Int. J. EXP. Pathol.. 71 :741 [1990]) showed that Mab 23C6 and LM609, which were grouped as Mabs recognizing they? epitope of CD51 , may recognize the intact avβ3 complex. Thus to confirm that the Mabs binding to the σv ?3 complex recognize different epitopes from that of 23C6, the staining of 293-15D with f luoresceinated 23C6 was examined in the presence of 100 fold higher level of unlabeled Mabs and analyzed by FACSCAN. The results in Figure 3 showed that Mabs 9D4.9.1 and 10C4.1.3 did not interfere at all with the binding of fiuoresceinated-23C6 to 15D cells. In the same experiment, the same amount of unlabeled 23C6 completely inhibited the binding of F-23C6 while the irrelevant control Mab did not have any effect. Thus it was confirmed that at least these two Mabs indeed recognize epitopes different from those recognized by 23C6. In contrast, Mab 9G2.1 .3 at high concentration, has some inhibitory effect on the F- 23C6 binding but could not completely block the F-23C6 binding. Thus it is concluded that Mab 9G2.1.3 recognizes a different epitope from the one recognized by F-23C6, but these epitopes appear to be closely orientated. Since it has reported that LM 609 (Cheresh, et al., J. Biol. Chem.. 262:17703-1771 1 [1987]) recognizes the same epitope as 23C6 it is concluded that our Mabs recognized different epitopes from the one recognized by Mab LM 609. EXAMPLE 4
Inhibition of the binding of 293-15D cells to ligands (fibrinogen and vitronectin) bv Mabs. Microplates (NUNC, Breakapart C8 Maxi Sorp) were coated with 100 //l/well of 10 μglml of fibrinogen or vitronectin overnight at 4°C. After being washed three times in PBS, plates were blocked with 1 % BSA in PBS for 1 hr and then were incubated with various concentrations of Mabs for 30 min followed by the addition of 100 ml of 61Cr labeled 293- 15D cells. Plates were centrifuged at 600 rpm for 2 min and incubated for 90 min at 37°C. At the end of the incubation, plates were washed three times and 61Cr labeled 293-15D cells bound to the ligand were counted by a gamma counter. E1Cr labeled 293-15D transfectants expressing avβ3 were prepared as follows. Cells were grown in F12/DMEM medium containing 10% FCS, 0.1 % glucose and 2mM glutamine for 40 hr, were harvested by treatment with 10 mM EDTA in PBS for 2 min, washed twice in PBS and resuspended to be 5 x 107 cells/ml in culture medium without FCS. 0.5 ml of 293-15D cells were then incubated with 250 mCi B1Cr and for 1 hr at 37°C. At the end of the incubation, excess unbound 61Cr was removed by washing three times in F12/DMEM medium and resuspended to be 6 x 10ε cell/ml in culture medium without FCS. Fig. 4A-B depicts the results.
The top panel of Figure 4 shows the binding of 61Cr-293 15 D to fibrinogen coated wells in the presence of various concentration of Mabs. All three Mabs inhibit the binding of 61Cr-293 15 D to fibrinogen very effectively. The strongest inhibition was shown with Mab 9D4.9.1. The bottom panel shows the binding of 61Cr-293 15 D to vitronectin coated wells. Under the conditions tested, 9D4.9.1 and 10C4.1 .3 could inhibit this interaction but 9G2.1 .3 and 23C6 showed a very weak inhibition, if any. In general, it was harder to inhibit the interaction between σvβ3 transfected cells to vitronectin than the interaction between avβ3 transfected cells to fibrinogen.
EXAMPLE 6 Inhibition of αv_ff3 integrin binding to Fibrinogen and Vitronectin bv monoclonal antibodies.
Microtiter plates were coated with 100 .l/well of 10 μg/ml of purified fibrinogen or vitronectin, overnight at 4°C. After washing three times in PBS, the plates were blocked with
1 % BSA for 1 hour at room temperature. After washing the plate in PBS, 61Cr-293-15D cells preincubated with various concentrations of Mabs for 30 minutes on ice were transferred to the ligand coated plate. The plates were then centrifuged at 600 rpm for 2 minutes and incubated for 90 minutes at 37°C. At the end of the incubation, the plates were washed three times in PBS and the 61Cr labeled 293-15D cells bound to the ligand were counted by a gamma counter. The results, which parallel those in Example 4, are depicted in Figs. 5a and 5b.
EXAMPLE 7 Histochemical staining of frozen sections of human tissues and bone imprints Frozen sections of human osteoclastoma tumors and bone imprints from human fetal limb bones (14 weeks gestation), newborn rabbit and rat bone, embryonic chick bone and adult red deer antler, and from the following tissues of adult human origin (liver, kidney, pancreas, colon, ileum, heart, lung, thymus, tonsil, spleen, placenta, skin, uterine cervix, umbilical cord, breast carcinoma, malignant melanoma, smears of peripheral blood and bone marrow ononuclear cells) were prepared as described [Horton, M.A. etal., Cancer Res. 45. 5663-5669 (1985)]; these were air-dried, fixed in acetone for 10 minutes at room temperature and stored at -20°C until use. Slides were brought to room temperature, rehydrated in PBS then incubated with 150 μ\ of 1 % FCS/PBS containing 1//g of purified Mabs for one hour. After washing in 1 % FCS/PBS the slides were incubated sequentially with biotinylated anti-mouse Ig and then with avidin-biotin-horseradish peroxidase complex at the manufacturer/s recommended dilutions (Vector Lab, Burlingame, CA) for one hour. After further washes, the bound peroxidase was developed in 0.1 mg/ml diaminobenzidine tetrahydrochloride containing 0.07% H202 (Organon Teknina Corp. Durham, NC) in pBS and counter stained with 0.5% methyl green for 5 minutes. The slides were then dehydrated in graded alcohols, then cleared in xylene and mounted in permountant (Fisher Scientific Co., San Francisco, CA) for microscopy.
Figure 6 shows clear membrane staining of multinucleated osteoclasts from human bone imprints and frozen sections of giant cell tumor of bone using Mab 10C4.1.3; a control Mab did not show staining. Mabs 9D4.9.1 and 9G2.1.3 showed a similar staining pattern to Mab 10C4.1.3. None of the Mabs recognized osteoclast vitronectin receptor in conventional formalin-fixed and paraffin-embedded tissue sections.
We have examined the binding of these Mabs to osteoclasts present in bone imprints from rat, rabbit, chicken and deer in comparison to human.
Bone imprints were stained with mAbs followed by F-goat anti-mouse IgG. The level of fluorescence staining was examined by fluorescent microscopy and graded as weak ( +), moderate (+ +) and strong (+ + +) or absent (-).
Table 3 Determination of the mAb binding to osteoclasts from various species
Figure imgf000019_0001
Mab 9G2.1.3 recognizes osteoclasts from rabbit, chicken, deer in addition to human; this distribution is similar to that seen with 23C6 (Horton etal., 1985, oo. cit.). In contrast, Mabs 9D4.9.1 and 10C4.1.3 only recognized human osteoclasts; to date, no αvβ3 complex- specific Mabs have shown such species selectivity.
The distribution of the antigens recognized by the three Mabs was analyzed by immuπohistochemistry on frozen sections from the above-listed tissues of normal adult and fetal origin (data not shown). Mab 9D4.9.1 stained platelets (and megakaryocytes in bone marrow) intensely in all tissues studied. In addition, vascular endotheiium was stained, variably and weakly, in all tissues. Mab 9G2.1 .3 also stained vascular endotheiium, but failed to react with platelets and megakaryocytes. Both antibodies stained kidney (glomerulus, tubulest, hepatic sinusoids, colonic and ileal smooth muscle, placenta (cyto- and syncytiotrophoblasts) and neoplastic melanocytes in malignant melanoma. In contrast, Mab 10C4.1.3 failed to stain or gave a much weaker reaction in tissues recognized by Mab 9G2.1.3. For example, Mab 10C4.1.3 did not stain intestinal smooth muscle or placenta.
The antigenic specificities recognized by these Mabs were further investigated by examining their binding to various cell lines including human melanoma tumor cells, osteosarcoma cells and normal human umbilical vein endothelial cells (HUVEC) by flow microfluorometry. Mabs 9D4.9.1 and 9G2.1.3 bound strongly to various human melanoma cells and less to MG-63 human osteosarcoma cells and HUVEC. In contrast, Mab 10C4.1.3 bound weakly to only one of the human melanoma cell lines, M-21. These results further confirm that 10C4.1.3 recognizes a novel antigenic epitope.
The following antibody producing hybridomas have been deposited with the American Type Culture Collection, 12301 Parklawn Drive, Rockville, MD, USA (ATCC): Antibody ATCC Pep. No. Deposit Date
10C4.1.3 HB 1 1029 29 April 1992
9G2.1.3 HB 11030 29 April 1992
9D4.9.1 HB 11031 29 April 1992 These deposits were made under the provisions of the Budapest Treaty on the international Recognition of the Deposit of Microorganisms for the Purpose of Patent Procedure and the Regulations thereunder (Budapest Treaty). This assures maintenance of a. viable culture for 30 years from the date of deposit or for the enforceable life of the patent or for a period of five years after the last request or for the effective life of the patent, whichever is longer. The organisms will be made available by ATCC under the terms of the Budapest Treaty, and subject to an agreement between Genentech, inc. and ATCC, which assures permanent and unrestricted availability of the progeny of the culture to the public upon issuance of the pertinent U.S. patent or upon laying open to the public of any U.S. or foreign patent application, whichever comes first, and assures availability of the progeny to one determined by the U.S. Commissioner of Patents and Trademarks to be entitled thereto according to 35 USC §122 and the Commissioner's rules pursuant thereto (including 37 CFR §1.14 with particular reference to 886 OG 638).
The assignee of the present application has agreed that if the culture on deposit should die or be lost or destroyed when cultivated under suitable conditions, it will be promptly replaced on notification with a viable specimen of the same culture. Availability of the deposited strain is not to be construed as a license to practice the invention in contravention of the rights granted under the authority of any government in accordance with its patent laws. In respect of those designations in which a European patent is sought, a sample of the deposited microorganism will be made available until the publication of the mention of the grant of the European patent or until the date on which the application has been refused or withdrawn or is deemed to be withdrawn, only by the issue of such a sample to an expert nominated by the person requesting the sample. (Rule 28(4) EPC) Although the foregoing refers to particular preferred embodiments, it will be understood that the present invention is not so limited. It will occur to those ordinarily skilled in the art that various modifications may be made to the disclosed embodiments without diverting from the overall concept of the invention. All such modifications are intended to be within the scope of the invention.

Claims

CLAIMS:
I . An anti-σv ?3 monoclonal antibody composition capable of binding to substantially the same avβ3 epitope recognized by a monoclonal antibody selected from the group consisting of 10C4.1 .3, 9G2.1 .3 or 9D4.9.1 .
2. The monoclonal antibody composition of claim 1 capable of (1 ) binding osteoclasts, and (2) inhibiting the binding of σvβ3 expressing cells to fibrinogen.
3. The monoclonal antibody composition of claim 2 capable of inhibiting the binding of avβ3 expressing cells to vitronectin.
4. The monoclonal antibody composition of claim 2 recognizing the avβ3 complex.
5. The monoclonal antibody composition of claim 4 binding to an epitope recognized by monoclonal antibody 10C4.1.3 or 9G2.1.3.
6. The monoclonal antibody composition of claim 2 recognizing the β3 portion of the avβ3 molecule.
7. The monoclonal antibody composition of claim 6 binding to an epitope recognized by monoclonal antibody 9D4.9.1 and having an affinity for avβ3 equal to or greater than monoclonal antibody 9D4.9.1.
8. The monoclonal antibody composition of claim 2 that is substantially incapable of binding llbllla.
9. The monoclonal antibody composition of claim 2 that is substantially incapable of binding avβ3 expressing cells other than osteoclasts.
10. An isolated nucleic acid encoding an antibody of claim 1.
I I . A hybridoma cell which produces a monoclonal antibody of claim 1.
12. A hybridoma cell which produces a monoclonal antibody of claim 5.
13. A hybridoma cell which produces the monoclonal antibody of claim 7.
14. A hybridoma cell which produces a monoclonal antibody of claim 9.
15. The hybridoma cell deposited under American Type Culture Collection Accession Number ATCC HB 1 1029 (referred to herein as 10C4.1.3).
16. The hybridoma cell deposited under American Type Culture Collection Accession Number ATCC HB 1 1030 (referred to herein as 9G2.1.3).
17. The hybridoma cell deposited under American Type Culture Collection Accession
Number ATCC HB 11031 (referred to herein as 9D4.9.1 ).
18. A monoclonal antibody produced by the hybridoma cell of claim 15.
19. A monoclonal antibody produced by the hybridoma cell of claim 16.
20. A monoclonal antibody produced by the hybridoma cell of claim 17.
21 . A pharmaceutical composition comprising a therapeutically effective amount of an antibody of claim 1 .
22. An immunoassay comprising the steps of:
(a) contacting a test sample with a monoclonal antibody of claim 1 , and
(b) determining the amount of σvβ3 in the test sample that is bound to the monoclonal antibody.
23. A monoclonal antibody composition comprising a monoclonal antibody which is capable of binding avβ3 on osteoclasts but is substantially incapable of binding to avβ3 on human umbilical vein endotheiium.
24. The monoclonal antibody composition of claim 23 which is labelled with a detectable marker.
25. The monoclonal antibody composition of claim 23 which is covalently bonded to a water insoluble matrix.
26. The monoclonal antibody composition of claim 23 which is bispecific.
27. The monoclonal antibody composition of claim 26 wherein the antibody is specific for σv Z and for a T-cell marker.
28. The monoclonal antibody composition of claim 27 wherein the T-cell marker is CD3, CD4, CD8, CD18, CD11a, CD1 l b, or CDI Ic.
29. The monoclonal antibody composition of claim 23 which comprises an Fc domain having an immunoglobulin effector function.
30. The monoclonal antibody composition of claim 23 which is monovalent.
31. The monoclonal antibody composition of claim 23 which comprises an Fc domain of IgA, IgD, IgE, IgG or IgM.
32. The monoclonal antibody composition of claim 23 wherein the antibody is bound to a cytotoxin.
PCT/US1993/002987 1992-04-03 1993-03-30 ANTIBODIES TO ALPHAvBETA3 INTEGRIN WO1993020229A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
EP93908677A EP0633945B1 (en) 1992-04-03 1993-03-30 ANTIBODIES TO ALPHAvBETA3 INTEGRIN
US08/307,844 US5578704A (en) 1992-04-03 1993-03-30 Antibody to osteoclast alphavbeta3 ntegrin
JP51764493A JP3353209B2 (en) 1992-04-03 1993-03-30 Antibodies to αvβ3 integrin
AU39413/93A AU680411B2 (en) 1992-04-03 1993-03-30 Antibodies to alphavbeta3 integrin
CA002132091A CA2132091C (en) 1992-04-03 1993-03-30 Antibodies to alphavbeta3 integrin
DE69322860T DE69322860T2 (en) 1992-04-03 1993-03-30 ANTIBODIES AGAINST ALPHA V BETA 3 INTEGRIN

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US86267992A 1992-04-03 1992-04-03
US07/862,679 1992-04-03
US2591393A 1993-03-03 1993-03-03
US08/025,913 1993-03-03

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US2591393A Continuation-In-Part 1992-04-03 1993-03-03

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US08/432,618 Continuation US5652110A (en) 1992-04-03 1995-05-02 Antibodies to αvβ3 integrin

Publications (1)

Publication Number Publication Date
WO1993020229A1 true WO1993020229A1 (en) 1993-10-14

Family

ID=26700459

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1993/002987 WO1993020229A1 (en) 1992-04-03 1993-03-30 ANTIBODIES TO ALPHAvBETA3 INTEGRIN

Country Status (8)

Country Link
US (5) US5578704A (en)
EP (1) EP0633945B1 (en)
JP (1) JP3353209B2 (en)
AT (1) ATE175241T1 (en)
AU (1) AU680411B2 (en)
CA (1) CA2132091C (en)
DE (1) DE69322860T2 (en)
WO (1) WO1993020229A1 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994015958A2 (en) * 1993-01-08 1994-07-21 Tanabe Seiyaku Co., Ltd. Peptide inhibitors of cell adhesion
EP0719859A1 (en) * 1994-12-20 1996-07-03 MERCK PATENT GmbH Anti-alpha V-integrin monoclonal antibody
EP0754059A1 (en) * 1994-03-18 1997-01-22 The Scripps Research Institute Methods and compositions useful for inhibition of angiogenesis
WO1998040488A1 (en) * 1997-03-12 1998-09-17 Smithkline Beecham Corporation Anti-alphabeta3 humanized monoclonal antibodies
WO1998046265A1 (en) * 1997-04-11 1998-10-22 G.D. Searle & Co. Methods for using antagonistic anti-avb3 integrin antibodies
WO1998046264A1 (en) * 1997-04-11 1998-10-22 G.D. Searle & Co. Antagonistic anti-avb3 integrin antibodies
WO1999066910A2 (en) * 1998-06-24 1999-12-29 Peter Dietsch Agents for specifically inhibiting osteoclastic bone resorption
WO2000009143A1 (en) * 1998-08-13 2000-02-24 G.D. Searle & Co. Multivalent avb3 and metastasis-associated receptor ligands
US6056958A (en) * 1994-12-09 2000-05-02 Dupont Pharmaceuticals Method of treatment of arterial and venous thromboembolic disorders
WO2000034780A2 (en) * 1998-12-04 2000-06-15 Novartis Ag METHODS AND COMPOSITIONS USEFUL FOR TARGETING ACTIVATED VITRONECTIN RECEPTOR αvβ¿3?
WO2001017563A2 (en) * 1999-09-08 2001-03-15 Bayer Aktiengesellschaft Integrin-mediated drug targeting
EP1194173A1 (en) * 1999-04-29 2002-04-10 Vanderbilt University X-ray guided drug delivery
EP1219305A1 (en) * 2000-12-27 2002-07-03 Bayer Aktiengesellschaft Conjugates of integrin receptor antagonists and a cytostatic agent having specifically cleavable linking units
US6500924B1 (en) 1996-05-31 2002-12-31 The Scripps Research Institute Methods and compositions useful for inhibition of angiogenesis
US6833373B1 (en) 1998-12-23 2004-12-21 G.D. Searle & Co. Method of using an integrin antagonist and one or more antineoplastic agents as a combination therapy in the treatment of neoplasia
US7049140B1 (en) 1999-04-29 2006-05-23 Vanderbilt University X-ray guided drug delivery
US7163681B2 (en) 2000-08-07 2007-01-16 Centocor, Inc. Anti-integrin antibodies, compositions, methods and uses
US7220824B1 (en) 2000-08-28 2007-05-22 Bayer Aktiengesellschaft Integrin-mediated drug targeting
US7288390B2 (en) 2000-08-07 2007-10-30 Centocor, Inc. Anti-dual integrin antibodies, compositions, methods and uses
US7306925B2 (en) 2001-11-09 2007-12-11 Vanderbilt University Phage antibodies to radiation-inducible neoantigens
US9340581B2 (en) 2001-10-03 2016-05-17 Washington University Ligands to radiation-induced molecules
US10449261B2 (en) 2014-07-24 2019-10-22 Washington University Compositions targeting radiation-induced molecules and methods of use thereof

Families Citing this family (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5770198A (en) * 1988-05-18 1998-06-23 The Research Foundation Of The State Of New York Platelet-specific chimeric 7E3 immunoglobulin
US5169930A (en) * 1990-01-05 1992-12-08 La Jolla Cancer Research Foundation Fibronectin receptor
US7132260B2 (en) * 1991-04-05 2006-11-07 The General Hospital Corporation DNA encoding parathyroid hormone receptor
US7150974B1 (en) 1991-04-05 2006-12-19 The General Hospital Corporation Parathyroid hormone receptor binding method
ATE287898T1 (en) * 1991-04-05 2005-02-15 Gen Hospital Corp PARATHYROID HORMONE RECEPTOR AND ITS CODING DNA
AU6235294A (en) * 1993-02-02 1994-08-29 Scripps Research Institute, The Methods for producing polypeptide binding sites
US5817768A (en) * 1995-06-07 1998-10-06 The New York Blood Center, Inc. Monospecific antibodies against a subunit of fibrinogen
AU4134497A (en) * 1996-09-03 1998-03-26 Chugai Seiyaku Kabushiki Kaisha Anti-integrin alpha3 antibody complexes
DE69739295D1 (en) * 1996-12-09 2009-04-23 Merck Patent Gmbh Soluble, recombinant alphaV beta3 adhesion receptor
US20010011125A1 (en) * 1997-01-30 2001-08-02 William D. Huse Anti-alpha v beta 3 recombinant human antibodies, nucleic acids encoding same and methods of use
US6590079B2 (en) * 1997-01-30 2003-07-08 Ixsys, Incorporated Anti-αvβ3 recombinant human antibodies, nucleic acids encoding same
US6596850B1 (en) 1998-01-30 2003-07-22 Ixsys, Incorporated Anti-αv3β3 recombinant human antibodies, nucleic acids encoding same
US6210904B1 (en) * 1997-10-14 2001-04-03 Merck & Co., Inc. Anticoagulant test
US6531580B1 (en) * 1999-06-24 2003-03-11 Ixsys, Inc. Anti-αvβ3 recombinant human antibodies and nucleic acids encoding same
US6037176A (en) * 1999-06-25 2000-03-14 Isis Pharmaceuticals Inc. Antisense inhibition of integrin beta 3 expression
EP1223980B1 (en) * 1999-10-28 2003-05-21 Hofbauer, Reinhold Use of csf-1 inhibitors
US7740841B1 (en) 2000-01-28 2010-06-22 Sunnybrook Health Science Center Therapeutic method for reducing angiogenesis
DK1297016T3 (en) * 2000-05-12 2006-07-10 Vlaams Interuniv Inst Biotech Use of placental growth factor inhibitors to treat pathological angiogenesis, pathological arteriogenesis, inflammation, tumor formation and / or vascular leakage
MXPA03007878A (en) * 2001-03-02 2004-07-08 Medimmune Inc Methods of preventing or treating inflammatory or autoimmune disorders by administering integrin alphav beta3 antagonists.
US8658773B2 (en) 2011-05-02 2014-02-25 Immunomedics, Inc. Ultrafiltration concentration of allotype selected antibodies for small-volume administration
EP1487492A4 (en) * 2002-03-04 2008-10-01 Medimmune Inc The prevention or treatment of cancer using integrin alphavbeta3 antagonists in combination with other agents
JP2005525368A (en) * 2002-03-04 2005-08-25 メディミューン,インコーポレーテッド Method for preventing or treating disorders in which an integrin αvβ3 antagonist is administered in combination with an HMG-CoA reductase inhibitor or bisphosphonate
JP2005535572A (en) * 2002-04-12 2005-11-24 メディミューン,インコーポレーテッド Recombinant anti-interleukin-9 antibody
EP1517704A1 (en) * 2002-06-28 2005-03-30 Vlaams Interuniversitair Instituut voor Biotechnologie vzw. Use of antibodies against flt-1 for the treatment of osteoporosis
US20050186204A1 (en) * 2002-06-28 2005-08-25 Peter Carmeliet Use of antibodies against FLT-1 for the treatment of osteoporosis
CA2514653A1 (en) * 2003-01-30 2004-08-12 Medimmune, Inc. Uses of integrin alphavbeta3 antagonists
CA2521826C (en) * 2003-04-11 2013-08-06 Jennifer L. Reed Recombinant il-9 antibodies and uses thereof
WO2005044978A2 (en) * 2003-07-15 2005-05-19 Washington University Methods of treating, preventing and inhibiting cancer metastasis and tumor formation
US7271245B2 (en) * 2004-02-13 2007-09-18 The Scripps Research Institute Methods and compositions for inhibition of metastasis
US7351739B2 (en) * 2004-04-30 2008-04-01 Wellgen, Inc. Bioactive compounds and methods of uses thereof
US8187595B2 (en) 2004-05-07 2012-05-29 The University Of North Carolina At Chapel Hill Monoclonal antibodies for enhancing or inhibiting insulin-like growth factor-I
ES2343424T3 (en) * 2004-05-07 2010-07-30 The University Of North Carolina At Chapel Hill PROCEDURE TO POWER OR INHIBIT THE GROWTH FACTOR 1 SIMILAR TO INSULIN.
WO2005117978A2 (en) 2004-06-04 2005-12-15 Genentech, Inc. Method for treating multiple sclerosis
JP2008518023A (en) 2004-10-27 2008-05-29 メディミューン,インコーポレーテッド Regulation of antibody specificity by altering affinity for cognate antigens
JP5164829B2 (en) * 2005-03-24 2013-03-21 トロンボジェニクス・ナムローゼ・フエンノートシャップ Novel anti-PLGF antibody
US7867490B2 (en) * 2005-06-30 2011-01-11 Vib Vzw Treatment of liver cirrhosis and its complications
MY149159A (en) 2005-11-15 2013-07-31 Hoffmann La Roche Method for treating joint damage
WO2008011216A2 (en) * 2006-05-16 2008-01-24 Pro-Pharmaceuticals, Inc. Galactose-pronged polysaccharides in a formulation for antifibrotic therapies
EP2851064A3 (en) 2006-07-11 2015-08-05 Roy C. Levitt Rhinosinusitis prevention and therapy with proinflammatory cytokine inhibitors
WO2008016431A2 (en) * 2006-07-29 2008-02-07 Robert Lamar Bjork Bi-specific monoclonal antibody (specific for both cd3 and cd11b) therapeutic drug
WO2008021237A1 (en) 2006-08-10 2008-02-21 Arubor Corporation Localized therapy of lower airways inflammatory disorders with proinflammatory cytokine inhibitors
AR073295A1 (en) 2008-09-16 2010-10-28 Genentech Inc METHODS TO TREAT PROGRESSIVE MULTIPLE SCLEROSIS. MANUFACTURING ARTICLE.
JP5522717B2 (en) * 2008-09-19 2014-06-18 国立大学法人佐賀大学 Atopic dermatitis detection method and prophylactic / therapeutic agent screening method
AU2009299744B2 (en) 2008-10-02 2015-08-20 Life Sciences Research Partners Vzw Inhibition of PLGF to treat Philadelphia chromosome positive leukemia
WO2010075249A2 (en) 2008-12-22 2010-07-01 Genentech, Inc. A method for treating rheumatoid arthritis with b-cell antagonists
WO2010075058A1 (en) * 2008-12-23 2010-07-01 Ge Healthcare Limited Application of 99mtc peptide-based compound as a bone marrow imaging agent
CA2749501C (en) 2009-02-13 2017-01-10 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US8287865B2 (en) 2009-09-16 2012-10-16 Immunomedics, Inc. Class I anti-CEA antibodies and uses thereof
WO2011068845A1 (en) 2009-12-02 2011-06-09 Immunomedics, Inc. Combining radioimmunotherapy and antibody-drug conjugates for improved cancer therapy
EP2785739B1 (en) 2011-12-01 2017-03-15 ThromboGenics N.V. Improving trabeculectomy outcome
WO2013101771A2 (en) 2011-12-30 2013-07-04 Genentech, Inc. Compositions and method for treating autoimmune diseases
WO2013116287A1 (en) 2012-01-31 2013-08-08 Genentech, Inc. Anti-ig-e m1' antibodies and methods using same
EP2885002A4 (en) 2012-08-14 2016-04-20 Ibc Pharmaceuticals Inc T-cell redirecting bispecific antibodies for treatment of disease
MX2015002465A (en) 2012-08-31 2015-11-06 Univ North Carolina Monoclonal antibodies for enhancing or inhibiting insulin-like growth factor 1 (igf-1).
SI2900277T1 (en) 2012-12-13 2022-05-31 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and sn-38 for improved efficacy and decreased toxicity
WO2014130648A1 (en) 2013-02-20 2014-08-28 Galectin Therapeutics, Inc. Method for treatment of pulmonary fibrosis
CN106132436B (en) 2014-02-21 2021-06-15 Ibc药品公司 Disease therapy by inducing an immune response to TROP-2 expressing cells
CN106029098A (en) 2014-02-25 2016-10-12 免疫医疗公司 Humanized RFB4 anti-CD22 antibody
US9580495B2 (en) 2014-06-24 2017-02-28 Immunomedics, Inc. Anti-histone therapy for vascular necrosis in severe glomerulonephritis
JP6678941B2 (en) 2014-10-07 2020-04-15 イミューノメディクス、インコーポレイテッドImmunomedics, Inc. Use of neoadjuvant antibody-drug conjugates
JP6746845B2 (en) 2015-04-22 2020-08-26 イミューノメディクス、インコーポレイテッドImmunomedics, Inc. Isolation, detection, diagnosis and/or characterization of circulating TROP-2 positive cancer cells
CN114796501A (en) 2015-06-25 2022-07-29 免疫医疗公司 Methods of treating cancer with combinations of antibodies and therapeutic agents
SI3316885T1 (en) 2015-07-01 2021-09-30 Immunomedics, Inc. Antibody-sn-38 immunoconjugates with a cl2a linker
EP3359572A2 (en) 2015-10-06 2018-08-15 H. Hoffnabb-La Roche Ag Method for treating multiple sclerosis
EP3207937A1 (en) 2016-02-17 2017-08-23 Royal College of Surgeons in Ireland A method of treating or preventing sepsis
KR102101561B1 (en) * 2018-07-25 2020-04-20 가톨릭대학교 산학협력단 Angiogenesis target constrast medium and a method for producing the same
US20220133913A1 (en) * 2019-07-24 2022-05-05 Korea Basic Science Institute SINGLE DOMAIN ANTIBODY TARGETING aVB3 INTEGRIN
WO2022157336A1 (en) 2021-01-22 2022-07-28 Royal College Of Surgeons In Ireland Treatment of coronavirus
WO2022239720A1 (en) 2021-05-10 2022-11-17 公益財団法人川崎市産業振興財団 Antibody having reduced binding affinity for antigen

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989005155A1 (en) * 1987-11-19 1989-06-15 Scripps Clinic And Research Foundation Monoclonal antibody against the rgd-directed adhesion receptor of endothelial cells
US5057604A (en) * 1988-08-03 1991-10-15 Washington University Novel monoclonal antibodies
WO1992008739A1 (en) * 1990-11-13 1992-05-29 The Scripps Research Institute Antibodies that bind to a ligand-induced binding site on integrin and induce integrin activation

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4707352A (en) * 1984-01-30 1987-11-17 Enzo Biochem, Inc. Method of radioactively labeling diagnostic and therapeutic agents containing a chelating group
US5169930A (en) * 1990-01-05 1992-12-08 La Jolla Cancer Research Foundation Fibronectin receptor
AU6235294A (en) * 1993-02-02 1994-08-29 Scripps Research Institute, The Methods for producing polypeptide binding sites

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989005155A1 (en) * 1987-11-19 1989-06-15 Scripps Clinic And Research Foundation Monoclonal antibody against the rgd-directed adhesion receptor of endothelial cells
US5057604A (en) * 1988-08-03 1991-10-15 Washington University Novel monoclonal antibodies
WO1992008739A1 (en) * 1990-11-13 1992-05-29 The Scripps Research Institute Antibodies that bind to a ligand-induced binding site on integrin and induce integrin activation

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
CANCER RESEARCH vol. 45, 1985, PHILADELPHIA,USA pages 5663 - 5669 HORTON ET AL 'MONOCLONAL ANTIBODIES TO OSTEOCLASTOMAS (GIANT CELL BONE TUMORS):DEFINITION OF OSTEOCLAST-SPECIFIC CELLULAR ANTIGENS' cited in the application *
ISI ATLAS OF SCIENCE:IMMUNOLOGY vol. 1, no. 1, 1988, PHILADELPHIA,USA pages 35 - 43 HORTON 'OSTEOCLAST-SPECIFIC ANTIGENS' *
JOURNAL OF BIOLOGICAL CHEMISTRY vol. 262, no. 36, 1987, BALTIMORE,USA pages 17703 - 17711 CHERESH ET AL 'BIOSYNTHETIC AND FUNCTIONAL PROPERTIES OF AN ARG-GLY-ASP-DIRECTED RECEPTOR INVOLVED IN HUMAN MELANOMA CELL ATTACHMENT TO VITRONECTIN,FIBRINOGEN,AND VON WILLEBRAND FACTOR' cited in the application *
JOURNAL OF BONE AND MINERAL RESEARCH vol. 7, no. 3, March 1992, NEW YORK,USA pages 345 - 351 HELFRICH ET AL 'INTEGRINS ON RAT OSTEOCLASTS:CHARACTERIZATION OF TWO MONOCLONAL ANTIBODIES (F4 AND F11) TO RAT BETA-3' *
NESBITT ET AL 'LEUCOCYTE TYPING IV.WHITE CELL DIFFERENTIATION ANTIGENS EDS:KNAPP ET AL' 1989 , OXFORD UNIVERSITY PRESS , OXFORD *
TISSUE ANTIGENS vol. 33, 1989, COPENHAGEN page 357 NESBITT ET AL 'EPITOPE ANALYSIS OF THE VITRONECTIN RECEPTOR ALPHA AND BETA CHAINS' *

Cited By (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994015958A2 (en) * 1993-01-08 1994-07-21 Tanabe Seiyaku Co., Ltd. Peptide inhibitors of cell adhesion
WO1994015958A3 (en) * 1993-01-08 1994-09-29 Tanabe Seiyaku Co Peptide inhibitors of cell adhesion
EP0754059A4 (en) * 1994-03-18 1997-07-30 Scripps Research Inst Methods and compositions useful for inhibition of angiogenesis
US7595051B2 (en) 1994-03-18 2009-09-29 The Scripps Research Institute Methods of treating neovascular glaucoma, macular degeneration and capillary proliferation with alphavbeta3-specific antibodies
EP1410807A1 (en) * 1994-03-18 2004-04-21 The Scripps Research Institute Methods and compositions useful for inhibition of angiogenesis
US6887473B1 (en) 1994-03-18 2005-05-03 The Scripps Research Institute Inhibition of angiogenesis in disease states with an anti-αvβ3 monoclonal antibody
EP1468695A1 (en) * 1994-03-18 2004-10-20 The Scripps Research Institute Methods and compositions useful for inhibition of angiogenesis
US7354586B2 (en) 1994-03-18 2008-04-08 The Scripps Research Institute Methods of treating arthritis and diabetic retinopathy with αvβ3-specific antibodies
US7482007B2 (en) * 1994-03-18 2009-01-27 The Scripps, Research Institute Methods of inhibiting angiogenesis to treat cancer with alphavbeta3-specific antibodies
EP0754059A1 (en) * 1994-03-18 1997-01-22 The Scripps Research Institute Methods and compositions useful for inhibition of angiogenesis
US6056958A (en) * 1994-12-09 2000-05-02 Dupont Pharmaceuticals Method of treatment of arterial and venous thromboembolic disorders
KR100450368B1 (en) * 1994-12-20 2005-01-31 메르크 파텐트 게엠베하 Anti-αV-integrin monoclonal antibody
EP0719859A1 (en) * 1994-12-20 1996-07-03 MERCK PATENT GmbH Anti-alpha V-integrin monoclonal antibody
US5985278A (en) * 1994-12-20 1999-11-16 Merck Patent Gesellschaft Mit Beschrankter Haftung Anti-αV-integrin monoclonal antibody
CN1117763C (en) * 1994-12-20 2003-08-13 默克专利股份有限公司 Anti-aV-integrin monoclonal antibody
US6500924B1 (en) 1996-05-31 2002-12-31 The Scripps Research Institute Methods and compositions useful for inhibition of angiogenesis
EP1491632A3 (en) * 1997-03-12 2006-01-04 Smithkline Beecham Corporation Anti-alphavbeta3 humanized monoclonal antibodies
US7230083B2 (en) 1997-03-12 2007-06-12 Smithkline Beecham Corporation Anti-alphabeta3 humanized monoclonal antibodies
US7504102B2 (en) 1997-03-12 2009-03-17 Smithkline Beecham Corporation Anti-αvβ3 humanized monoclonal antibodies
EP1491632A2 (en) * 1997-03-12 2004-12-29 Smithkline Beecham Corporation Anti-alphavbeta3 humanized monoclonal antibodies
WO1998040488A1 (en) * 1997-03-12 1998-09-17 Smithkline Beecham Corporation Anti-alphabeta3 humanized monoclonal antibodies
US6193968B1 (en) 1997-04-11 2001-02-27 The Burnham Institute Methods for using anti-αvβ3 integrin antibody
US6171588B1 (en) 1997-04-11 2001-01-09 G. D. Searle & Company Anti-αvβ3 integrin antibody antagonists
WO1998046264A1 (en) * 1997-04-11 1998-10-22 G.D. Searle & Co. Antagonistic anti-avb3 integrin antibodies
WO1998046265A1 (en) * 1997-04-11 1998-10-22 G.D. Searle & Co. Methods for using antagonistic anti-avb3 integrin antibodies
WO1999066910A3 (en) * 1998-06-24 2000-06-15 Peter Dietsch Agents for specifically inhibiting osteoclastic bone resorption
WO1999066910A2 (en) * 1998-06-24 1999-12-29 Peter Dietsch Agents for specifically inhibiting osteoclastic bone resorption
WO2000009143A1 (en) * 1998-08-13 2000-02-24 G.D. Searle & Co. Multivalent avb3 and metastasis-associated receptor ligands
WO2000034780A3 (en) * 1998-12-04 2000-10-19 Novartis Ag METHODS AND COMPOSITIONS USEFUL FOR TARGETING ACTIVATED VITRONECTIN RECEPTOR αvβ¿3?
WO2000034780A2 (en) * 1998-12-04 2000-06-15 Novartis Ag METHODS AND COMPOSITIONS USEFUL FOR TARGETING ACTIVATED VITRONECTIN RECEPTOR αvβ¿3?
US6833373B1 (en) 1998-12-23 2004-12-21 G.D. Searle & Co. Method of using an integrin antagonist and one or more antineoplastic agents as a combination therapy in the treatment of neoplasia
US7049140B1 (en) 1999-04-29 2006-05-23 Vanderbilt University X-ray guided drug delivery
US7875454B2 (en) 1999-04-29 2011-01-25 Vanderbilt University X-ray guided drug delivery
EP1194173A1 (en) * 1999-04-29 2002-04-10 Vanderbilt University X-ray guided drug delivery
EP1194173A4 (en) * 1999-04-29 2003-05-14 Univ Vanderbilt X-ray guided drug delivery
WO2001017563A2 (en) * 1999-09-08 2001-03-15 Bayer Aktiengesellschaft Integrin-mediated drug targeting
WO2001017563A3 (en) * 1999-09-08 2002-07-11 Bayer Ag Integrin-mediated drug targeting
US7163681B2 (en) 2000-08-07 2007-01-16 Centocor, Inc. Anti-integrin antibodies, compositions, methods and uses
US7288390B2 (en) 2000-08-07 2007-10-30 Centocor, Inc. Anti-dual integrin antibodies, compositions, methods and uses
US8071729B2 (en) 2000-08-07 2011-12-06 Centocor, Inc. Anti-integrin antibodies, compositions, methods and uses
US8071333B2 (en) 2000-08-07 2011-12-06 Centocor, Inc. Anti-integrin antibodies, compositions, methods and uses
US7550142B2 (en) 2000-08-07 2009-06-23 Centocor, Inc. Anti-integrin antibodies, compositions, methods and uses
US7220824B1 (en) 2000-08-28 2007-05-22 Bayer Aktiengesellschaft Integrin-mediated drug targeting
WO2002051444A1 (en) * 2000-12-27 2002-07-04 Bayer Aktiengesellschaft Conjugates of integrin receptor antagonists and a cytostatic agent having specifically cleavable linking units
EP1219305A1 (en) * 2000-12-27 2002-07-03 Bayer Aktiengesellschaft Conjugates of integrin receptor antagonists and a cytostatic agent having specifically cleavable linking units
US9340581B2 (en) 2001-10-03 2016-05-17 Washington University Ligands to radiation-induced molecules
US10086073B2 (en) 2001-10-03 2018-10-02 Washington University Ligands to radiation-induced molecules
US8012945B2 (en) 2001-11-09 2011-09-06 Vanderbilt University Phage antibodies to radiation-inducible neoantigens
US7306925B2 (en) 2001-11-09 2007-12-11 Vanderbilt University Phage antibodies to radiation-inducible neoantigens
US8927288B2 (en) 2001-11-09 2015-01-06 Vanderbilt University Phage antibodies to radiation-inducible neoantigens
US10449261B2 (en) 2014-07-24 2019-10-22 Washington University Compositions targeting radiation-induced molecules and methods of use thereof

Also Published As

Publication number Publication date
DE69322860D1 (en) 1999-02-11
CA2132091C (en) 2008-09-16
DE69322860T2 (en) 1999-07-01
AU680411B2 (en) 1997-07-31
ATE175241T1 (en) 1999-01-15
US5652110A (en) 1997-07-29
EP0633945B1 (en) 1998-12-30
JPH07505528A (en) 1995-06-22
EP0633945A1 (en) 1995-01-18
US5652109A (en) 1997-07-29
CA2132091A1 (en) 1993-10-14
AU3941393A (en) 1993-11-08
US5578704A (en) 1996-11-26
US6359126B1 (en) 2002-03-19
US6369204B1 (en) 2002-04-09
JP3353209B2 (en) 2002-12-03

Similar Documents

Publication Publication Date Title
US5652110A (en) Antibodies to αvβ3 integrin
Hubbard et al. Identification of rat hepatocyte plasma membrane proteins using monoclonal antibodies.
Kramer et al. Human microvascular endothelial cells use beta 1 and beta 3 integrin receptor complexes to attach to laminin.
US7060271B2 (en) Inhibitory immunoglobulin polypeptides to human PDGF beta receptor
CA2140538C (en) Monoclonal antibodies that block ligand binding to the cd22 receptor in mature b cells
US5599790A (en) Fibrinogen γ chain polypeptide and compositions thereof
CA2103893A1 (en) Monoclonal antibodies against receptor-induced binding sites
Ravdin et al. Production of mouse monoclonal antibodies which inhibit in vitro adherence of Entamoeba histolytica trophozoites
EP0369816A2 (en) Monoclonal antibodies specific for human polymorphic epithelial mucin
US5580780A (en) Vascular adhesion protein-(VAP-1) and VAP-1-specific antibodies
EP0726914B1 (en) Cell surface protein expressed on human cortical thymocyte and their use
US5320942A (en) Method of diagnosing the presence of abnormal epithelial tissue using monoclonal antibodies to the A6 B4 cell surface protein
EP0229794B1 (en) Monoclonal antibodies to gamma interferon, their production and use
Kunicki et al. The Pla Alloantigen System Is a Sensitive Indicator of the Structural Integrity of the Amino-terminal Domain of the Human Integrin β3 Subunit
EP0190049B1 (en) Method for selecting hybridomas producing antibodies specific to inaccessible cell surface antigens
EP0644768B1 (en) Methods and compositions for inhibiting endothelial cell and fibrinogen mediated inflammation
AU609382B2 (en) Monoclonal antibodies to terminal deoxynucleotidyl transferase
Le Guern et al. A monoclonal antibody directed against a synthetic peptide reacts with a cell surface rabbit class I MHC molecule
HUBBARD et al. Identification of Rat Membrane Proteins
EP0478272A2 (en) Gp98 cell adhesion molecule and integrin complex containing it

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP US

AL Designated countries for regional patents

Kind code of ref document: A1

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

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: 2132091

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 08307844

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 1993908677

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1993908677

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 1995 432618

Country of ref document: US

Date of ref document: 19950502

Kind code of ref document: A

Ref document number: 1995 432542

Country of ref document: US

Date of ref document: 19950502

Kind code of ref document: A

WWG Wipo information: grant in national office

Ref document number: 1993908677

Country of ref document: EP