CA2259371A1 - Therapeutic multispecific compounds comprised of anti-fc.alpha. receptor antibodies - Google Patents

Therapeutic multispecific compounds comprised of anti-fc.alpha. receptor antibodies Download PDF

Info

Publication number
CA2259371A1
CA2259371A1 CA002259371A CA2259371A CA2259371A1 CA 2259371 A1 CA2259371 A1 CA 2259371A1 CA 002259371 A CA002259371 A CA 002259371A CA 2259371 A CA2259371 A CA 2259371A CA 2259371 A1 CA2259371 A1 CA 2259371A1
Authority
CA
Canada
Prior art keywords
binding
antigen
receptor
binding molecule
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002259371A
Other languages
French (fr)
Inventor
Yashwant M. Deo
Robert Graziano
Tibor Keler
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
ER Squibb and Sons LLC
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2259371A1 publication Critical patent/CA2259371A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/33Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Clostridium (G)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/14Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from fungi, algea or lichens
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against Fc-receptors, e.g. CD16, CD32, CD64
    • 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
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell

Abstract

Multispecific compounds comprising at least one binding determinant which binds to the Fcx-receptor on an effector cell. The other binding determinant(s) binds(s) to one or more antigens on a target cell, e.g., the Neu/Her-2 proto-oncogene product or the epidermal growth factor receptor on cancer cells, or to Candida antigens on infected cells. Examples are biospecific and trispecific antibodies. Therapeutic use of said multispecific compounds for treatment of cancers or pathogen infections.

Description

CA 022 ~ 9371 I s s s - o l - o ~
Wo 98/02463PCT/US97/12013 Therapeutic Multispecific Compounds Comprised of Anti-Fca Receptor Antibodies Background of the Invention 5Receptors for the Fc portions of immunoglobulins are important in triggering many of the protective functions of monocytes, macrophages and polymorphonuclear cells. Receptors for IgG (Fcy receptors or Fc~R) on these cells have been extensively investigated and bispecific molecules targeting these receptors have been constructed. (See e.~. European Patent No. 0 255 249 entitled l'Monoclonal 10 Antibodies to Fc Receptor for Immunoglobulin G on Human Mononuclear Pha~ocytes".
which is co-owned by Applicants.) In addition, clinical trials of bispecific molecules (BsAb) which have specificity for the Fc~R and the HER-2/neu anti~en, which is found on breast or ovarian cancers. indicate that these molecules are both safe and efficacious (Valone, Frank H. et al. 1995- J of Clin. Oncol. 13(9): 2281 -2292).
15IgA receptors Fca receptors (FcaR or CD89) are also capable of promoting effector cell function. Binding of ligand to FcaR triggers phagocytosis and antibody mediated cell cytotoxicity in leukocytes and FcaR-bearin~ cell lines. Fca receptors can also cooperate with receptors for I~G on effector cells in enhancing the phagocytosis of target cells. Monoclonal antibodies of the IgM (Shen. L. el al., 1989 J
lmmunol. 143: 4117) and IgG (Monteiro, R.C. et al.. 1992 J. Immunol~ 148: 1 764) classes have been developed against FcaR.
IgA is abundant in the human body (Kerr, M.A. 1990. Biochem. J
271 :285-296). A single class of IgA Fc receptor. FcaRl or CD89~ which binds to monomeric IgA has been identified and characterized (Albrechtsen, M. et al.~ 1988 Immunol. 64:201;MonteiroR.~etal., 1990J. Exp. Med, 171:597). FcaRIis constitutively expressed primarily on cytotoxic immune effector cells including monocytes, macrophages~ neutrophils, and eosinophils (Morton, H.C.. et al.~ 1996Critical Reviews in Immunology 16:423). FcaRI expression on a sub-population of Iymphocytes (Morton, H.C.~ et al., I g96 Critical Reviews in Immunology 16:423). and on glomerular mes~n~i~3l cells has been reported (Gomez-Guerrero, C.~ et al. . 1996 J.
Immunol. 156:4369-4376). Its expression on monocytes and PMN can be enhanced b~
TNF-a (Gesl, A., et al., 1994 Scad. J. Immunol. 39:151-156; Hostoffer~ R.U'., et al 1994, The J. Infectious Diseases 170:82-87)~ IL-1. GM-CSF. LPS or phorbol esters(Shen L.. et al., J. Immunol. 152:4080-4086; Schiller, C.A. et ul., 1994. Immunolog~, .
81 :598-604), whereas IFN-~ and TGF-~I decrease FcaRI expression (Reterink, T.J.F.. et al., 1996, Clin. Exp. Immunol. 103: 161 - 166). The o~-chain of human ~caRI is a heavil~

CA 022~9371 1999-01-0~
WO 98102463 PCT/USg7/I2013 glycosylated, type one transmembrane molecule belonging to the Ig super-gene family which also includes receptors for lgG and I~E. One gene located on chromosome 19encodes several alternatively spliced isoforms of the FcaRI alpha chain (55- 110 kDa;
Morton, H.C.~ et al., 1996 Critical Revie-vs in Immunology 16:423). Mvelocytic FcaRI
5 has been shown to be associated with the FcR ~-chain which is implicated as playing a role in FcaRI signal transduction (Morton, H.C. et al. 1995, J. Biol. Chem. 270:29781;
Pfefferkorn. L.C., el al. 1995~ J. Immunol. 153:3228-3236~ Saito. K. e~ al.~ 1995, J
Allergy Clin. Immunol. 96: 1152).
FcaRI binds both antigen-complexed and monomeric I~AI and IgA2 (Mazangera, R.L. et c~l., 1990 Biochem. J. 272:159-165)~ consistent with the receptor being saturated in vivo with monomeric IgA in the same manner as Fc yR and ~c~RI are saturated with IgG and IgE respectively. Cross-linking FcaRI on myeloid effector cells~
by polymeric IgA, IgA immune complexes~ or mAb specific for epitopes within or outside the ligand binding domain, stimulates degranulation. superoxide release~15 secretion of infl~mmzltory cytokines. endocytosis and phagocytosis (Patty~ C., A.
Herbelin~ A. Lihuen~ J.F. Bach? and R.C. Monteiro. 1995 Imn?UnOIo~,J. 86:1-5; Stewart.
W.W., R.L. Maz Yegera? L. Shen, and M.A. Kerr. 1994 J. Leucocyle Biolog~. 56:481 -487; Stewart~ W.W., and M.A. Kerr. 1990. Immunology. 71 :328-334; Shen~ L. 1992. J.
Leukocyte Biology. 51 :373-378.). These physiological responses triggered via FcaRI can 20 be important in the first line of humoral defense on mucosal surfaces (Morton~ H.C.. M.
van Egmond~ and J.G.J. van de Winkel. 1996 C~ritical Revie~s in ImmunologJ,. 16:423).
Thus FcaRI is a clinically relevant trigger receptor on cytotoxic immune effector cells and its activity can be exploited to develop novel immunotherapies. The cytotoxic potential of FcaRI has not been carefully explored since almost all monoclonal 25 antibody (mAb) based therapies are being developed with mAbs of IgG class which do not bind to FcaRI.

Summary of the Invention The present invention relates to multispecific therapeutic molecules with 30 binding deterrninzlnt.~ for immunoglobulin A (IgA) receptors. IgA is the predominant antibody class in fluids on mucosal surfaces. and IgA receptors (Fca receptors~ FcaR or FcaRI) are found on white blood cells including macrophages. monocytes. neutrophils.
eosinophils and Iymphocytes. The bispecific and multispecific molecules of the invention can be used as therapeutic agents to harness the cytolysis and phagocytosis 35 capabilities of these white blood cells. enhancing the attack of these cells against cancer cells, cells of infectious microorg~nism~. and cells infected with patho~ens.

.... . ..

CA 02259371 l999-01-0~

In one aspect~ the invention includes bispecific binding molecules~
comprising a first binding determinant which binds an Fco~ receptor and a secondbinding determinant which binds one or more target antigens. Preferably~ the first determinant binds a site on the Fco~R that is different from the binding site for S endogenous IgA, so that binding of the molecules of the invention is not blocked or is not substantially blocked by IgA. In a preferred embodiment~ the target antigen bound by the second binding deterrninant of the bispecific molecules of the invention is a cancer cell antigen. In a more preferred embodiment the cancer cell antigen is an antigen of a cancer of the breast. ovary, testis~ lung~ colon, rectum, pancreas~ liver, central nervous system, head and neck. kidney, bone, blood or Iymphatic system. In another preferred embodiment, the target antigen is an infectious disease antigen from a pathogen or patho~en-infected cell. In yet another embodiment, the invention features treatment of an autoimmune disease with a composition that binds and modulates areceptor for IgA~ causing modulation of the receptor such that further binding of IgA to that receptor is decreased. In a different embodiment~ the invention provides compositions that bind and do not modulate a receptor for IgA, so that the effector cells of a subject are arrned with the bispecific and multispecific molecules and can bind an antigen on a pathogen or on a cancer.
A preferred embodiment of bispecific molecules of the subject invention comprise molecules with binding determinants for a receptor of the human EGF-like receptor family, for a carcinoembryonic antigen~ for a gastrin releasing peptide receptor antigen, and for a mucine antigen, which are overexpressed by certain tumor cells.
The bispecific molecules of the invention encompass molecules that are comprised in pan of binding determinants of antibodies, and the molecules of theinvention include those that are engineered to include at least one antibody or an antibody fragment. The bispecific binding molecules of the invention preferably comprise a binding determinant from an IgG antibody or IgG fragment, including an Fab, Fab'~ F(ab')2. Fv, and single chain Fv. A binding deterrninant, including an Fab, Fab', F(ab')2. Fv, and single chain Fv, can be obtained also from an IgA antibody or an antibody of another isotype. A preferred bispecific binding molecule of the invention comprises a first binding determinant that is at least a functional fragment of antibody A77 and a second binding determinant that binds a cancer cell antigen~ a pathogen antigen, or an antigen on an infected cell. Other preferred bispecific binding molecules of the invention comprise a first binding deterrninant that is at least a functional fragment of antibody A3, A59 or A6~, which are similar to A77 in affinity to thereceptor for IgA and are not blocked by IgA of the subject. The invention includes CA 022~9371 l999-ol-o~

nucleic acid sequences encoding the VH and V~ regions of the A77 antibod~ and the predicted amino acid sequences of these regions~ and these sequence are preferably used for hllm:~ni7ing the A77 binding determinants for therapeutic multispecific molecules.
Preferably the second binding determinant of the molecules of the invention is at least a functional fragment of antibody 520C9, antibody H425 or antibody CC49. A preferred embodiment carries one binding determinant for FcaR and one for the HER/nel~ antigen found for example on tumors of the breast ovary~ and lung.
Several methods of producing bispecific binding molecules are encompassed by the invention including by chemical linkage of the bindinP
10 determinants~ and by recombinant genetic methods. Recombinant bispecific molecules encoded by nucleic acid sequences carrying genes encoding binding deterrninants which are thus ~enetically linked are encompassed by the invention. Further~ bispecific binding molecules of the invention are produced by cell fusion of two antibody-producing cell lines carrying the respective nucleic acid sequences encoding the bindino 15 determinants~ such as hybridoma cell lines~ to obtain a progeny cell line producing the bispecific molecule of the invention.
In addition to bispecific binding molecules~ the instant invention encompasses multispecific binding molecules which comprise at least a first binding deterrninant which binds an Fca receptor and a second binding determinant which binds 20 a target antigen~ and at least a third binding determinant. Binding of the first determinant of these multispecific binding molecules to FcaR is not blocked or inhibited by human immunoglobulin A, so there is little or no competition for bindin,~ by endogenous IgA molecules. Multispecific binding molecules encompass bispecific and trispecific compositions, and those with four or more binding determinants. A preferred 25 embodiment of a trispecific binding molecule carries an additional binding determinant that binds to an Fc receptor that is not an Fca receptor, including for example a binding determinant for CD2, CD3, Fcy receptor, Fc~ receptor~ Fc~ receptor and/or Fcll receptor~
these determinants being in addition to the first binding determinant to Fc(x receptor.
The most preferred embodiment of an additional binding determinant for an FcR is a 30 determinant for Fcy receptor. For a multispecific binding molecule of the invention carrying a binding determinant for an Fcy receptor. binding to FcyR is not inhibited bv human IgG, since the molecule binds Fcy at a different epitopic site from IPG binding of FcyR. By incorporating at least binding deterrninant for each of FcaR and FcyR into a single molecule~ the therapeutic capability of the molecule is increased to enhance 35 affinity and kinetics of binding of white blood cells to tumor cells or cells of pathogenic CA 022~9371 1999-ol-0~

organisms or pathogen-infected cells. increasing opportunities for cytolysis andphagocytosis of these targets.
Another preferred embodiment of the multispecific binding molecules with a determinant for Fc~, is a molecule that carries a third binding determinant that 5 binds to a second target antigen or a second target epitope on a cancer cell, a pathogen.
or a pathogen-infected cell. The preferred means of producing these molecules is by chemical linkage of the binding determinants~ however the invention encompasses also multispecific binding moJecules which are recombinantly produced~ or which are produced by cell fusion of two or more cell lines each of which carries the nucleic acid 10 sequences encoding the binding determinants. Preferably at least one binding determinant is an antibody or an antibody fragment, and to improve the success of the outcome during continued treatment of humans. the binding determinant is a humanized antibody~ which is engineered to minimi7~ the number of foreign epitopes born by the molecule.
A preferred embodiment of the multispecific binding molecules of this invention is comprised of one or more binding determinants for target cancer cell antigens, particularly cancer cell antigens from breast. ovary, testis, lung, colon, rectum, pancreas, liver, central nervous system. head and nec}~. kidney. bone. blood andIymphatic system cancers. A different preferred embodiment of the invented 20 multispecific binding molecules comprises. as the target antigen, infectious disease antigens from pathogens or pathogen-infected cells. In one embodiment? the target antigens are infectious disease antigens and antigens expressed on infected cells~ for example. antigens from infections by bacteria, fungi, protozoa. and viruses. ln a more preferred embodiment. the target antigen is from a pathogenic fungus. including an 25 antigen from a pathogenic yeast. In a most preferred embodiment, the target antigen is from a species of Candida. for example, Candida albicans.
Suitable targets among cancer cell antigens are preferably members of the human EGF-like receptor family. more preferably the cancer cell antigen is an EGF
receptor, and most preferably the cancer cell antigen is HER-2/neu~ HER-3. HER-4. or a 30 heteromultimeric receptor comprised of at least one HER subunit. Additional preferred cancer cell antigens include carcinoembryonic antigen, gastrin releasing peptide receptor antigen, and TAG 72.
A most preferred multispecific binding molecule comprises at least a first binding determinant that is at least a functional fragment of antibody A77 and a second 35 binding determinant that binds an antigen of a cancer celJ. a cell of a pathogenic org~ni~m, or a pathogen-infected cell. In l~le~.ed embodiments of A77-derived , ., _ . . .

CA 022~9371 l999-01-0~
WO 98/02463 PCT/US97tl2013 multispecific binding molecules with a cancer antigen binding determinant. the preferred second binding determinant is at least a funclional fragment of antibod~ 520C9 or antibody CC49. The first binding determinant for an Fca receptor preferably binds a receptor on a white blood cell. The types of white blood cells to which the molecules 5 bind are preferably macrophages~ monocytes, neutrophils. eosinophils. and Iymphocytes.
Yet another aspect of the invention comprises multispecific binding molecules in which the molecule includes at least one antigen from a pathogen orpathogen-infected cell, or an antigen from a cancer cell. The molecules of this particular embodiment can serve to deliver these antigens as a vaccine directly to the antigen 10 presenting cells of the immune system to immunize the recipient against an infectious disease or a cancer. These antigens can be taken from known antigenic protein sequences of bacteria, viruses~ fungi and protozoans. and from cells infected with these pathogen. or from cancer cells. to immunize the recipient.
The multispecific binding molecules of the invention comprise bindino 15 determin~nt.~ from antibody or antibody fragment molecules which preferably are IgG or IgG fragments. Antibody fragments are preferably Fab. Fab'~ F(ab')2~ F(ab')3~ Fv, or single chain Fv as sources of binding determinants for construction of the multispecific binding molecules. Antibody fragments can be obtained from an IgG isotype class of antibody~ for example from a hybridoma producing a monoclonal IgG antibody~ or from 20 a polyclonal IgG preparation. In a preferred embodiment, a polyclonal IgG preparation is obtained from an animal that has been immunized with an antigen from a pathogenic org~ni~m, more preferably from a pathogenic fungus.
Another feature of multispecific binding molecules in which the first binding det~rmin~nt binds FcaR and the second binding determinant binds an anti~en of 25 a target cell, encompasses a third binding determinant which binds to a different antigen on the same target cell as the second binding determinant. Further, embodiments encompass a third binding determinant which binds to a different epitope on the same target antigen as the second binding determinant. These deternnin:~nt~ provide a two-fold binding capacity of the multispecific molecule to the target to link it to an immune 30 effector cell, for cytolysis and phagocytosis.
The invention also provides a method for elimin~ting an unwanted cell in a subject~ comprising ~lmini~tering to the subject a therapeutically effective dose of a multispecific binding molecule~ which comprises at least a first binding determinant which binds an Fca receptor and a second binding determinant which binds an antigen 35 on the unwanted cell, in a pharrnaceutically acceptable carrier. For human subjects~ the binding determinants derived from an antibody can be hllm~ni7~ d. Further. therapeutic CA 022~9371 l999-ol-o~

treatment can be monitored hy obtaining a biological sample from the subject during the course of treatment. Another embodiment of the method f'or elimin~ling unwanted cells in a subject involves treating the subject in addition with an agent that enhances the number or activity of Fca receptors~ for example~ treating the subject with a cytokine.
5 Preferred cytokines for a-lmini.~tration during treatment with the invented composition treatment include at least one of G-CSF. GM-CSF, IFN-~, and TNF~ and protocols involving treatment with the molecules of the subject invention and more than one additional therapeutic agent are envisioned.
In another embodiment, the effector cells of the subject can be armed 10 against an antigen, by administering to the subject a therapeutically effective dose of a multispecific binding molecule, which comprises at least a first binding determinant which binds an Fca receptor and a second binding determinant which binds the antigen.
in a pharmaceutically acceptable carrier. The effector cells thus armed with themultispecific molecule do not modulate or down-regulate the Fca receptors on their 15 surface, and are capable of binding to a target antigen. For human subjects. the bindinc determinants derived from an antibody can be hllm~ni7Pd. Further~ therapeutic treatment can be monitored by obtaining a biological sample from the subject during the course of treatment.
Another preferred embodiment of the invention is a method for 20 elimin~ting an unwanted cell in a subject~ comprising obtaining an aliquot of a sample of blood or blood cells from said subject, treating said blood or blood cells ex vivo with a therapeutically effective dose of a multispecific binding molecule of the invention in a pharm~eeutically acceptable carrier, said binding molecule comprising a first binding deterrninant which binds an Fca receptor and a second binding determinant which binds 25 one or more target antigens, and returning said treated blood or blood cells to the subject.
Preferably, the cells of the sample of blood are isolated and expanded in culture~ and more preferably~ the cells of said sample of blood are treated with agents that enhance the number or activity of Fca receptors. For human subjects, the binding determin~nts derived from an antibody can be hllm~ni7ed Further, therapeutic treatment can be30 monitored by obtaining a biological sample from the subject during the course of treatment.
In an aspect, the invention provides a method for treatment of a subject with an infectious disease. comprising a-lmini.~tration to the patient of a therapeuticall~
effective dose in a pharmaceutically acceptable carrier of a multispecific binding 35 molecule, wherein a first binding determinant binds an Fca receptor and a second CA 022~9371 1999-01-0~

binding determinant binds a target antigen from a pathogen or a pathogen-infected cell~
enhancing the capacity of the immune system to elimin~te the infection.
In yet another embodiment, the invention provides a method for immunizing a subject against a cancer antigen or an antigen found on a pathogen or a cell infected by a. comprising a-lmini~tration in a pharmaceutically acceptable carrier of a composition of a multispecific binding agent bearing one or more antigens of apathogenic infectious organism, or of an antigen of infected cells~ or of a cancer cell A
preferred embodiment of an infectious organism is a pathogenic fungus. including a pathogenic yeast; a more preferred embodiment is a species of Candida. For humansubjects, the binding deterrninants derived from an antibody can be h~lm~ni7~d. Further, therapeutic treatment can be monitored by obtaining a biological sample from thesubject during the course of treatment.
The invention provides also a method for identifying for an a,~ent which modulates Fca receptors on the surface of cells~ involving contacting a sample of cells carrying Fca receptors with the agentA method for identifying for an agent whichmodulates Fca receptors on the surface of cells~ comprising contacting a sample of cells carrying Fca receptors with the agent and determining Fca receptor activity in the sample with the agent. and in a control sample with an antibody that modulates Fca receptors such as antibody A77? and in another control sample with cells not contacted with said agent or with antibody; then Fca receptor activities in the samples are compared. such that a sample of cells contacted with the agent having statistically significant less Fca receptor activity than control cells not contacted with agent, or having statistically significantly as low Fca receptor activity as cells in a sample with the antibody, identifies an agent which modulates Fca receptors on the surface of cells.
In another embodiment~ the invention provides a method for designing an agent which modulates Fca receptors for treatment of autoimmune disease by obtaining a three dimensional model of the A77 anti-Fca receptor binding site using the sequence deterrninants of A77 heavy and light chain variable regions. This method involves comparing the amino acid residues of the A77 variable region with that of heavy and light chain variable regions of antibodies of known three dimensional structure,determining placement of non-homologous amino acid residues within the main peptide chain of the binding region of the VH and VK sites, such that the size. shape and charge of the A77 anti-Fca receptor binding site is determined, screening a library of molecules to obtain those of suitable size. shape and charge by computer modeling that aremimetics of the A77 binding site, and screening such candidates of ~plo,oliate size, CA 022~9371 l999-ol-o~

shape and charge for activity as potential modulators of Fco~ receptors. such that an a~ent which modulates Fca receptors is designed.

Brief Description of the Drawings Figure I is a gel filtration HPLC elution profile of a A77X520C9 bispecific molecule p,ep~dlion, in which the peak at 9.86 min. (-I SOKD) represents a F(ab')3 heterocomplex comprising one A77 F(ab') and two 520C9 F(ab ) molecules, and the peak at 10.43 min. represents a F(ab~)2 heterocomplex comprising one F(ab') molecule each of A77 and 520C9; the gel filtration HPLC was perforrned using a TSK-3000 column.
Figure 2 is a graph showing the extent of binding of A77X520C9. an anti-FcaRXanti-HER2/neu bispecific antibody (BsAb), as a function of concentration in ~g/ml. to neutrophils (PMN, open squares) and to monocytes (closed squares). in which mean fluorescence intensity analyzed by FACScan is the measure of binding.
Figure 3 is a cytometric analysis showing the extent of binding of A77X520C9, an anti-FcaRXanti-HER2/neu BsAb (at 10 llglml, final concentration) to effector cells in heparinized whole blood, aRer incubation for one hour at 0~C;
phycoerythrin-anti mouse IgG was added, erythrocytes were Iysed, and samples were analyzed by FACScan; data show substantial binding to monocytes (middle panel) and neutrophils (bottom panel), but not Iymphocytes (top panel).
Figure 4 is a graph showing binding of A77X520C9, the anti-FcaRXanti-HER2/neu BsAb (diamonds), and binding of another BsAb, MDX210~ also carrying a determinant for HER2/neu (open squares), to target SKBR-3 breast tumor cells, as a function of BsAb concentration in ~lg/ml~ in which mean fluorescence intensity analyzed by FACScan is the measure of binding.
Figure 5 is a graph showing binding to target SKBR-3 breast tumor cells of a further yl~p~dlion of A77X520C9, an anti-FcaRXanti-HER2/neu BsAb~ as a function of concentration in ~lg/ml; SKBR-3 cells were incubated with various concentrations of A77X520C9 (circles), A77 F(ab')2 (triangles), or 520C9 F(ab')2(squares) on ice; cells were washed, stained with anti-mouse IgG conjugated withfluorescein iso-thiocyanate (FITC), and analyzed by FACScan.
Figure 6 is a graph showing A77XH425 BsAb binding to A431 target cells overexpressing EGF-R, in which mean fluorescence intensity analyzed by FACScan is the measure of binding; cells were incubated with various concentrations of A77XH425 (circles), A77F(ab')2 (triangles), or with H425 F(ab')2 (squares); cells were . . .

CA 022~9371 l999-ol-o~

incubated with antibody as indicated on ice, washed, stained with anti-human IgGconjugated with FITC and analyzed by FACScan Figure 7 is a graph showing A77X520C9 BsAb-mediated antibody-dependent cellular toxicity by cell lysis of SKBR-3 breast tumor target cells by5 neutrophil effector cells. at a ratio of effector to target cells of 200 to one, as a function of BsAb concentration in ~lglml, with two different preparations of BsAb.
Figure 8 is a graph showing A77X520C9 BsAb-mediated antibody-dependent cellular toxicity by cell Iysis of SKBR-3 breast tumor target cells by effector cells in whole blood, using independent duplicate BsAb preparations. as a function of BsAb concentration in ~Lg/ml.
Figure 9 is a bar graph showing inhibition by anti-Fc(xR A77 F(ab')2 o~
A77X520C9 BsAb-mediated killing of breast tumor cells by monocytes~ compared to absence of inhibition by addition of anti-FcyR M22 F(ab')2.
Figure 10 is a graph showing A77X520C9 BsAb-mediated cytotoxicity by Iysis of target SKBR-3 cells by effector PMN cells purified from whole blood; target SKBR-3 cells were labeled with 10011Ci of 51 Cr for I hour prior to combining with PMNs and another preparation of BsAb indicated here as "BSM" (circles); and thismixture of target cells, PMNs and BsAb in the presence of 50~g/ml of A77 F(ab')2(squares); and this mixture in the presence of M22 F(ab')2 (triangles)~ each in a 'U
bottom microtiter plate, cell and antibody mixtures were incubated for 16 hours at 37~C~
supernatants were collected and analyzed for radioactivity, and cytotoxicity wascalculated by the formula: % Lysis = (experimental CPM-target leak CPM / detergent Iysis of CPM target leak CPM) X100%. BsAb-dependent Iysis = % Lysis with BsAb -% Lysis without BsAb. with an effector:target ratio of 200: 1; error bars represent +/- the standard deviation of the mean of values obtained from triplicate experiments.
Figure 11 is a graph showing A77X520C9 BsAb-mediated cytotoxicity by Iysis of target SKBR-3 cells, using as effector cells monocytes purified fromLeukopaks. with an effector:target ratio of 100:1; other conditions and symbols were the same as those in the description of Figure 10.
Figure 12 is a graph showing A77X520C9 BsAb-mediated cytotoxicity by Iysis of target SKBR-3 cells~ using the effector cells in whole blood. other conditions and symbols were the same as those in the description of Figure 10.
Figure 13 is a graph showing cytoxicity of A431 target cells that overexpress EGF-R, mediated by A77XH425 BsAb (indicated here as "BSM"; circles) and PMN effector cells purified from whole blood; cells were incubated also with BsAb in the presence of 50 ~g/ml of A77 F(ab')2 (squares) or M22 F(ab')2 (triangles); target CA 022~937l l999-Ol-o~

cells were labeled with 100~Ci of 51 Cr for I hour and cytotoxicity was determined as in the brief description of Figure 10.
Figure 14 is a graph showing A77XH425 BsAb mediated Iysis of A431 target cells by monocytes purifled from whole blood by Leukopaks (E:T ratio 100: 1);
5 other conditions and symbols were the same as those in the brief description of Figure 13.
Figure 15 is a graph showing A77XH425 BsAb mediated Iysis of A431 target cells by the effector cells in whole blood; other conditions and symbols were the same as those in the brief description of Figure 13.
I o Figure 16 is a graph showing Anti-TAG 72 x A77 BsAb-mediated antibody-dependent cellular cytoxicity by neutrophils of TAG 72-bearing tumor cells~ as a function of BsAb antibody concentration in llg/ml.
Figure 17 is a graph showing stimulation by the BsAb composition A77XTT of growth of tetanus toxoid (TT)-specific T cells~ measured by a spectorphotometric assay of cellular lactate dehydrogenase~ by monocyte presentation of this antigen by the BsAb~ compared to that of uncomplexed TT, as a function of concentration of each antigen in llg/ml.
Figure 18 is a graph showing the extent of A77X520C9 BsAb-mediated phagocytosis~ as a function of concentration in ,ug/ml, of SKBR-3 breast tumor target cells at a ratio of 15 neutrophils per target cell (circ~es), or 3 monocytes (squares) per target cell.
Figure 19 is a flow cytometric analysis of BsAb-mediated phagocytosis of SKBR-3 cells by monocyte-derived macrophages (MDM): SKBR-3 target cells were labeled with the lipophilic red fluorescent dye, PKH 26~ and cultured with MDM in the absence or presence of A77XS20C9 BsAb (or control antibodies) at 37~C for 24 hours;
MDM and non-phagocytized tumor cells were recovered with trypsin. and were stained with a FITC labeled anti-CD14, and samples were analyzed by two color fluorescence by FACScan. Percent phagocytosis was calculated as the number of dual-positive target cells (ingested by MDM! divided by the total number of target cells.
Figure 20 is a graph showing phagocytosis of SKBR-3 cells with A77X520C9 BsAb, in which specific phagocytosis of SKBR-3 cells was induced by A77X520C9 BsAb (circles) as compared to the presence of both un-coupled F(ab')o fragments of A77 and 520C9 (triangles). The BsAb-mediated phagocytosis was blocked by the addition of 10 ~g/ml A77 F(ab')2 (squares). BsAb-dependent phagocytosis was 3~ calculated as: % phagocytosis with BsAb - % phagocytosis without BsAb.

CA 022~9371 l999-ol-o~

Figure 21 is a photographic and flow cytometric illustration of neutrophil-mediated phagocytosis of the fungal pathogen. Candida albicans, in the presence of A77Xanti-Candida (A77Xa-Candida) BsAb; panels A and B are representative photomicrographs. and panels C and D are FACScan analyses. of neutrophils that were 5 mixed with C albicans cells in the absence (panels A and C) and presence (panels B and D) of the BsAb.
Figure '72 is a graph showing the extent of BsAb-mediated killing. as a function of concentration in ~lg /ml, of SKBR-3 breast tumor target cells by neutrophils treated with G-CSF (top panel). or with G-CSF and IFN-y (bottom panel).
Figure 23 is a graph showing the extent of BsAb-mediated killing. as a function of concentration in ~lglml. of SKBR-3 breast tumor target cells by effector cells which are IFN-y-treated monocytes (top right panel)~ by TNF-treated monocvtes (bottom panel). or b,v untreated monocytes (top leR panel).
Figure 24 is a bar graph showing that pre-incubation of effector cells with human IgA does not affect binding of A77 mAb, compared to a control incubated in the absence of IgA
Figure 25 is a bar graph showing modulation of Fca receptors by incubation of cells with A77 mAb at concentrations ranging from 0.001 to 10 llg/ml~
showing extent of change in number of cell surface receptors as a function of incubation with increasing A77 concentration, in comparison with control mAb 520C9 at 10 ~lg/ml Figure 26 is a bar graph showing that BsAb binding to monocytes or to PMN effector cells does not mediate FcaR modulation: modulation of FcaR after A77X520C9. A77 F(abl)2 or A77 mAb binding to PMN and monocytes was examined by flow cytometry, with various concentrations of antibodies added directly to whole blood and incubated overnight at 37~C in medium containing 5%C02. Erythrocytes were lysed. and the surface level of FcaR expression on PMN and monocytes was deterrnined by incubation with an anti-huIgA-PE probe at 4~C Modulation was calculated as: [ I -(MFI of sarnplelMFI of no antibodylBSM control~] x 100%.
Figure 27 shows the DNA sequence of the light chain variable Vlc re ion of the gene encoding the A77 anti-FcaR antibody (SEQ ID NO: 5). and the predicted arnino acid residue sequence (SEQ ID NO: 6).
Figure 28 shows the DNA sequence of the heavy chain variable VH
region of the gene encoding the A77 anti-FcaR antibody (SEQ ID NO: 7), and the predicted amino acid residue sequence (SEQ ID NO: 8).

, .

CA 022~9371 Isss-ol-o~

Detailed Description of the lnvention I Definitions Definitions of the terms and phrases as used herein should have the 5 meanings indicated below. An antibody (or fra~ment thereof~ is used in the invention as a component of multispecific agents which cause association of a cytolytic, phagocytic white blood cell with a tumor cell, or unwanted infectious disease agent or infected cell.
Antibodies suitable for use in the methods of the invention are available in the art (e.g.~
from the American ~ype Culture Collection, l~ockville, MD? or commercially, e.g.~ from 10 Becton-Dickinson or Immunotech) or can be prepared by standard techniques formaking antibodies. The term "antibody" as used herein refers to immunoglobulin molecules and immunologically active determinants of immunoglobulin molecules~ i.e..
molecules that contain an antigen binding site which specifically binds (immunoreacts with) an antigen. Structurally, the simplest naturally occurring antibody (e.g.~ IgG) 15 comprises four polypeptide chains, two copies of a heavy (H) chain and two of a light (L) chain~ all covalently linked by disulfide bonds. Specificity of binding in the large and diverse set of antibodies is found in the variable (V) determinant of the H and L
chains; regions of the molecules that are primarily structural are constant (C) in this set.
Structurally. a naturally occurring antibody (e.g.~ IgG, Mr 150 kDa) 20 consists of four polypeptide chains. two copies of a heavy (H) chain and two of a light (L) chain (Mr 25 kDa), the four chains being covalently linked by disulflde bonds.
Specificity of antigen binding by each molecule that comprises the large and diverse set of antibodies is found in the variable (V) determinant of the H and L chains: regions of the molecules that are primarily structural are constant (C) in this set. IgA (Mr 160 kDa) 25 is the predominant antibody in secretions (saliva~ tears~ mild~ nasal mucus and gastrointestinal and respiratory secretions). IgA can exist as a monomer. dimer, and higher multimeric forms (J. Kendrew, Ed., The Encyclopedia of Molecular Biology,1994, Blackwell Science, Oxford). A further isotype is IgM can be found as a membrane-bound monomer (Mr 190 kDa) on B cells~ and as a circulating secretory form 30 as a pentamer (Mr ca. 950 kDa) that differs from the bound form at the N-terminus of the heavy chain due to a~ternative splicing. The heavy chain of the pentamer is attached by disulfide bridges to a J chain molecule. Minor antibody isotypes include IgD (Mr 175 kDa)~ expressed on cell surfaces, and IgE. IgE (total Mr 190 kDa) comprises 0.0003%
of serum immunoglobulin, however it can be substantially elevated in an allergic subject 35 such as an asthmatic~ and is medically important as the major mediator of immediate type hypersensitivity.

CA 022~937l l999-Ol-o~

The binding sites of the proteins that comprise an antibody. i.e.. the antigen-binding functions of the antibody. are localized by analysis of fragments of a naturally-occurring antibody. Thus~ antigen-binding fragments are also intended to be designated by the terrn "antibody." Examples of binding fragments encompassed v.~ithin 5 the term antibody include: a Fab fragment consisting of the VL. VH, CLand C~l domains; an Fd fragment consisting of the VH and CH I domains; an Fv fragment consisting of the VL and VH domains of a single arm of an antibody; a dAb fragment (Ward et al.~ 1989 Nature 341 :544-546 ) consisting of a VH domain; an isolated complementarity determining region (CDR); and an F(ab')~ fragment~ a bivalent 10 fragment comprising two Fab' fragments linked by a disulfide bridge at the hinge region.
These antibody fragments are obtained using conventional techniques well-known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies. The term "antibody" is further intended to include bispecific and chimeric molecules having at least one antigen binding determinant derived from an 15 antibody molecule. Furthermore. although the H and L chains of an Fv fragment are encoded by separate genes~ a synthetic linker can be made that enables them to be made as a single protein chain (known as single chain antibody, sAb; Bird et al. 198~ Science 242:423-426; and Huston et al. 1988 PNAS 85:5879-5883) by recombinant methods.
Such single chain antibodies are also encompassed within the term "antibody". and may 20 be utilized as binding determinants in the design and engineering of a multispecific binding molecule. Antibody fragments are also useful for modulating the number of receptors for that antibody on the surface of cells, and for obtaining agents that mimic this activity. by screening for such agents in an assay for modulation of the receptor.
Polyclonal antibodies are produced by immunizing animals. usually a 25 m~mm~l, by multiple subcutaneous or intraperitoneal injections of an immunogen (antigen) and an adjuvant as appropriate. As an illustrative embodiment~ animals are typically immunized against a protein, peptide or derivative by combining about I !lg to 1 mg of protein capable of eliciting an immune response, along with an enhancingcarrier plepaldlion. such as Freund's complete adjuvant. or an aggregating agent such as 30 alum, and injecting the composition intradermally at multiple sites. Animals are later boosted with at least one subsequent ~1mini.~tration of a lower amount, as 1/5 to 1110 the original amount of immunogen in Freund's complete adjuvant (or other suitable adjuvant) by subcutaneous injection at multiple sites. Animals are subsequently bled.
serum assayed to determine the specific antibody titer. and the animals are again boosted 35 and assayed until the titer of antibody no longer increases (i.e., plateaus).

CA 022~9371 1999-01-0~
wo 98/02463 PCT/USg7/12013 Such populations of antibody molecules are referred to as "polyclonal"
because the population comprises a large set of antibodies each of which is specific for one of the many differing epitopes found in the immunogen, and each of which is characterized by a specific affinity for that epitope. An epitope is the smallest 5 deterrninant of antigenicity. which for a protein. comprises a peptide of six to eight residues in length (Berzofsky, J. and I. Berkower, (1993) in Paul, W.~ Ed., Fundamental ImmunologY, Raven Press, N.Y., p.246). Antibody affinities for the antigen range from low, e.g. 1()-6 M. to high. e.g., lo-l 1 M. The polyclonal antibody fraction collected from nlzlmm~ n serum is isolated by well known techniques, e.g. by chromatography 10 with an affinity matrix that selectively binds immunoglobulin molecules such as protein A. to obtain the I~G fraction. To enhance the purity and specificity of the antibody. the specific antibodies may be further purified by immunoaffinity chromatography using solid phase-affixed immunogen. The antibody is contacted with the solid phase-af'fixed immunogen for a period of time sufficient for the immunogen to immunoreact with the 15 antibody molecules to form a solid phase-affixed immunocomplex. Bound antibodies are eluted from the solid phase by standard techniques. such as by use of buffers of decreasing pH or increasing ionic strength, the eluted fractions are assayed. and those containing the specific antibodies are combined.
The term "monoclonal antibody" or "monoclonal antibody composition"
20 as used herein refers to a preparation of antibody molecules of single molecular composition. A monoclonal antibody (mAb) composition displays a single binding specificity and affinity for a particular epitope. Monoclonal antibodies can be prepared using a technique which provides for the production of antibody molecules by continuous growth of cells in culture. These include but are not limited to the 25 hybridoma technique originally described by Kohler and Milstein (1975, Nature256:495-497; see also Brown et al. 1981 J Immunol 127:539-46; Brown et al., 1980. J
Biol Chem 255:4980-83; Yeh et al., l 976, PNAS 76:2927-31; and Yeh et al. ., 1982. Int.
J. Cance) 29:269-75) and the more recent human B cell hybridoma technique (Kozbor e al., 1983. Immunol Today 4:72), EBV-hybridoma technique (Cole et al. .. 1985, 30 Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96), and trioma teclmiques.
Tumor specific mAb of human IgA class are not available. Also. it is likely that serum IgA (up to 4.0mg/ml) may interfere with the activity of IgA mAbs under physiological conditions. Another approach employs bispecific antibody 35 molecules to enable FcaRI-dependent cell-mediated cytotoxicity of tumor targets.
Bispecific molecules (BsAb) which simultaneously bind to tar~et cells (tumor cells.

~ , . . .

CA 022~9371 1999-01-0~

pathogens) and a trigger receptor (e.g. CD3~ CD~. FcyR) on immune effector cells have been described (Michon. J.. et al. 1995, Blo~l, 86~ 4-1130: Bakacs. T., ef ~JI. 1995.
Inlernarional Immunology~ 7,6:947-955) BsAbs can be generated from hetero-hybridomas~ or by chemically or genetically linking F(ab') fragments of two antibodies 5 with different specificities or a F(ab') fragment and a ligand (Graziano. R.F.. et cll. 1995.
In Bispecific Antibodies M.W. Fanger. editor R.G. Landes Company/Austin. TX;
Goldstein. J. et al., 1997 J. Immunol. 158:872-879). BsAbs produced using a trigger receptor-specific antibody. that binds outside the natural ligand binding domain of the trigger receptor, can circumvent interference by serum antibodies and recruit immune 10 effector cells in the presence of saturating concentration of the natural ligand (Fanger M. et al., 1989~ Immunol. Today, 10,3:~2-99) This strategy has been used to produce Fc rR-speciflc BsAbs, which mediate antibody-dependent cellular cytotoxicity (ADCC) of tumor cells in the presence of monomeric or aggregated lgG (Michon. J.. et al. 1995.
Blood, 86:112q-1130: Bakacs, T.?et al. 1995, International Immunology, 7.6:947-955)~
15 and have shown promising results in clinical settings, Deo. Y.M., et al., 1997. Immunol.
Tod~a~, 18:127-135. Four FcaRI-specific mAb. identifled as A3, A59, A62 and A77,which bind FcaRI outside the lgA ligand binding domain. have been described (Monteiro, R.C. et al., 1992, J. Immunol. 148:1764).
A monoclonal antibody can be produced by the following steps. In all 20 procedures, an animal is immunized with an antigen such as a protein (or peptide thereof) as described above for preparation of a polyclonal antibody. The immunization is typically accomplished by a~1ministering the immunogen to an immunoloc~icallycompetent m~mm~l in an immunologically effective amount. i.e., an amount sufficient to produce an immune response. Preferably, the m~mm~l is a rodent such as a rabbit. rat 25 or mouse. The m~mm~l is then m~inl~ined on a booster schedule for a time period sufficient for the m~mm~l to generate high affinity antibody molecules as described. A
suspension of antibody-producing cells is removed from each immunized m~mm~l secreting the desired antibody. After a sufficient time to generate high affinity antibodies, the animal (e.g., mouse) is sacrificed and antibody-producing lymphocytes 30 are obtained from one or more of the Iymph nodes, spleens and peripheral blood. Spleen cells are preferred. and can be mechanically separated into individual cells in a physiological medium using methods well known to one of skill in the art. The antibody-producing cells are immortalized by fusion to cells of a mouse myeloma line Mouse Iymphocytes give a high percentage of stable fusions with mouse homologous35 mvelomas, however rat, rabbit and frog somatic cells can also be used. Spleen cells of the desired antibody-producing ~nim~l~ are immortalized by fusing with myeloma cells.

CA 022~9371 1999-01-0~

.~vIXI-064CPPC ~ 1~/ ~ ~~~ ~~~~~~-: . .

generally in the presence of a fusing agent such as polyethylene glycol. Any of a number of myeloma cell lines suitable as a fusion partner are used with to standard techniques, for example. the P~-NSI/I-Ag4-1~ P3-x63-Ag8.653 or Sp2/O-Agl4 myeloma lines, available from the American Type Culture Coliection (ATCC)~
5 Rockville, Md.
The fusion-product cells, which include the desired hybridomas~ are cultured in selective medium such as HAT medium, designed to eliminate unfused parental myeloma or Iymphocyte or spleen cells. Hybridoma cells are selected and are grown under limiting dilution conditions to obtain isolated clones. The supernatants of 10 each clonal hybridoma is screened for production of antibody of desired specificity and affinity, e.g., by immunoassay techniques to deterrnine the desired antigen such as that used for immunization. Monoclonal antibody is isolated from cultures of producing cells by conventional methods, such as ammonium sulfate precipitation~ ion exchan_e chromatography, and affinity chromatography (Zola et al., Monoclonal Hybridoma Antibodies: Techniques And Applications, Hurell (ed.), pp. 51 -52, CRC Press, 198~).
Hybridomas produced according to these methods can be propagated in culture in vitro or in ~ivo (in ascites fluid) using techniques well known to those with skill in the art.
For therapeutic use of antibodies of non-human origin in hurnans. the non-human "forelgn" epltopes ehclt Immune response In the patlent. If sufficlentlv ~0 developed, a potentially lethal disease known as HAMA (human antibodies against mouse antibody) may result. To elimin~te or minimi7e HAMA, it is desirable to engineer chimeric antibody derivatives, i.e., "hllm~ni7ed" antibody molecules that combine the non-human Fab variable region binding determinants with a human constant region (Fc). Such antibodies are characterized by equivalent antigen specificity and affinity of monoclonal and polyclonal antibodies described above, and are less imrnunogenic when ~mini.~tered to humans, and therefore more likely to be tolerated by the patient.
Chimeric mouse-human monoclonal antibodies (i.e., chimeric antibodies) can be produced by recombinant DNA techniques known in the art. For example, a gene encoding the Fc constant region of a murine (or other species) monoclonal antibody molecule is digested with restriction enzymes to remove the region encoding the murine Fc, and the equivalent portion of a gene encoding a human Fc constant region is substituted. (see Robinson e~ al., International Application WO 87/02671; Akira, et al., European Patent Application 184,187; Taniguchi, M., European Patent Application -- 35 171,496; Morrison et al., European Patent Application 173,494; Neuberger et al - International Application WO 86/01533; Cabilly et al. U.S. Patent No. 4,816,567;
__ SUBSTITUTE PAGE
AMEN~ED SHEE~

CA 022~9371 1999-01-0~
.~XI-064CPPC

Cabilht et al., European Patent Application I '5~0'3: Better el al. 1988 Science240:1041-1043); Liu et al. 1987 P~ S 84:3439-3443; Liu e~ al., 1987, J Immunol.
I ~9:3521 -3526; Sun et al. 1987 PAAS 84:' 14-218: Nishimura et al. ., 1987, Canc. Res.
47:999-1005; Woodetal. (1985).~ature314:446-449;andShawetal.. 1988,J. hatl Cancer Inst. 80: 1553- 1559.) The chimeric antibody can be further hllm~ni7ed by replacing sequences of the Fv variable region which are not directly involved in antigen binding with equivalent sequences from human Fv variable regions. General reviews of h~ ni7edchimeric antibodies are provided by Morrison, S. L., 1985, Science 229:1202-1207 and by Oi et al., 1986, BioTechniques 4:214. Those methods include isolating, manipulating, and expressing the nucleic acid sequences that encode all or part of immunoglobulin Fv variable regions from at least one of a heavy or light chain. Sources of such nucleic acid are well known to those skilled in the art and, for example, may be obtained from 7E3, an anti-GPIIbIIIa antibodv producing hybridoma. The recombinant DNA encoding the chimeric antibody. or fragment thereof, can then be cloned into an appropriate expression vector. Suitable hllm~ni7ed antibodies can alternatively be produced by CDR substitution U.S. Patent 5,225.539; Jones et al. 1986 Nature 3 ~ I :5~2-5'5; Verhoeyan et al. . 1988 Science 239: 1534; and Beidler et al. 1988 J Immunol.
141 :4053-4060).
Human mAb antibodies directed against human proteins can be generated using transgenic mice carrying the complete human immune system rather than the mouse system. Splenocytes from these transgenic mice immunized with the antigen of interest are used to produce hybridomas that secrete human mAbs with specific affinities for epitopes from a hurnan protein (see, e.g., Wood e~ al. International Application WO
91 /00906, Kucherlapati et al. PCT publication WO 91 / 10741; Lonberg et al.
International Application WO 92/03918; Kay et al. International Application WO
92/03917; Lonberg, N. et al. 1994 Nature 368:856-859; Green, L.L. et al. 1994 ,~Jature Genet. 7:13-21; Morrison, S.L. etal. 1994Proc. Natl. Acad Sci. USA 81:6851-6855;Bruggeman et al.. 1993 Year Immunol 7:33-40; Tuaillon et al. 1993 PNAS 90:3720-3724; Bruggeman etal.. 1991 Eur JImmunol 21:1323-1326).
Monoclonal antibodies can also be generated by other methods well known to those skilled in the art of recombinant DNA technology. An alternative method, referred to as the "combinatorial antibody display" method, has been developed to identify and isolate antibody fragments having a particular antigen specificity, and can 35 be utilized to produce monoclonal antibodies (for descriptions of combinatorial antibody display see e.g., Sastry et al. 1989 Pl\~S 86:5728; Huse et al. 1989 Science 246: 1275;
_ SUBSTITUTE PAGE
~MEN~E~ SHEET

CA 022~9371 l999-01-0~

and Orlandi et al. 1989 PNAS 86:3833). After immunizing an animal with an immunogen as described above~ the antibody repertoire of the resulting B-cell pool is cloned. Methods are generally known for obtaining the DNA se4uence of the variable regions of a diverse population of immunoglobulin molecules by using a mixture of oligomer primers and PCR. For instance~ mixed oligonucleotide primers corresponding to the 5' leader (signal peptide) sequences and/or framework I (FRI ) sequences. as we]l as primer to a conserved 3' constant re~ion primer can be used for PCR amplification of the heavy and light chain variable regions from a number of murine antibodies (Larrick el al, l 991~ Biotechniques 11: 152- 156). A similar strategy can also been used to amplify 10 human heavy and light chain variable regions from human antibodies (Larrick et al.
1991~ Me~hods. Companion to Methods in Enzymology 2: 106- 110).
The term "complement" refers to a set of more than 30 serum proteins that are universally present without prior exposure to a particular antigen (see~
Liszewski, M. et al.~ 1993~ Fundamental Immunol., 3rd Ed., W. Paul Ed. Ch. 26 "The 15 Complement System" p. 917). The function of the complement system is modification of the membrane of an infectious agent~ and promotion of an infl~mm~tory response through cell action. Complement proteins are converted to active forms by a series of proteolytic cleavages. Production of a reactive C3b protein can occur ~uickly and efficiently via the "classical" complement pathway, or slowly and inefficientlv via the 20 "alternative" pathway. C3 is secreted by monocytes and macrophages: a complex of Factors B and D and properdin cleave C3 to yield the products C3a and C3b. Theseproducts promote mast cell degranulation, releasing infl~mm~tory molecules such as histamine, proteases, Iysozyme, acid hydrolases. and myeloperoxidase. Opsonization of target cell membranes promotes Iysis and phagocytosis.
As used herein, the term "cytokine" means a protein hormone that can mediate immune defenses against "foreign" substances or org~nism~. General properties of cytokines are reviewed, for example, by Abbas, A. et al. Cell and Molecular Immunology, 2nd Ed.. 1994, Saunders~ Philadelphia. Infl~mm~tory cytokines include tumor necrosis factor (TNF), interleukin I ~ (IL-I ~). IL-6, and y-interferon (IFN-y).
30 Production of cytokines by the host can be stimulated by a microbial product. such as lipopolysaccharide (LPS), or by a foreign antigen.
Cytokines can be produced by cells of the immune system. for example.
T cells and basophils. and can act on a nearby other cell (paracrine action). or on the producing cell (autocrine action), or can be released into the circulation to act on a 3~ distant cell (endocrine). Categories of function of cytokines include: mediation of natural immunity; regulation of Iymphocyte activation, growth, and differentiation:

CA 022S9371 lg99-ol-OS

regulation of immune-mediated inflammation; and stimulation of leukocyte growtl1 and differentiation.
Cytokine function is initiated by binding to a specific receptor on a tar,~et cell. For example~ the 17kD TNF polypeptide which functions as a trimer, is produced 5 by phagocytes and T cells. It binds to a specific TNF-receptor located on~ for example. a neutrophil or an endothelial cell to activate the responses of infl~mm~tion. One such response in these target cells is production of IL -l ,B~ which in turn provokes production of IL-6. Both TNF and IL - I ~ act on thymocytes to initiate a signal cascade clllmin:~ting in increased expression of genes encoding Ig proteins. Similarly~ IFN-y binds to specific 10 cell receptors to stimulate expression of different sequences. These cytokines also bind to receptors on liver cells to activate expression of proteins of the acute phase of immune response.
Other cytokines can be anti-inflammatory in their effects on the immune system, for example~ IL-4. IL-10. and IL-13 (Joyce. D. e~ al. 1994. Eur. J. Immunol. ~4:
2699-2705; Zurawski~ et al . 1994, Immunol. Today 15: 19-26). IL- 10 thus reduces the pro-inflamm:~tory effects of TNF by down-regulating surface TNF receptor (TNF-R) expression~ increasing production of soluble TNF-R~ and inhibiting the release of TNF.
Further, the function of human IL- 13 protein~ studied by stimulation of monocytes with LPS~ inhibits production of IL-I a~ IL-I ~. IL-6, IL-8. MIP-l a, 20 TNF-a ~ IL- 10, GM-CSF and G-CSF. Further, production of IL- I ra (receptor antagonist)~ a soluble form of the IL- I receptor~ is enhanced. These anti-inflamm~tor~
properties are similar to those of IL-4 and IL-10.
Immune response to "foreign" antigens comprises the notion that "self' proteins and other molecules expressed within an org~nism~ are not antigenic or 25 immunogenic to that org~ni~m. In fact~ discrimination between isologous or homologous determinants and foreign, or heterologous determinants is achieved throu,~h maturation of the immune system of an organism during development of the immune system. A system of selection against immune cells bearing antibodies with binding determin~nt~ to "self" occurs~ so that when mature the immune system does not attack 30 proteins or other molecules native to the organism. In certain pathological conditions known as "autoimmune diseases," however. such discrimination is not as accurate~ and endogenous structures may be subject to attack from the immune system. Examples of autoimmune ~ e~es and conditions in which there is autoimmune exacerbation of symptoms include systemic lupus erythematosus. myasthenia gravis. multiple sclerosis.
35 and rheumatoid arthritis. Compositions of the instant invention which are capable of binding to a site on the Fca receptor, by virtue of comprising a binding determinant of CA 02259371 1999 - 01- 0~

wo 98/02463 an antibody for a site on this receptor, can also modulate the number of these receptors on the cell surface~ and accordingly are potential agents for treatment of autoimmune diseases. Further, amino acid residue sequence data of the Fv regions of the antibody binding determin~rlt is the basis for obtainin~ a three-dimensional model of the protein features~ such as size, charge, and shape of the set of residues which comprise this binding site~ so that agents which mimic this binding site may be designed.
The agents of the invention are ~rimini~tered to subjects in biolo~ically compatible forrns suitable for phannaceutical a-lministration in vivo to produce a therapeutic response against a cancer or an infectious disease. By "biolo~icallycompatible forrn suitable for a-lmini~tration in vivo" is meant a form of the protein to be administered in which any toxic effects and side effects are outwei~hed by the therapeutic effects of the composition.
The term "subject~" as used herein. refers to a living animal or human in need of susceptible to a condition, in particular a "cancer or infectious disease" as defined below. The subject is an organism possessing leukocytes capable of responding to antigenic stimulation and growth factor stimulation. In preferred embodiments the subject is a m~mm~l, including hllm~n.~ and non-human rn~mmz~l~ such as dogs. cats~
pigs~ cows. sheep, goats, horses, rats. and mice. In the most preferred embodiment. the subject is a human. The terrn "subject" does not preclude individuals that are entirely normal with respect to cancer, infectious disease, or norrnal in all respects.
The term 'Ipatient~'' as used herein, refers to a human subject who has presented at a clinical setting with a particular symptom or symptoms suggesting one or more therapeutic regimens. A patient may be in need of further categorization byclinical procedures well-known to medical practitioners of the art (or may have no further disease indications and appear to be in any or all respects norrnal). A patient's diagnosis may alter during the course of disease progression~ such as development of further disease symptoms, or remission of the disease, either spontaneously or during the course of a therapeutic regimen or treatment, or rediagnosis as being entirely of norrnal condition.
The terrn "inf'ectious disease" is meant to include disorders caused by one or more species of bacteria, viruses, fungi. and protozoans~ which are disease-producina org~ni.~m~ collectively referred to as "pathogens." The terrn "fungi" is meant to include the yeasts. In this invention, pathogens are exemplified~ but not limited to, Gram-positive bacteria such as Enterococcus fecalis, Hemophilus pneumoniae, Listeria monocytogenes, Mycobacterium tuberculosis, M leprae. Proprionibacterium acnes, Staphylococcus aureus~ S epidermis, S. intermedias, Streptococcus hemolyticus. S.

CA 022~9371 l999-ol-o~

pneur~10niae; Gram-negative bacteria such as Flavobacterium meningosepficum, Helicobacte~ pylori~ Hemophilus pneumoniae. H. influen7ae, Klebsiella pneumonia,Neisseria gonorrhoeae! Pseudomonas aeru~inosa~ Shi~ella dysenteria~ Salmonella typhi, S. paratyphi. Escherichia coli serotype 0157, Chlamydia species~ ~elicobacter 5 species; viruses such as HIV-1,-2, and -3, HSV-I and -Il~ non-A non-B non-C hepatitis virus, pox viruses, rabies viruses, and Newcastle disease virus; fungi such as Candida albicans. C. ~ropicalis, C. krusei, C. pseudotropicalis, C. parapsilosis, C. quillermondii, C. stellatoidea, Aspergillus fumigatus, A. niger, A. nidulans, A. flavus, A. terreus, Absidia corymbi.fèra, A. ramosa, Cryptococct~s neoforms~ Histoplasma capsulatum,10 Coccidioides immitis~ Pneumocystis carinii, Rhizopus arrhizl~s, R. oryzae, Mucor pusillus and other fungi; and protozoa such as Entamoeba histolytica, Enlamoeba coli.
Giardia lamblia, G. intestinalis, Eimeria sp.. Toxoplasmn sp., Cr~)ptosporidium pa~ vum, C'. mt~ris. C. baileyi, C. meleagridis, C. wrai/i, and C. nosarum. Obtaining unique epitopes from these organisms by screening proteins and by assaying peptides in vitro 15 are commonly known to those skilled in the art.

Il. Multispecific molecule.s The instant invention relates in one embodiment to recombinant multispecific molecules. which have affinity for and are capable of binding at least two 20 different entities. Multispecific molecules can include bispecific molecules comprised of a binding determinant for an Fc receptor and a binding determinant for a target. The preferred multispecific molecules for the instant invention include molecules which are comprised of at least one copy of a binding determinant which binds specifically to an Fca receptor or target: or molecules comprised of at least one binding determinant 25 which binds an Fca receptor, one binding determinant for a target and additionally one or more binding deterrnin~nts that recognize other molecules. A preferred multispecific molecule is a bispecific antibody (BsAb), which carries at least two different binding determin~nts? at least one of which is of antibody origin.
A "binding determinant for an Fca receptor" refers to an antibody, a 30 functional antibody fragment (e.g., Fab fragment) or a ligand such as an engineered binding protein that recognizes and binds an Fca receptor on an effector cell. Preferred antibodies for use in the subject invention bind the Fca receptor on an effector cell (white blood cell) at a site which is not bound by endogenous immunoglobulin A (IgA).
Most preferably. the anti-Fca receptor VH and VL portion binds a human FcaR.
35 Preferred hnm~ni7~d anti-FcaR monoclonal antibodies are described, the teachings of which are fully incorporated herein by reference. The antibody that comprises the BsAb CA 022~937l l999-0l-05 or multispecific molecule of the invention may be whole. i.e. havin~ heavy and light chains or any fra~ment thereof, e.g. Fab or (Fab')~ fragment. The antibody l'urther may be a light chain or heavy chain dimer~ or any minim~l fragment thereof such as a Fv or a single chain construct as described in Ladner et al. (U.s. Patent No. 4.946.778~ issued 5 August 7, 1990). the contents of which is expressly incorporated by reference.An "effector cell" as used herein refers to an immune cell which is a leukocyte or a Iymphocyte. Specific effector cells express specific Fc receptors and carry out specific immunc functions. For example monocytes macrophages~
neutrophils. eosinophils. basophils. and Iymphocytes which express Fca~R are involved 10 in specific killin~ of tar~et cells and presentin~ antigens to other components of the immune system. or binding to cells that present antigens. ~he expression of a particular FcR on an effector cell can be re~ulated by humoral factors such as cytokines. For example. expression of Fc~RI has been found to be up-regulated by interferon gamma (IFN-~). This enhanced expression increases the cytotoxic activity of Fc~RI-bearing 1~ cells against targets.
The recombinant antibodies or antibody fragments which specifical]y bind to an Fc receptor~ are preferably "h~lm~ni7ed" i.e. carry portions derived from a human antibody, but having at least a portion of a _omplementarity determining region (CDR) derived from a non-human antibody. Ordinarily that portion which is 20 "hllm~ni7ed" is selected to provide specificity of the hum~ni7~d antibody to bind a human Fc receptor. The hllm~ni7ed antibody has CDR portions derived from a non-hun~an antibody and the "constant" portions of the antibody molecule are of human ongln.
The portion of the non-human CDR inserted into the human antibody is 25 selected to be sufficient for allowing bindin~ of the hllm~ni7Pd antibody to the Fca receptor. A sufficient portion may be selected by inserting a portion of the CDR into the human antibody and testing the binding capacity of the created hnm~ni7ed antibody using flow cytometry or enzyme linked immunosorbent assay (ELISA).
Allof the CDRs of a particular human antibody may be replaced with at 30 least a portion of a non-human CDR or only some of the CDRs may be repJaced with non-human CDRs. It is onJy necessarv to repJace the number of CDRs required for binding of the hum~ni7~d antibody to the Fc receptor.
An antibody can be hnm~ni7ed by any method, which is capable of replacing at least a portion of a CDR of a human antibody with a CDR derived from a 35 non-human antibody. Winter describes a method which may be used to prepare the hnm~ni7t d antibodies of the present invention (UK Patent Application GB 21 88638A, CA 022~9371 l999-ol-o~
WO 98/02463 PCTIUS97tl2013 filed on March 26. 1987). the contents of which is expressly incorporated bv reference.
The human CDRs may be replaced with non-human CDRs using oligonucleotide site-directed mutagenesis as described in International Application WO 94/10332 entitled~
Humanized An~ibodies to Fc Receptors for Immuno~lobuli~? G on Human Mononuclea~
5 Pha~;ocy~es.
In addition to an anti-Fc~ receptor portion~ the claimed multispecific molecules can comprise a binding determinant for a target i.e. an antibody, a functional antibody fragment or a ligand that recognizes and binds a pathogen (e.g.. viruses, bacteria. fungi, protozoa), a pathogen infected cell. a cancer or tumor cell (e.g.. breast.
10 ovarian, prostate~ testicular, lung, colon, rectum, pancreas, liver, central nervous system head and neck, kidney, bone, blood, and Iymphatic system) or other unwanted cell in a subject (e.g., a human or animal) or an antigen or modified form thereof. Additionally.
the target portion may comprise or be directed against an antigen. A preferred embodiment contains an antigen that can be used to induce a specific immune response I S against a chronic infection, against a tumor or cancer cell, or to deplete antigen in the circulation. A different particularly preferred embodiment has an antigen that is attached to a multivalent molecule containing a binding determinant for an FcR. which stimulates the immune system by directing the antigen to an antigen presenting cell.
ln one embodiment of the invention, the multispecific molecule contains 20 a binding determinant or ligand which interacts with a molecule. In a preferred embodiment. the binding determinant binds a protein. e.g., a protein on a target cell.
such as a cancer cell, or a cell of an infectious disease agent or the agent itself or an infected cell. Preferred binding determinants include antibodies. fragments of antibodies, and receptors for grov~th factors or differentiation factors. For example, a 25 multivalent molecule can comprise an epiderrnal growth factor (EGF). or at least a portion or modified form that is capable of interacting with a receptor, e.g.. an epidermal growth factor receptor EGF-R, or an antibody to EGF-R. A particularly preferred embodiment of the invention comprises a BsAb carrying a binding determinant for an human EGF-like eceptor, including the EGF-R, HER2/neu. HER3. HER4, etc. In yet 30 another preferred embodiment, the binding determinant is for the tumor antigen TAG 7 found e.g. on tumors of the breast, colon. and ovary.
In another preferred embodiment of the invention. the ligand is a small peptide, such as bombesin, gastrin-releasing peptide (GRP). Iitorin, neuromedin B. or neuromedin C. The sequences of the peptides can be found, e.g.. in U.S. Patent No.
5.217.955, the content of which is incorporated herein by reference. The ligand can also be a modified form of any of these peptides. The modification can increase binding to ... . . .

CA 0 2 2 ~ 9 3 7 1 1 9 9 9 - o l - 0 ~

the receptor. decrease binding~ or not affect the binding to a receptor. The modification of the ligand can also transform an agonist into an antagonist, such that the ligand inhibits rather than stimulates cell proliferation. Modification of the ligand can be an addition a deletion, a substitution~ or a modification of at least one amino acid.
In another preferred embodiment of the invention, a multispecific or bispecific molecule comprises an antigen. As used herein, the term "antigen" means any natural or synthetic immunogenic substance, a fragment or portion of an immunogenic substance, a peptidic epitope, or a hapten. In one embodiment of the invention, a bi- or multispecific molecule is employed to target an antigen, e.g., tetanus toxoid to the cell to enhance the processes of internalization and presentation by these cells, and ultimately, to stimulate an immune response therein. In a specific embodiment~ the bispecific binding agent specifically binds the antigen (either directly, to an epitope of the antigen~
or indirectly to an epitope attached to the antigen) and~ at the same time. binds a surface receptor of an antigen-presenting cell which can internalize antigen for processing and presentation. In another embodiment, the antigen is linked to the multi- or bispecific molecule and at the same time binds a surface receptor of an antigen-presenting cell. In a preferred embodiment the antigen is covalently attached to the multispecific molecule by genetic or chemical means The receptor-binding component of the bi- or multispecific molecule (and thus the bi- or multispecific molecule, itself) binds the receptor of the anti~en-presenting cell at a site different and distinct from the naturally-occupying ligand. Thus, binding of the multispecific molecule occurs without competition by the natural ligand for the receptor. As a result, binding to the receptor will not be prevented by physiological levels of the ligand and the targeted receptor will remain capable of binding the molecule of the invention and the ligand.
One type of antigen can be an allergen. An "allergen" refers to a substance that can induce an allergic or asthmatic response in a susceptible subject. The number of allergens that elicit a sensitive response in a proportion of a population is enormous, and includes pollens, insect venoms, animal dander, dust mite proteins, fungal spores and drugs (e.g. penicillin). Examples of natural animal and plant allergens include proteins specific to the following genera: Felis (~elis domesticus); Canis (Canis familiaris); Dermatophagoides (e.g. Dermatophagoides,farinae); Periplaneta (e.g.Periplaneta americana); Ambrosia (Ambrosia artemiisfolia; Lolium (e.g Loliun1 perenne or Lolium multiflorum); Cryptomeria (Cryptomeria japonica): Alternaria (Alternaria alternata); Alder; Alnus (Alnus gultinosa); Betula (Betula verrucosa);
3~ Quercus (Quercus alba~; Olea (Olea europa); Artemisia (Artemisia vulgaris); Plantago (e.g. Plantago lanceolata); Parietaria (e.g. Parietaria o,~ficinalis or Parietaria judaica . . .

CA 0 2 2 ~ 9 3 7 1 1 9 9 9 - o l - o ~

- Blattella (e.g. Blattella ~ermanica); Apis (e.g. Api.s multiflorum): Cupress2~s ~e.g.
Cupressus sempervirens~ Cupressus ari onica and CupressZIs macrocarpa): Juniperus (e.g. Juniperus sabinoides Juniperus virginiana. Jllniperl~s communis and Juniperus ashei) Thuya (e.g. Thuya orientalis); Chamaecvparis (e.g. Chamaecypa~-is obtusa);
5 Agropyron (e.g. Agropyron repens); Secale (e.g. Secale cereale): Triticu~77 (e.g. Triticum aestivum); Dac~ylis (e.g. Dac~ylis glomerata); ~estuca (e.g. Festuca elatio~); Poa (e.g.
Poa pratensis or Poa compressa); Avena (e.g. Avena sativa); ~olcus (e.g. Holcus lanatus); Anthoxanthum (e.g. Anthoxanthum odoratl~m); Arrhenatherum (e.g.
Arrhenatherum elatius); ~grostis (e.g. Agrostis alba): Phleum fe.~;. Phleum pralense);
10 Phalaris (e.g. Phalaris arundinacea); Paspalum (e.g. Paspalum notatum): Sor~hum (e.g.
Sorghum halepensis); and Bromus (e.g. Bromus inermis).
Many allergens are found in airborrle pollens of ragweed, grasses, or trees~ or in fungi. animals. house dust~ or foods. As a class~ they are relati~ely resistant to proteolytic digestion. Preferable allergens are those which bind to IgE on mast cells 15 and basophils, thereby causing a range of symptoms from infl;lmm~tion and asthma to a type I anaphylaxis hypersensitivity reaction.
In another preferred embodiment, a binding determinant is specific for an antigen on an infectious disease agent or an infected cell, as defined supra. A binding determinant can be obtained from a polyclonal antibody, which can be produced by20 injection of a rabbit or other suitable animal with a fragment or component of a pathogen~ for example, a cell wall component of a pathogen. IgG can be purified from the serum of the immunized animal, F(ab')~ can be prepared~ and used as the source of the target binding determinant. For example, polyclonal antibody prepared by immunizing a rabbit with a preparation of Candida albicans fragments can be obtained.
25 and used to prepare anti-C. albicans IgG F(ab')2~ which can be coupled to a binding determinant for FcaRI by chemical methods described herein.
In some cases. it may be desirable to couple a substance which is weakly antigenic or nonantigenic in its own right (such as a hapten) to a carrier molecule, such as a large immunogenic protein (e.g., a bacterial toxin) for a~ministration. In these 30 instances, the bispecific binding reagent can be made to bind an epitope of the carrier to which the substance is coupled~ rather than an epitope of the substance itself.
The antigen that can be linked either directly~ or indirectly~ to a multi- or bispecific molecule of the invention can be soluble or particulate: it may carry B cell epitopes, T cell epitopes or both. The antigen can be bacteriah fungal, viral or parasitic 35 in origin. Often, the antigen will comprise a component of the surface structure of a pathogenic org~ni~m, or a surface structure in a cell infected by a pathogenic organism.

CA 022~9371 1999-01-0~

MXI-06~CPPC

For example~ the antigen can comprise a viral surface structure such as an envelope Plvcoprotein of human immunodeficiency virus (HIV) or the surface antigen of hepatitis virus. In addition? the antigen can be associated with a diseased cell, such as a tumor cell. against which an immune response may be raised for treatment of the disease. The 5 antigen can comprise a tumor-specific or tumor-associated antigen, such as the HER-2/neu proto-oncogene product which is expressed on human breast and ovarian cancer cells (Slamon ef al. (1989) Science 24~:707). Another important cancer antigen which comprises a target of the BsAb of this invention is TAG 72, which has been identified on about 90~/0 of colorectal cancers, 85% of breast tumors, and 95% of ovarian tumors (Johnson ef al.1986 Cancer Res. ~6:850-897; Bodmer, M. el al., European Patent Specification 0 348 442 Bl; Mezes, P. ef al. Intemational Application WO 93/12231).
The cells of a subject can be exposed ex vivo or in vivo to the multispecific molecules of the invention, to target an antigen to antigen-presenting cells.
Immune cells are separated and purified from subject blood, exposed to a multispecific I S molecule comprising the antigen, or the cells can be exposed to the antigen together with a multispecific molecule having a binding determinant for the antigen. After stimulation~ cells are returned to the subject. Cells to be used in this procedure can also be treated with cytokines or other factors, for the purpose of, for example, up-regulating numbers of receptors per cell. Further, in vivo or ex vivo therapeutic use of the molecules can be enhanced by treatment of the subject with one or more cytokines or gro~th factors.
The method of this invention can be used to enhance or reinforce the immune response to an antigen. For exarnple, the method is valuable for the treatment of chronic infections, such as hepatitis and AIDS, where the unaided immune system is unable to overcome the infection. It can also be used in the treatment of the acute stages of infection when reinforcement of immune response against the invading organism may be necessary.
The method can be used to reduce the dose of antigen required to obtain a protective or therapeutic immune response or in instances when the host does notrespond or responds minim~lly to the antigen. Although generally desirable, the lowering of effective dose can be especially desirable when the antigen is toxic to the host such as is the case for allergies. Methods and uses for bi- or multispecific molecules comprising one or more antigens or comprising one or more binding determinants, e.g., an antibody interacting with an antigen, are further described in the published International Application WO 92/05793.

SUBSTITUTE PAGE
AMEN~D S~EET

CA 022~9371 l999-ol-o~
111 Methods for Makin~ MMltispecific Molecules The multispecific molecules described above can be made by a number of methods. For example, both specificities can be encoded in the same vector and expressed and assembled in the same host cell. This method is particularly useful where 5 the multi-specific molecule is a mAb x mAb. mAb x Fab~ Fab x F(ab'), or ligand x Fab fusion protein. A bispecific molecule of the invention can also be a single chain bispecific molecule, such as a single chain bispecific antibody. a single chain bispecific molecule comprising one single chain antibody and a binding determinanL or a single chain bispecific molecule comprising two binding determin~nt~. Multispecific 10 molecules can also be single chain molecules or may comprise at least two single chain molecules. Methods for preparing bi- or multivalent antibodies are described forexample described in U.S. Patent Number 5~260.203; U.S. Patent Number 5.4~5.030:U.S. Patent Number 4.881,175; U.S. Patent Number 5~132.405; U.S. Patent Number 5.091,513; U.S. Patent Number 5,476~786; U.S. Patent Number 5~013~653: U.S. Patent Number 5,258.498; and U.S. Patent Number 5.482,858.
Binding of the single chain molecules to their specific targets can be confirmed by bispecific ELISA, familiar to those skilled in the art. Alternatively. each specificity of a multispecific molecule can be generated separately and the resulting proteins or peptides chemically conjugated to one another. For example~ two hum~ni7ed antibodies or antibody fragments can be conjugated via sulfhydryl bonding of the C-terminus hinge regions of the two heavy chains as described in Examples, infra The BsAbs of the present invention can be prepared by conjugating the anti-FcR and anti-target portions using methods described in the following Examples or those well-known in the art. For example~ a variety of coupling or cross-linking agents can be used for covalent conjugation. Examples of cross-linking agents include protein A, carbodiimide, N-succinimidyl-S-acetyl-thioacetate (SATA). N-succinimidyl-3-(2-pyridyldithio)propionate (SPDP), and sulfosuccinimidyl 4-(N-maleimidomethyl) cyclohexane-l-carboxylate (sulfo-SMCC) (see e.g., Karpovsky et al. Ig84 J. Exp. Med.
160: 1686; Liu, MA et al. .1985 Proc. Natl. Acad. Sci. USA 82:8648). Other methods include those described by Paulus (Behring Ins. Mitt. 1985 No. 78, 118-132): Brennan el al. (Science 1985 229:81-83). and Glennie et al. (J. Immunol. 1987 139: 2367-2375).
Examples of other cross-linking agents include ortho-phylenedimaleimide (o-PDM).protein A, carbodiimide. In the preferred embodiment for BsAb, the conjugating agent is o-PDM. Other preferred conjugating agents are SATA and sulfo-SMCC. both available from Pierce Chemical Co. (Rockford, IL).

, CA 022~9371 l999-ol-o~

Based on their ability to bind FcR bearing immune cells and specific target cells. a particular embodiment of a multispecific molecule can be administered to a subject to treat or prevent reoccurrence of a variety of diseases or conditions, includin~: cancer (e.g., breast~ ovarian, testicular, prostate~ lung, brain. colon~ rectum.
5 pancreas. Iiver, central nervous system~ head and neck~ kidney, bone~ blood and Iymphatic system), pathogenic infections such as viral (such as HIV~ HTLV and FELV)~
protozoan (such as Toxoplasma gondii)~ fungal (such as Candida albicans); and bacterial (such as S~aphylococcus ~ureu*, Slreptococcus hemoly~icus and Mycobacterium tuberculosis). Another aspect of the invention provides molecules that are useful for 10 vaccination against diseases and cancer by including an anti~en from disease or~ni.~ms from infected cells, f~om gene products of disease or~ni.~m.~ or from cancer cells. For these purposes, the invention provides compositions which are multispecific molecules that link the useful operative antigen to a binding determinant that directs the anti~en to the immune system. An Example provided herein describes a molecule which functions 15 to target tetanus toxoid directly to FcaR on monocytes, resulting in stimulation of T
cells at lower doses than is required by free tetanus toxoid.
For use in therapy, an effective amount of an appropriate multispecific molecule can be A-lmini~tered to a subject by any mode that allows the molecules to exert their intended therapeutic effèct. Preferred routes of ~-lmini~tration include oral, 20 transdermal (e.g., via a patch), and injection (subcutaneous. intravenous, parenteral.
intraperitoneal, intrathecal. etc.). The injection can be in a bolus or a continuous r mIuslon.
Further~ the cells of a tissue, e.g. blood. may be removed from a patient.
fractionated and cultured if appropriate to expand the cell number, treated er vivo with 25 the multispecific multivalent composition in a pharmaceutically acceptable carrier. and returned to the patent for therapy. During the ex vivo culture and expansion~ a particular cell type may be selected, e.g. a monocyte population. Further, eX vivo cultured cells may be treated at various points during eX vivo culture and expansion. with agents to modify certain functional Fc(xR molecules. Agents include but are not limited to~
30 ~rowth factors, cytokines, Iymphokines such as IFN-r, G-CSF, TNF, and GM-CSF. and interleukins such as IL-2, IL-lO and IL-12.
A multispecific molecule is ~tlmini.stered in conjunction with a pharmaceutically acceptable carrier. As used herein. the phrase "pharmaceutically acceptable carrier" is intended to include substances that can be coadministered with a 35 multispecific molecule and allows the molecule to perform its intended function.
Examples of such carriers include solutions~ solvents. dispersion media. delay agents.

CA 022~9371 l999-ol-o~
W O 98/02463 PCTrUS97/12013 emulsions and the like. The use of such media for pharrnaceutically active substances are well known in the art. and are discussed infi a. Any other conventional carrier suitable for use with the molecules falls within the scope of the instant invention.
Further~ therapy with the multispecific multivalent binding molecule may be coordinated S into a treatment regimen with other similar molecules, or with traditional chemotherapeutic agents such as cis-platin, AZT, DDI, adriamycin, tetracycline, cefachlor, nystatin. and acyclovir.
Combinatorial libraries can be screened to obtain members of the library with a desired binding activity, and to identify the active species, hy methods that have been described (see, e.g., Gordon et al., J Med. Chem., op. cit. ). These include affinity chromatography with an appropriate "receptor" to isolate ligands for the receptor.
followed by identification of the isolated ligands by conventional techniques (e.g.~ mass spectrometry and NMR). Prel'erably, the soluble receptor is conjugated to a label (e.g fluorophores, spectrophotometric enzymes, radioisotopes, or luminescent compounds) that can be detected to indicate ligand binding. Alternatively~ immobilized compounds can be selectively released and allowed to diffuse through a membrane to interact with a receptor.
Combinatorial libraries of compounds can also be synthesized with "tags"
to encode the identity of each member of the library (see, e.g., W.C. Still et al.
International Application WO 94/08051). In general, this method features the use of inert but readily detectable tags, that are attached to the solid support or to the compounds. When an active compound is detected the identity of the compound is determined by identification of the unique accompanying tag. This tagging methodpermits the synthesis of large libraries of compounds which can be identified at very lou levels among to total set of all compounds in the library.
Specific binding proteins with high affinities for targets can be made according to methods known to those in the art. For example, proteins that bind specific DNA sequences may be engineered, and proteins that bind a variety of targets~ especially protein targets (Ladner, R.C., et al., U.S. Patent 5,233,409: Ladner, R.C.. et al.. U.S.
Patent 5~403,484) may be engineered and used in the present invention as the FcalR
binding deterrninant or as the target binding determinant~ as apl,l opl iate. Further. the methods of these libraries can be used in screens to obtain binding determinants that are mimetics of the structural determin~nt~ of antibodies.
In particular, the Fv binding surface of a particular antibody molecule interacts with its target ligand according to principles of protein-protein interactions, hence sequence data for VH and VL (the latter of which may be of the 1C or i~ chain type) CA 022~9371 1999-01-05 is the basis for protein engineering techniques known to those with skill in the art.
Details of the protein surface that comprises the binding deterrninants can be obtained from antibody sequence information. by a modeling procedure using previousJy determined three-dimensional structures from other antibodies obtained from NMR
5 studies or crytallographic data. See for example Bajorath, J. and S. Sheriff~ 1996, Proleins: S~ruct. Func~.. and Gene~. 2~ (2), 152-157; Webster~ D.M. and A. R. Rees, 1995, "Molecular modeling of antibody-combining sites." in S. Paul, Ed., A~ethods in Molecular Biol. 51 Antibody Engineering Protocols, Humana Press, Totowa. NJ, pp 17-49; and Johnson. G., Wu. T.T. and E.A. Kabat. 1995, "Seqhunt: A program to screen ali~ned nucleotide and arnino acid sequences," in Methods in Molecular Biol. 51 op ci~
pp 1-15.
Breast and ovarian cancers are sex hormone dependent cancers. Breast tumors may be characterized by abnormally expressed receptors, e.g. those of the_uman-EGF-like receptor family (HER), for example HER-2. -3, and 4. The invention is not }imited to these embodiments of HER antigens. The natural HER ligand, Heregulin. can be incorporated into a bispecific antibody (BsAb) or multispecific molecule, as a means to target a breast tumor cell expressing one or more HER receptor during cancer. Further. a heregulin molecules are binding determinants for heterodimeric HER receptors cont~inin~, eg. a monomer of each of HER-~, -3 or -4 in combination.
Additional examples of sex hormone-dependent cancer include prostate cancer (Smith, P. ( 1995), Cancer Survey* T/ol. 23 Preventing Prost~fe Cancer. Imper.
Cancer Research Fund and testicular cancers). The growth of hormone-dependent cancer types is promoted by male hormones (e.~.~ androgens such as testosterone and 2~ dihydrotestosterone). Removal of the testes (castration) was for many years the standard method of preventing secretion of male hormones by the gonads, to reduce growth of the cancer. Currently, secretion of male hormones is suppressed by chemical means byinterfering with production of luteinizing hormone (LH), which regulates synthesis of male hormones. Similar considerations are applicable to other sex hormone-dependent cancers, such as breast or ovarian cancer, so that patients with these diseases or in a population prone to these diseases, are usually not ~-lministered sex hormones as therapeutic or replacements. Multispecific molecules of the invention can comprise binding determinants for sex hormones, to reduce the concentration and suppress tumor growth.
In a preferred embodiment, the methods of this invention include ~lministration, for example, to a cancer patient, of a multispecific multivalent binding CA 022~9371 l999-ol-o~
WO 98t02463 PCT/US97/12013 molecule preparation comprising at least one binding determinant with affinity for a tumor marker or a tumor-specific protein of the cancer to be treated, for example, the nestin protein for brain cancers. The nestin protein~ which is expressed during normal m~n~m~lian fetal development~ is also expressed on tumors of the central nervous5 system, including most forms of brain cancer (McKay, D.G. Ronald~U.S. Patent No.
5,338,839. 8/16194). It is also expressed on melanomas on the skin and on melanomas that have metastasized (V.A. Florenes, R. Holm. O. Myklebost, U. Lendahl. O. Fodstad.
Cancer Res. ~: 354-6, 1994), to other organs and are difficult to detect and treat. The preferred site of delivery for treatment of a brain tumor with the molecules of this 10 invention is directly into the central nervous system or directly, to the brain via spinal injection or fine needle delivery. For a metastatic cancer, a preferred delivery route would be by direct in3ection into the circulation~ or by the ex vivo blood methods described herein.
Other tumor types for which the methods of this invention are 15 exemplified by. but are not limited to, Wilm's tumor (A.J. Buckler, et al. U.S. Patent No.
~.350,840) a pediatric kidney cancer due to occurrence of a somatic mutation in the patient's single copy of a gene normally found in two intact copies. Wilm's tumor can be cured surgically in 95% of cases. and a bispecific or multispecific multivalent bindin~
protein is envisioned to be suitable as an adjunct therapeutic modality for surgical 20 patients. Other examples of known cancer-associated proteins for which the compositions of matter and methods of the current invention are suitable include those associated with gastrointestinal cancer (R. Fishel et al.. International Application WO
95/14085, 05/26/95), those characterized by development of multiple drug resistance during chemotherapy (J.M. Croop et al ., U.S. Patent No.5,198.344)~ and a large number 25 of oncogenes well known to the skilled artisan such as Rb, ras, and c-m,l!c. the sequences of which are available for analysis to those with skill in the art. The compositions of this invention are, for example, suitable for inhibition of secreted enzymes such as matrix metal~oproteinases, which are considered to potentiate tumor metastasis (Liotta. L.A., et al. 1991 Cell 64:327-336). In the latter embodiment, a multispecific binding molecule 30 with a binding determinant to the matrix metalloproteinase and another for FcaR would facilitate inhibition and clearance of these enzymes from in situ activit~c If used in conjunction with standard surgical and chemotherapeutic regimens, the compositions are foreseen to reduce cancer re-occurrence and enhance long-term survival.

CA 022~9371 1999-01-0~

I l Pharmaceulical Composi~ions The compounds of the invention can be incorporated into pharmaceutical compositions suitable for ~(lmini.~tration to a subject in vivo. In a preferred embodiment, the pharmaceutical composition comprises either a multispecific molecule (compound, or agent) of the invention and a pharmaceutically acceptable carrier. In yet another embodiment of the present invention, the pharrnaceutical composition can be administered by combination therapy. i.e., combined with other agents. For example the combination therapy can include a composition of the present invention with at least one anti-cancer agent~ at least one antibiotic. at ]east one cytokine~ at least one vaccine?
or other conventional therapy. Exemplary anti-cancer agents include cis-platin, adriamycin, and taxol. Exemplary antibiotics include isoniazid~ rifamycin, and tetracycline. Exemplary cytokines include G-C~SF. GM-CSF. interleukins and interferons.
As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings~ antibacterial and antifungal agents~ isotonic and absorption delaying agents, and the like that are physiologically compatible. Preferably, the carrier is suitable for intravenous. intramuscular, subcutaneous, parenteral? spinal or epidermal ~f~mini.~tration (e.g, by injection or infusion). Depending on the route of a-lmini~tration. the active compound may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound, or for controlled release.
A "pharmaceutically acceptable salt" refers to a salt that retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects (see e.g., Berge, S.M.~ el al. (1977) J. Pharm Sci. 66:1-19).
Exarnples of such salts include acid addition salts and base addition salts. Acid addition salts include those derived from nontoxic inorganic acids, such as hydrochloric~ nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, phosphorous and the like, as well as from nontoxic organic acids such as aliphatic mono- and di-carboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, aromatic acids~ aliphatic and aromatic sulfonic acids and the like. Base addition salts include those derived from alkaline earth metals~ such as sodium, potassium, ma~nesium. calcium and the like~ as well as from nontoxic organic amines~ such as N.N'-dibenzylethylene~i~mine. N-methylglucamine, chloroprocaine, choline, diethanolamine, ethylene~i~mine, procaine and the like.
A composition of the present invention can be ~rlministered by a variety of methods known in the art. As will be appreciated by the skilled artisan~ the route CA 022~9371 1999-01-0~
W O 98/02463 PCT~US97/12013 and/or mode of ~clmini~tration will vary depending upon the desired results. The active compounds can be prepared with carriers that will protect the compound against rapid release? such as a controlled release formulation, including implants~ transdermal patches~ and microencapsulated delivery systems. Biodegradable, biocompatible 5 polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid~
collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See. e g., Sustained and Controlled Release Drug Delivery Systems, J.R. Robinsom ed.. Marcel Dekker, lnc., New York~ 1978.
To ~tlmini~ter a compound of the invention by certain routes of ~lmini.stration, it may be necessary to coat the compound with~ or co-~llmini~ter the compound with, a material to prevent its inactivation. For example~ the compound may be ~lmini.~tered to a subject in an appropriate carrier. for example~ liposomes~ or a diluent. Ph~ ceutically acceptable diluents include saline and aqueous buffer 15 solutions. Liposomes include water-in-oil-in-water CGF emulsions as well as conventional liposomes (Strejan e~ al., (1984) J. Neuroimmunol. 7:27). Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous p~epal~lion of sterile injectable solutions or dispersion. The use of such media and agents for pharmaceutically active substances is well known in the 20 art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the pharmaceutical compositions of the invention is contemplated. Supplementary active compounds can also be incorporated into the compositions.
Therapeutic compositions typically must be sterile and stable under the 25 conditions of manufacture and storage The composition can be formulated as a solution, microemulsion. Iiposome, or other ordered structure suitable to high drug concentration. The carrier can be a solvent or dispersion medium cont~ining~ forexample~ water, ethanol, polyol (for example, glycerol, propylene glycol. and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can 30 be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. In many cases, it will be preferable to include isotonic agents. for example. sugars, polyalcohols such as mannitol, sorbitol. or sodium chloride in the composition.
Prolonged absorption of the injectable compositions can be brought about by including 35 in the composition an agent that delays absorption~ for example, monostearate salts and gelatin.

.

CA 02259371 1999-01-0~

Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appro~l-ate solvent with one or a combination of ingredients enumerated above, as required, followed by sterilization microfiltration.
Generally~ dispersions are prepared by incorporating the active compound into a sterile 5 vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying (Iyophilization) that yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
Dosage regimens are adjusted to provide the optimum desired response (e.g., a therapeutic response). ~or example, a single bolus may be ~lmini.stered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. lt is especiall~
advantageous to formulate parenteral compositions in dosage unit forrn for ease of 1 j a~mini.~tration and uniforrnity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined ~uantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forrns of the invention are dictated by and directly dependent on (a) the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.
Examples of pharmaceutically-acceptable antioxidants include: (I ) water soluble antioxidants, such as ascorbic acid~ cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate. butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylene(li~mine tetraacetic acid (EDTA), sorbitoL tartaric acid, phosphoric acid, and the like.
For the therapeutic compositions, forrnulations of the present invention include those suitable for oral, nasal, topical (including buccal and sublingual)~ rectal.
vacinal andlor parenteral a~mini~tration. The formulations may conveniently be presented in unit dosage forrn and may be prepared by any methods well known in the art of pharmacy. The arnount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated. and the particular mode of ~lmini~tration. The amount of active ingredient CA 022~9371 l999-ol-o~

which can be combined with a carrier material to produce a single dosage form will generally be that amount of the composition which produces a therapeutic effect.Generally, out of one hundred per cent, this amount will range from about 0.01 per cent to about ninety-nine percent of active ingredient, preferably from about 0.1 per cent to 5 about 70 per cent. most preferably from about I per cent to about 30 per cent.Transdermal patches offer the advantage of providing controlled delivery of a compound of the present therapeutic inventions to the body. Such dosage forms can be made by dissolving or dispersing the composition in the proper medium. Absorption enhaulcers can also be used to increase the flux of the composition across the skin. The 10 rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the composition in a polymer matrix or gel. Devices, including patches~
which transdermally deliver a composition by iontophoresis or other electrically-assisted methods can also be employed in the present invention~ including. for example. the devices described in U.S. Patent Nos. 4,708,716 and 5,372,579.
The phrases "parenteral administration" and "~tlmini~tered parenterally"
as used herein means modes of administration other than enteral and topical lmini~tration, usually by injection, and includes, without limitation, intravenous~
intramuscular~ intraarterial, intrathecal, intracapsular. intraorbital~ intracardiac.
intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, 20 subcapsular, subarachnoid, intraspinal~ epidural and intrasternal injection and infusion.
Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions of the invention include water~ ethanol.
polyols (such as glycerol, propylene glycol, polyethylene glycol. and the like) and suitable mixtures thereof vegetable oils~ such as olive oil, and injectable organic esters.
25 such as ethyl oleate. Proper fluidity can be m:~int~inefl for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
These compositions may also contain adjuvants such as preservatives wetting agents, emulsifying agents and dispersing agents. Prevention of presence of 30 microorg~nism~ may be ensured both by sterilization procedures, supra~ and by the inclusion of various antibacterial and antifungal agents. for example, paraben.
chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride. and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought 35 about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.

~, CA 022~9371 1999-01-0~

When the compounds of the present invention are ~lmini.stered as pharrnaceuticals, to humans and :~nim~l~, they can be given alone or as a pharmaceutical composition cont~ining, for example~ 0.01 to 99.5% (more preferably~ 0. I to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
S Regardless of the route of ~lrnini.stration selected. the compounds of the present invention, which may be used in a suitable hydrated form. and/or the pharmaceutical compositions of the present invention. are formulated into pharrnaceutically acceptable dosage forms by conventional methods known to those of skill in the art.
A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the phalmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the I ~ dosage until the desired effect is achieved. In general. a suitable daily dose of a compositions of the invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above. It is preferred that a~lmini.stration be intravenous, intrarnuscular, intraperitoneal, or subcutaneous, preferably a-lmini~tered proximal to the site of the target. If desired, the effective daily dose of a therapeutic compositions may be ~dministered as two, three. four. five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally. in unit dosage forms. While it is possible for a compound of the present invention to be~-lmini.ctered alone. it is preferable to a~1minister the compound as a pharrnaceutical formulation (composition).
Therapeutic compositions can be ~clmini.~tered with medical devices known in the art. For example, in a preferred embodiment, a therapeutic composition of the invention can be atlmini~tered with a needleless hypodermic injection device, such as the devices disclosed in U.S. Patent Nos. S,399,163, 5.383,851, 5,312.335, 5.064~413, 4,941,880, 4,790,824, or 4,596,556. Examples of well-known implants and modules useful in the present invention include: U.S. Patent No. 4,487,603, which discloses an implantable micro-infusion pump for dispensing medication at a controlled rate;
U.S. Patent No. 4.,486.194, which discloses a therapeutic device for a~lmini.~tering medicants through the skin; U.S. Patent No. 4,447,233~ which discloses a medication infusion pump for delivering medication at a precise infusion rate; U.S. Patent No. 4,447,224, which discloses a variable flow implantable infusion apparatus for CA 022~9371 1999-01-0~
WO 98/02463 PCT~US97/12013 continuous drug delivery: U.S. Patent No. 4~439~196, which discloses an osmotic drug delivery system having mulli-chamber compartments; and U. S. Patent No. 4.475.196.
which discloses an osmotic drug delivery system. These patents are incorporated herein by reference. Many other such implants, delivery systems~ and modules are well known to those skilled in the art.
In certain embodiments, the compounds of the invention can be formulated to ensure proper distribution in vivo. For example~ the blood-brain barrier (BBB) excludes many highly hydrophilic compounds. To ensure that the therapeuticcompounds of the invention cross the BBB (if desired)~ they can be forrnulated~ for 10 example, in liposomes. For methods of manufacturing liposomes, see~ e.g.~ U.S. Patents 4,522,811; 5.374,548; and 5,399~331. The liposomes may comprise one or more moieties which are selectively transported into specific cells or organs ~ thus enhance tar,~eted drug delivery (see, e.g., V.V. Ranade (1989) J. Clin. Pharmacol. 29:685).
Exemplary targeting moieties include folate or biotin (see e.g., U.S. Patent 5.416,016 to 15 Low et al.); mannosides (Umezawa et al., (1988) Biochem. Biophys. Res. Commun.
153:1038); antibodies (P.G. Bloeman e~ al. (1995) FEBSLett. 357:140; M. Owais e~(1995) Antimicrob. Agents Chemother. 39: 180); surfactant protein A receptor (Briscoe et al. ( I 995) Am. J. Physiol. 1233: 134)~ different species of which may comprise the formulations ofthe inventions~ as well as components ofthe invented molecules; pl20 20 (Schreier et al. ( 1994) J. Biol. Chem. 269:9090); see also K. Keinanen; M.L. I .allkk~nen (1994) F~BSLett. 346:123; J.J. Killion; I.J. Fidler (1994) Immunomethods 4:273. ln one embodiment of the invention, the therapeutic compounds of the invention are formulated in liposomes; in a more preferred embodiment. the liposomes include atargeting moiety. In a most preferred embodiment, the therapeutic compounds in the 25 liposomes are delivered by bolus injection to a site proximal to the tumor or infection.
The composition must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the cont~min~ting action of microore~nicm~ such as bacteria and fungi.
A "therapeutically effective dosage" preferably inhibits tumor growth or 30 pathogen infection by at least about 20%~ more preferably by at least about 40%~ even more preferably by at least about 60%~ and still more preferably by at least about 80%
relative to untreated subjects. The ability of a compound to inhibit cancer or infectious disease can be evaluated in an animal model system predictive of efficacy in human tumors and infectious diseases. Alternatively~ this property of a composition can be 35 evaluated by ex~mining the ability of the compound to inhibit. such inhibition in vitro by assays well-known to the skilled practitioner. A therapeutically effective amount of a . ...

CA 022~9371 l999-ol-o~

therapeutic compound can decrease tumor size, prevent or delay death of infected tissues or organs decrease fever and while cell count? improve CD4 count or otherwise ameliorate symptoms in a subject. One oi'ordinary skill in the art would be able to determine such amounts based on such fàctors as the subject's size, the severity of the subject's symptoms, and the particular composition or route of ~clministration selected.
The invention is further illustrated by the following examples, which should not be construed as further limiting The contents of all references, pending patent applications and published patents, cited throughout this application. are hereby expressly incorporated by reference.
]O
EXAMPLES

The followin~ methodology described in the Materials and Methods section was used throughout these E~xamples. set forth below.
Materials and Methods Cell lines and Monoclonal antibodies The murine hybridoma producing cell line for the anti-FcaR antibody 20 used to construct the BsAb compositions described herein is A77 (Monteiro et al. 1992, J. lmmunol. 148: 1764-1770), and other anti-FcaRI hybridoma cell lines with properties the same or similar to A77 such as A3. A59 and A62 ((Monteiro et al. ~ 199~). can be used also. The hybridoma cell line for the anti-HER2/neu antibody is 520C9 (Frankel.
A. et al. 1985 J. Biol. Response Modif~ers 4:273-286; available from American Type 25 Culture Collection, ATCC, 12301 Parklawn Drive, Rockville, MD 2085~; accession number HB8696). Antibody H425 is a hllm~ni7~d anti-EGF-R antibody (Kettlesborough, C. et al., 1991, Prot. Eng. 4: 773-783). CC49 (anti-TAG 72) hybridoma was obtained from the ATCC (accession number HB 9459). mAb-producing cell lines were cultivated in Iscove's Modified Dulbecco's Medium (IMDM, Gibco/BRL, 30 Grand Island, NY) supplemented with 10% fetal bovine serum (FBS).
The mAb preparations were purified from the respective hybridoma supernatants byprotein A affinity chromatography (Bio-Rad, Richmond, CA).
SKBR-3 (Backm~n et al. Cancer Res 54: 2456-2461), a human breast carcinoma cell line. and SKOV-3 (ovarian carcinoma), which overexpress the HER2/neu 35 protooncogene, were obtained from ATCC. EGF-R over-expressing cell lines A431(skin carcinoma), HNS (head and neck carcinoma) and MDA-ME~-468 (breast CA 022~9371 l999-ol-o~
WO 98/02463 PCTtUS97/12013 carcinoma}~ were obtained from ATCC. Tumor cell lines were cultivated in Iscove's Modified Dulbecco's Medium (IMDM. Gibco/BRL~ Grand Island. NY) supplemented with 10% fetal bovine serum (FBS). The monocytoid cell line U937 (J. Immunol. 136:
1641 - 1647~ 1986) which expresses Fco~R was obtained from the ATCC and grown inRPMI-1640 plus 10% FBS (Gibco/BRL, Grand Island~ NY).
Candida albicans (strain ATCC 448.585) was grown in Sabouraud maltose broth (Difco, Detroit, MI) Preparation of Blood Cells Leukocytes were prepared from heparinized whole venous blood or from apheresis of norrnal human volunteers. Whole blood was diluted with RPMI containin~
5% dextran at a ratio of 2.5: 1 (v/v). The erythrocytes were allowed to sediment for 4 minutes on ice~ then the cells in the supernatant were transt'erred to a new tube and pelleted by centrifugation. The residual erythrocytes were removed by hypotonic Iysis.
15 The rem~ining Iymphocytes, monocytes and neutrophils were kept on ice until use in binding assays. For some experiments, neutrophils were separated from mononuclear cells by ficoll hypaque (Pharmacia-Upjohn, Piscataway~ NJ) gradient separation.
Monocytes were enriched from mononuclear cells by cold aggregation and settling through a cushion of fetal calf serum. Monocyte cultures were used fresh or were20 incubated at 37~C, 5% CO~ for 24 to 48 hours in teflon dishes at 4 x 106 cells/ml of MSFM containing 2.0% normal human serum type AB (Sigma, St. Louis~ MO) and 500 IRU/ml IFN-y (R&D Systems, Minneapolis, MN). Neutrophils were cultured for 24 to48 hours (37~C, 5% CO2) in AIM V media (Gibco/BRL~ Grand Island. NY) with 50 ng/ml G-CSF (Kindly provided by R. Repp, U. of Erlanger~ Germany) and 500 IRU/ml25 IFN-~.

Binding byflo~ cytometry The binding of the BsAb to FcaR and ~ER2/neu was assessed by flo~A-cytometry. Various concentrations of BsAb diluted in PBS, pH 7.4 containing 2mg/ml 30 BSA and 0.05% NaN3 (PBA), were incubated with SKBR-3 cells or with human leukocytes for one hour at 0~C. The cells were washed with PBA and incubated with a phycoerythrin labeled goat anti-mouse antibody for one hour at 0~C. The cells were washed and fixed with I % paraformaldehyde, and cell associated fluorescence wasanalyzed on a Becton Dickinson (Mountain View. CA) FACScan.
To assess whether IgA binding interfered with mAb A77 binding, A77 was incubated with TNF-treated cells in the presence of an excess of human IgA, and ~ .. . .

CA 022~9371 1999-01-0~

~XI-064CPPC ; ~.L - ~-. . , , ~

compared to controls incubated in the absence of IgA. A77 binding ~as detected with a phvcoerythrin-labeled goat anti-mouse antibody as above. Cells were washed and fi~ed with I % paraformaldehyde. and cell associated fluorescence was analyzed on a Becton Dickinson FACScan. In addition, binding human IgA to U937 cells was assessed in the 5 presence of A77 mAb. and compared to controls in the absence of excess A77 mAb.
IgA binding was detected with FITC labeled anti-human IgA antibod~

BsAb coupling procedure BsAb preparations were constructed using the method of Glennie et al. (J.
Immunol. 1987 139: 2367-2375). mAbs A77 (anti-FcaR), 520C9 (anti-HER2/~zeu) CC49 (anti-TAG 72) and H425 (anti-EGF-R) antibodies were produced by in vitro cultivation of the respective hybridoma cell lines. The antibody preparations were each digested with pepsin to produce F(ab')2 preparations~ and subsequently reduced to Fab' by addition of 10 mM mercaptoethanolamine (MEA) and incubation for 30 minutes atI 5 30~C. The Fab' fragments were applied to a Sephadex~ G-25 (Pharmacia-Upjohn.Piscataway, NJ) column equilibrated in 50mM Na Acetate, 0.5mM EDTA~ pH 5.3 (4~C).
One-half volurne of ortho-phenylenedimaleimide (o-PDM, 12mM) dissolved in dimethylformamide and chilled in a methanol/ice bath was added to the 5~0C9 Fab'. and the mixture was incubated 30 minutes at 0~C. The Fab'-maleimide was then separated from free o-PDM on Sephadex G-25 equilibrated in 50mM Na Acetate, 0.5mM EDTA, pH 5.3 (4~C).
For preparation of BsAb, the 520C9 Fab'-maleimide was added to an equimolar solution of the A77 Fab'. The reactants were concentrated under nitrogen to the starting volume using a Diaflo~ membrane in an Amicon~ (Lexington, MA) ~5 chamber, at 4~C, for 18 hours. The pH was then adjusted to 8.0 with 1 M Tris-HCI, pH
8.0, and the mixture was reduced with 10mM MEA (30 minutes, 30~C) and alkylated with 25 mM iodoacetamide. A77XCC49 and A77XH425 were produced using similar procedures. The bispecific F(ab')2 reactant preparation was separated from unreacted Fab's and other materials using a Superdex~ 200 column (Pharmacia-Upjohn, Piscataway, NJ) equilibrated in PBS.
Analysis of A77X520C9 BsAb by HPLC showed that this BsAb comprised of two main species; 75-85% of F(ab')2 heterodimer (100kDa, 10.43 mins) and 15-25% of F(ab')3 heterotrimer (~150 kDa, 9.86 mins) (Figure 1). Based on the method of preparation, the F(ab')3 species is expected to be comprised of two 520C9 F(ab') and one A77 F(ab'). A control experiment, in which a F(ab')-o-PDM derivative was incubated without a non-derivatized second F(ab'), confirmed that F(ab')-o-PDM
_ SUBSTITUTE PAGE
~ ~MEN~D SHEE~

CA 022~9371 1999-01-0~

MXI-064CPPC ~ 4~
~ . .
~ . .
~ ~ ~ ~ . O .

derivative does not cross-link with itself since all the hinge-SH _roups are occupied by o-PDM and no free -SH group is available for linkage (data not shown). Thus the F(ab')2 and F(ab'), species in these BsAb preparations are hetero-complexes of F(ab') fragments with two different specificities.
s ~ntibody dependent cellular cytofoxicity (~DCC) Human breast carcinoma cells, SKBR-3, which over-express HER2/neu, were used as target cells for determination of Iysis using multispecific compositions comprising binding determin~nt~ for HER2/neu. Other target cell lines were used in tests of molecules with different antigen-binding determin~nt~, for e~ample, A43 I cells for EGF-R, KLEB for TAG 72, etc. Effector cell samples were obtained by using heparinized whole blood, purified neutrophils (purified as described supra), or monocytes prepared from Leukopaks~ obtained from Advanced Biotechnologies Inc.
(Columbia, MD) as previously described (Guyre, P.M. et al. 1989, J: Immunol. 193:
1650). To prepare for use as effector cells, monocytes were cultured in Teflon(~containers in Macrophage Serum-Free Medium (GibcolBRL) containing 2% human serum for 24 to 48 hours. Target cells were labeled with 100 ~lCi of 51 Cr for one hour prior to combining with effector cells and antibodies in a U-bottom microtiter plate.
After incubation for 16 to 18 hours at 37~C, supernatants from each well were collected 0 and analyzed for radioactivity. Cytotoxicity was calculated by the formula: % Iysis =
(experimental CPM - target leak CPM/detergent Iysis CPM - target leak CPM) X 100%.
Further, specific Iysis = % Iysis with antibody - % Iysis without antibody. Assays were performed in triplicate.

Fluoresceination of ~ntibodies The pH of mAb solution was adjusted to 9.3 by the addition of 0.1 M
Na2CO3. Fluorescein iso-thiocyanate (FITC, Sigma, St. Louis, MO) was dissolved in DMSO at a concentration of 2mg/ml. Forty ~g of FITC was added for each milligramof mAb and incubated for two hours at room temperature. The fluoresceinated mAb was separated from the free FITC by G-25 chromatography.

l~odulation of Fcc~R by mAb ,4 77 The ability of mAb A77 to modulate the number of FcaR on the surface of human monocytes was assessed by incubating monocytes with various dilutions of MAb A77 37~C for 18 hours (or with mAb 520C9 as an isotype control). Monocytes were then washed with PBA, and incubated for one hour at 0~C in the presence of SUBSTITUTE PAGE
~t~liENG~D SHEET

CA 0 2 2 ~ 9 3 7 1 I g 9 9 - o l - o ~

human serum IgA at 10011g/ml. Cells were further washed with PBA, and IgA binding to FcaR~ was detected with FITC-labeled anti-human IgA antibody. Percent modulation = I - (MFI of the sample/MFI of the control ) x 100%, where MFI is the mean fluorescence ntensit~.

BsAb-mediated Phagocytosis Assay of monocyte and neutrophil-mediated phagocytosis of SKBR-3 cells was performed with SKBR-3 target cells labeled with the lipophilic red fluorescent dye PKH 26. Buffy coat cells purified from heparinized whole blood containing 10 monocytes, neutrophils~ and Iymphocytes were incubated with the labeled targets at 37~
for 6 hours in the absence or presence of BsAb. Monocytes and neutrophils were stained with FITC labeled anti-CD14 mAb (AML-2-23) at 0~C. and cells were washed and analyzed by two color fluorescence by FACScan. Percent phagocytosis is expressed as the percent of effector cells (monocytes or neutrophils) that have PKH 26 stain 15 associated with them.

Example 1. Bispecific AntibodY A77X520C9 Binds Effector Cells To deterrnine efficacy of the bispecific antibody BsAb A77X520C9 in killing breast cancer cells in a patient. ability to specifically bind to breast cancer cells in 20 culture was determined. For these experiments, cells of line SKBR-3. which overexpresses the HER2/neu oncogene~ were used. The 520C9 binding determinant derives from an anti-HER2/neu murine hybridoma (Frankel, A. et al., 1985 J. Biol.
Response Modifiers 4: 273-286).
Figure 2 shows that BsAb A77X520C9 bound to each of two types of 25 effector cells, neutrophils (PMNs) and monocytes. Figure 3 shows that, in whole blood~
A77X520C9 BsAb binds to Fcc~RI-expressing PMN and monocytes. but not to FcaR-negative Iymphocytes. This binding activity was not inhibited by the physiological levels of serum IgA in whole blood. which is sufficient to saturate all the Fc~RI
expressed in vivo.
~xample 2. Bispecific Antibod~/ A77X520C9 Binds Tar~et Breast Cancer Cells The BsAb A77X520C9 bound to the target breast tumor cells SKBR-3 derived from a breast tumor (Fig. 4) to the same extent as did the previously described BsAb MBX210 (Valone et al. 1995 J. Clin. Oncol. 13(9~: 2281-2292). Mean 35 fluorescence intensity (MFI) as a measure of binding was found to increase as a function of BsAb concentration when breast tumor cells were incubated with each of 0.1, 1.0 or CA 022~9371 l999-ol-o~

10 ~Lg/ml. The fraction of A77X520C9 BsAb that bound to breast tumor tar~et cells was e4uivalent to or greater than the fraction of the control BsAb MDX210 (FiC. 4) and binding to effector cells (Fig. 2) was similar as a function of concentration of each of the BsAbs.
Further data showing binding of A77X520C9 BsAbto HER2/neu positive SKBR-3 cells is shown in l~igure 5. As expected, only the BsAb and 520C9 F(ab')2 ~ but not A77 F(ab')2 control~ bound the target breast tumor cells.Data are shown in Figures 7-12 with a variety of effector cells~ with several independent bispecific antibody preparations~ and with varying concentrations of different preparations of BsAb.

Example 3 Bispecific Antibodv A77XH4~5 Binds Tar~et Skin Carcinoma Cells The monoclonal antibody H425 is specif1c for the EGF receptor (EGF-R).
which is overexpressed on cell lines derived from skin carcinoma (A431)~ and MDA-MB468 (breast carcinoma). The bispecific molecule A77XH425 which carries a binding determinant for EGF-R and f'or FcaRI. was prepared by the same chemical couplingprocedure as A77X520C9, and tested for binding to target cells by the methods described herein.
Similar to binding of A77X520C9 to HER2/neu-bearing target cells shown in Example 2 and Figures 2-5, Figure 6 shows that the EGF-R tumor-specificity of the BsAb A77XH425 causes this molecule to be bound to A431 tumor cells which overexpress EGF-R. Further, the tumor binding deterrninant H425 F(ab')2. but not A77 F(ab')2, which bears the FcaRI deterrninant. bound to the A431 target cells which overexpress EGF-R.
Example ~. Bindin~ of Bispecific Antibody A77X520C9 Mediates Effector LeukocYte Cytolvsis of HER2/neu-bearin~ Tar~et Cells Following determination of the ability of A77X520C9 to bind to breast tumor cells. its ability to bind both to target cells and to specific leukocytes such that cytolysis of the breast tumor cells is effected was determined also. Thus, antibody-dependent cellular cytolysis (ADCC) mPfli~ted by bispecific antibodies (BsAb; also referred to as bispecific molecules, BSM) of the invention is shown in Figures 7-16 as percent cytotoxicity or percent Iysis.
As shown in Figure 7, cytotoxicity of SKBR-3 cells mediated by A77X520C9 and neutrophil effector cellss (PMN) varied from 6% to 9% at 0.1 ~/ml,and increased to 20% to 30% at 1.0 ~Lg/ml. Similar data are shown for killing mediated by effector cells in whole blood in Figure 8. In Fi~ure 9 this cytotocity mediation by CA 022~9371 1999-01-0~

BsAb is shown to be inhibited by addition also of the Fc~ binding A77 F(ab')~. but not that of Fc~RI determinant of M22 F(ab')2.
Therapy of subjects with multispecific multivalent chemical compositions directly ~/~mini.~tered into the circulation requires that these agents function in whole 5 blood. Ability to function in whole blood~ i.e., to mediate cytolysis. is shown in Figure 8~ in which A77X520C9 preparations were found to mediate cytolysis of cultured breast tumor cells by blood effector cells. At 0.1 ,ug/ml. between 15% and 20% of the tumor cells were Iysed by A77X520C9, and at 1.0 ~g/ml cytolysis was approximately 25% to 30% of breast tumor cells. Since whole blood contains IgA at a concentration of 2 to 5 10 mg/ml, these data also show that cytotoxic activity of this BsAb is not inhibited by IgA.
These results show that BsAb can be delivered for therapeutic application it? vivo.
BsAb-mediated destruction of tumor cells by FcocRI-expressing cytotoxic effector cells was examined using freshly purified effector cells (monocytes and PMN) as well as whole blood as the source of effector cells. Figure 10 shows that I .0 ,ug/ml of A77X520C9 BsAb mediated up to 37% BsAb-dependent Iysis of HER2/nezJ positive SKBR-3 cells by purified PMN (neutrophils). This cytotoxic activity was dose dependent and saturated at 1.0 ~lg/ml of the BsAb. Fi~ure I I shows that A77X520C9 mediated up to 40% BsAb-dependent Iysis of SKBR-3 cells when purified monocytes were used as effector cells. Finally, A77X520C9 BsAb mediated up to 40% BsAb-dependent Iysis of the same target cells when whole blood was used as a source of effector cells (Figure 12). In the data shown in Figures 10-12, A77 F(ab')~ with the determinant for FcaRI, inhibited the ADCC activity of this BsAb~ but the anti-CD64 M22 F(ab')2 with the deterrninant for FcyRI, d id not. The background Iysis (in the absence of BsAb) in these experiments was about 10%.
Similarly, A77X520C9 BsAb mediated Iysis of another HER2/neu over-expressing tumor line, SKOV-3 (ovarian carcinoma line), in a whole blood ADCC assay.

Ex~mple 5 Binding of Bispecific Antibody A77X5H425 Mediates Effector LeukocYte CYtol~sis of EGF-R-bearin~ Tar~et Cells Similar to the data shown for BsAb for the tumor antigen ~ER2/neu.
A77XH425 BsAb, with the affinity determinant for EGF-R. mediated up to 52% BsAb-dependent Iysis of A431 target tumor cells with purified PMN as effector cells (Figure 13), up to 55% BsAb-dependent Iysis with purified monocytes as effector cells (Figure 14), and up to 43% BsAb-dependent Iysis with the whole blood a source of effector cells (Fi~ure 15). In these experiments A77 F(ab')2, but not M22 F(ab')2, inhibited the cytotoxic activity. The background Iysis (without BsAb) was about 10%.

CA 022~9371 1999-01-0~

These results show that the BsAb A77X520C9 can bind both effector and ~ER2/neu-bearing target cells. and can mediate target cytolysis by purified neutrophils and monocytes, and by these effector cells in whole blood. Lymphocytes in whole blood did not bind the BsAb. Further. the BsAb A77XH42~ can bind target cells that overexpress EGF-R~ and mediated cytolysis of these cells in the presence of effector cells.
In whole blood ADCC assays, A77XH425 BsAb-dependent Iysis (40-50% cytolysis) of HN5 (head and neck carcinoma line) and MDA-MB468 (breast carcinoma line), which express comparable levels of EGF-R as A431 (skin carcinoma 10 line), was observed.

Exa~?ple 6. Bispecific Antibody anti-TAG 72XA77 Mediates CvtolYsis of TAG 7~-Bearin, Tumor Cells The Examples above show that BsAb preparations are effective in 15 mediating cytolysis of cell of cell lines derived from tumors, bearing the tumor antigens HER2~neu, and EGF-R. To extend the potential use of BsAb. another composition with a binding determinant for the TAG 72 tumor marker was prepared by the coupling method described supra. The mucine antigen TAG 72 is found in the majority of breast, colon, ovarian and other cancers. A variety of antibodies that specifically bind TAG 72 20 are available, for example the monoclonal antibody produced by hybridoma CC49(ATCC HB 9459, Mezes, P. et al.~ International Application WO 90/04410).
CC49 was coupled to A77 to produce BsAb anti-TAG 72XA77. to target tumor cells bearing the TAG 72 antigen specifically to effector cells bearing FcaR.
Figure 16 shows neutrophil mediated antibody-dependent cytotoxicity bv the BsAb anti-25 TAG 72XA77 (constructed from CC49 and A77 mAb antibodies) of TAG 72-bearing tumor cells, as a function of concentration in ~lg/ml. The results in Figure 16 show that BsAb anti-TAG 72XA77 mediated cytolysis of tumor cells to a similar extent as BsAbs A77X520C9 for cells bearing HER2/neu, and A77XH425 for cells bearing EGF-R, shown in the Figures above.
Overall. Examples I -6 demonstrate cytolysis of tumor cells bearing three different tumor antigens mediated by the bispecific molecules of the invention. These Examples and other studies performed using cell lines taken from breast tumors. skin carcinoma, gastric carcinoma? head and neck carcinoma. and ovarian carcinoma~ shov~
that FcaRI-directed BsAb plcpaldtions coupled to binding deterrninants for a variety of 35 tumor antigens can be broadly applicable in a variety of m~lign~ncies. This observation is significant because target antigen restriction of FcyR-mediated ADCC activity of CA 022~9371 l999-01-0~
PCT~US97/12013 PMN has been reported for B eell Iymphoma associated anti~ens (Elasser. Det al 1996. Blood. 87,9:3803-3812).

~;xample 7. BsAb CvtolYsis Requires Access to FcaR Via the A77 Bindin~
Determinant To show that BsAb-mediated eytolysis of target eells is due to FeaR recognition by the A77 binding determinant, BsAb cytolysis was analyzed in the presenee of A77 F(ab')2 in Figures 9-15. If BsAb-mediated cytolysis functions bybinding to FeaR beeause of the A77-derived binding determinant~ then the addition of 10 A77 F(ab')2, but not an antibody with a different reeeptor binding deterrninant. eould eause inhibition of eytolysis of breast tumor eells.
The results shown in Figures 9-15 show that BsAb-mediated eytolysis funetions by binding to Feo~RI via the A77-derived binding determinant. In Figures 9-12. the addition of A77 F(ab')2 to the mixture of A77X520C9. target eells. and effector 15 cells causes inhibition of cytolysis, whi}e similar addition of an antibody that does not share the FeaRI has no effect on level of eytolysis. Figures 13-15 show this result also with BsAb A77XH425-mediated eytolysis of tumor eells: inhibition was observed in the presence 50 ~g/ml A77 F(ab')2, and not in the presenee of 50 ~g/ml of M22 F(ab')~.
M22 F(ab')2 speeifieally binds a different receptor, the Fe~RI receptor (Valone et cll., 20 1995 J Clin. Oncol. 13: 2281-2292).
These data show that BsAbs A77X520C9 and A77XH4'5 mediate eytolysis of tumor eells in a manner that is dependent on speeific binding of the BsAb to FcaRI on effector eells. Further, inhibition of BsAb killing by A77 F(ab')~ was found regardless of the nature of the effector eells.
~xample 8. Stimulation of T-eell Growth bv Antigen-Presentation Usin~ a Bispeeific Antibody For use of a BsAb for delivery of an antigen, the following proeedure was used to eouple the FcaRI binding determinant of mAb A77 to tetanus toxoid. Purified 30 tetanus toxoid (TT, Aeeurate Chemieal and Seientifie Company. Westbury, NY) was reaeted with sulfo-succidimidyl, 4-(N-maleimidomethyl) cyelohexane-l-earboxylate (s-SMCC, Pierce, Rockford. IL) at a molar ratio of 20 SMMC: I TT for two hours at room temperature. Free SMCC was removed from TT:SMCC by G-25 chromato~raphy. The F(ab')2 fragment of mAb, A77, was reduced to Fab' by incubating in the presence of 5 35 mM mercaptoethylamine (MEA, Sigma, St. Louis, MO) for 30 minutes at 30~C. Free MEA was removed from Fab' by G25 ehromatography. The A77 Fab' was incubated with the SMCC-treated TT for two hours at room temperature followed by an overnight - CA 022~9371 1999-01-0~
.\~IXI-06~CPPC ; 4~

incubation at 4~C. The A77-TT conjugate was purified from uncoupled Fab' by Superdex(~ 200 gel filtration (Pharmacia-Upjohn, Piscataway, NJ).
To assay T cell proliferation following antigen presention. TT-specific T
cell lines were generated from immunized individuals as previously described (Gosselin~-E.J. et al. J Immunol. 179:3477, 199~), monocytes were purified by cold aggregation as described previously (Guyre, P.M. et al. ~ Immunol. 143: 1650, 1989) and T cells (50ul of 106/ml) and autologous monocytes (50111 of 5Xl 05/ml) were added to wells of a 96 well tissue culture plate in the presence of various concentrations of TT or of A77-TT
and incubated at 37~ in a CO2 incubator. The relative number of T cells in each well 10 was assessed after incubation for four days by measuring lactate dehydrogenase (LDH) released from cells after Iysis using a kit purchased from Promega (Madison. WI). LDH
levels were quantified spectrophotometrically after addition of substrate and stop solution. Optical density was read at 490 nm using an ELISA plate reader (~Iolecular Devices. Palo Alto. CA).
Each data point presented in Figure 17 is the mean of data obtained from quadruplicate samples. with background value (monocytes and T cells in the presence of media only) subtracted. The A77-TT conjugate induced equivalent T cell proliferation at antigen doses that were 30- 100 fold lower than that of the uncoupled TT. These data show that directing the TT antigen to FcaRI by coupling to mAb A77 clearly enhanced '0 monocyte presentation of TT to TT-specific T cells. Similar antigen presentation can be achieved with antigens for allergies~ such as antigens of the dust mite, and cat and dog antigens, and for vaccine antigen presentation, such as for malaria, chicken pox. hepatitis virus C~ and other non-A-non-B hepatitis viruses.

25 Example 9 Bispecific Antibodv Binds Tar et Cells and Causes Pha ocvtosis bv Effector Cells To demonstrate BsAb mediation of phagocytosis, the following experiments were performed to determine the extent to which effector cells engulf SKBR-3 breast tumor cells or cell fragments in the presence of BsAb A77X520C9.
30 These studies tested phagocytosis of SKBR-3 by neutrophils, monocytes, and monocyte-derived macrophages (MDM). Phagocytosis of tumor cells mediated by Fcc~RI has not been described.
Tumor cells were labeled prior to exposure to leukocytes with lipophilic red fluorescent dye PKH26. Following incubation of effector cells with target cells for 6 35 hours, effector monocytes and neutrophils purified from whole blood were stained with FITC-labeled anti-CD14 mAb, and were analyzed by cell sorting. The results are shown in Figures 18-20. Percent phagocytosis is expressed as FITC-stained effector cells SUBSTITUTE PAGE
AMEN~D SHEET

CA 0 2 2 ~ 9 3 7 1 I g 9 9 - o l - o ~

associated with the lipophilic red dye As shown in Figure 18~ percent effector cells engaged in phagocytosis was on the order of 65% to 75%, depending on whether monocytes or neutrophils were tested. Increasing the bispecific antibody concentration above I .0 ~lg/ml did not enhance the percent of effector cells engaged in phagocytosis.
Monocytes differentiated into macrophages (MDM) are known to mediate phagocytosis oftumor cells (Ely? P.et al. 1996, Blood. 87:3813-3821). Todetermine if A77X520C9 BsAb could induce phagocytosis by MDM, these effector cells were incubated with dye-labeled SKBR-3 cells in the presence of varying concentrations of BsAb. The level of phagocytosis was determined by 2-color flow cytometric analysis, in which one fluorescent label is denoted FLI and the other is denoted FL2. Figure 19 shows that the MDM (panel 1, FL1+, FL2-) and SKBR-3 cells (panel 2, FL1-, FL2+) are distinguished from each other in a mixture of these two cell types by their unique fluorescence patterns (panel 3). When the A77~520C9 BsAb was added to the mixture of these target and effector cells, the BsAb mediated nearly a complete loss of tumor cells (panel 5, lower right quadrant). This was confirmed by an almost total lack of tumor cells that could be recovered from the BsAb cont~ininE wells as deterrnined by tumor specific ELISA (data not shown). As expected. MDM alone mediated some phagocytosis (~45%) of SKBR-3 cells without the BsAb (panel 3). However, addition of 0.1 llg/ml A77X520C9 was sufficient to enhance the phagocytosis to >95% (panel 5).
This BsAb-mediated phagocytic activity was almost completely inhibited by A77 F(ab')2 (panel 6). Furthermore, a mixture of uncoupled A77 F(ab')2 and 520C9 F(ab')2 could not enhance phagocytosis (panel 4), indicating the need for a conjugated A77 F(ab')x 520C9 F(ab') BsAb to target tumor cells to effector cells leading to activation of the effector cells (MDM).
Figure 20 shows that the BsAb-mediated phagocytic activity was dose dependent and saturated (nearly complete loss of tumor cells) at 0.1 ~lg/ml of the BsAb.
A77 F(ab')2 almost completely blocked this BsAb-mediated phagocytosis. Again, a mixture of uncoupled A77 F(ab')2 and 520C9 F(ab')2 had no activity to mediate phagocytosis.
The above-described studies show that~ in addition to promoting extracellular Iysis, the FcaRI-directed BsAb mediated potent phagocytic activity. In particular, when MDM were used as effector cells, nearly 100% of tumor cells were phagocytosed. It is likely that BsAb-mediated both ADCC and phagocytic activities simultaneously. and entire cells or fragments of Iysed cells were phagocytosed by the MDM via FcaRI. These two mech~ni.~m~ have been shown to occur in concert for FcrRI-mediated phagocytosis (Ely, P.et al. 1996? Blood. 87:3813-3821). It has been shown CA 022~9371 l999-ol-o~

that antigen presenting phagocytic cells (such as MDM) can present antigens via both class I and class 11 pathways after phagocytosis of antigen bearing particles (Falo~ ~.D.
et al. 1995. Nature Medicine. 1,7:649-656). Thus the potent phagocytosis of tumor cells Yia FcaRI can lead to activation of both humoral and cellular immune functions 5 specifically directed to tumor associated antigens.
These cytotoxic activities of Fca~l-directed BsAb are of therapeutic value because FcaRI expression is limited primarily to immune effector cells that are herein demonstrated to mediate BsAb-dependent cytotoxic activities, vi ., PMN, monocytes and macrophages ( Morton, H.C.et al. 1996.Critical Reviews in Immunolog~.
10 16:423). Because distribution of FcaRI is limited primarily to cytotoxic effector cells and because of its potent triggering activity, BsAb comprising a binding determinant to FcaRI can be of general use for cell-mediated immunotherapies. Furthermore. the approach described herein can be employed to prepare FcaRI-directed BsAb utilizing the existing high affinity tumor-specific IgG mAbs, that is, the binding determinant 15 components of the anti-FcaRI BsAb can be IgG molecules, since they are converted into F(ab')~ fragments prior to chemical coupling. Further~ for construction of recombinant BsAb~ the Fc portion of the IgG can be removed by appropriate restriction digestion and further exonuclease digestion as ap~ulopliate. The ability to use binding determinants from the Fab portion of IgG molecules will obviate the need to generate new IgA class 20 tumor specific mAbs toexploit the cytotoxic potential of FcaRI.

Example 10. Pha~ocytosis of the fun~al patho~en Candida alhicans Bispecific and multispecific antibody compositions of the invention can be used to direct FcaRI-bearing leukocytes a~ainst antigens from microbial pathogens, 25 for example, against bacteria, viruses, protozoan and metazoan parasites, and funs~i. By way of example, the following studies demonstrate that BsAb mediates phagocytosis of Candida albicans, a pathogenic yeast which causes deleterious infections in T-cell deficient immunocompromised patients.
A bispecific antibody directed against this pathogen was produced from 30 rabbit polyclonal-anti Candida IgG (Biodesign, Kennebunk, ME). F(ab')2 fragments of this polyclonal IgG were treated with sulfo-SMCC to add maleimide to free amino groups. This conjugate was reacted with equimolar A77 F(ab'), yielding the A77Xanti-Candida BsAb, which was purified from the uncoupled fragments by chromatography on Superdex 200 (Pharmacia, Piscataway, NJ). The A77Xanti-Candida bound effector35 and target cells consistent with the specificity of the component antibodies.

~, . . . ..

CA 022~9371 1999-01-0~

.~vL X I--064 C P P C , ~

Candida albicans cells cultured overnight at 37~C were harvested by centrifugation, washed three times with phosphate-buffered saline (PBS)~ and fluorescently labeled by incubation with FITC (Sigma, St. Louis, MO) at a concentration of 0.1 mg/ml in 0.1 M NaH2PO4/Na2HPO4 buffer (pH 9.6) for 30 minutes at 4~C.
Fluorescent fungal cells washed three times with PBS (5 x 105) were incubated for 30 minutes at 37~C with 2 x 105 isolated neutrophils in the presence of 10 ~lg/ml A77Xanti-Candida (A77 X ~Candida) BsAb; controls were incubated without BsAb.
Phagocytosis was quantitated by measuring FITC-fluorescence intensities of cells on a flow cytometer using PE-conjugated CD 11 b MoAb (Becton Dickinson~ San Jose~ CA)as a gate marker for neutrophils, and phagocytosis was determined also in cytospin preparations and evaluated by microscopy. Each experiment was repeated three times.
Phagocytosis of Candida by neutrophils, the most important effector cell population against pathogenic fungi such as Candida and Aspergillus species (Pizo, P
1984, Cancer 54 2649; Schaffner, A. et al, 1986, J. Clin. Invest. 78:511), and the stimulation of phagocytosis by the bispecific antibody of the invention. are shown in Fig. 21. In the presence of A77 X anti-Candida, 37.3% + 7.5 % of neutrophils were found to have phagocytosed fungal particles; in the absence ofthis BsAb 4.2 + 1.5% of neutrophils had ingested fungal particles (average of three experiments). Thus the FcaRI BsAb caused 8.9-fold stimulation of neutrophil phagocytosis of the fungal 20 pathogen as demonstrated photographically in panels A and B of Fig. 21, and by the shift in fluorescence intensity of the FITC-labeled fungal cells to that of PE-labeled neutrophils, in panels C and D.
FcaRI-directed BsAb can be used to combat a number of infectious diseases, since most infectious agents (bacteria, viruses, fungi etc.) express unique 25 antigens on their surface and several pathogen-specific antibodies have been described.
This technology can be applied to combat antibiotic resistant pathogens such as methicillin resistant Staphylococcus aureus, or other pathogenic bacterial species, and to pathogenic fungal species in addition to Candida.

30 Example 11. BsAb-Mediated Cytolysis With Effector Cells Treated With CYtokines and Growth Factors Figure 22 shows percent cytotoxicity of SKBR-3 breast tumor target cells, as a function of concentration of A77X520C9 BsAb, by neutrophil effector cells pre-incubated with G-CSF (Panel A) or both G-CSF and IFN-l~ (Panel B). Neutrophils 35 were pre-incubated with these factors overnight at 37~ prior to the cytotoxicity assay. In comparison with the data described in Example 4, in which neutrophils were not pre-incubated with cytokines, percent cytotoxicity was here found to be enhanced by pre--SUBSTITUTE PAGE
AMEND~D SHEET
. . .

CA 022~9371 l999-ol-o~

incubation of effector cells with both G-CSF and IFN-~ such that at 0.1 ~g/ml BsAb~
greater than 10% of tumor cells were specifically Iysed compared to less than 10% seen in Example 1, and at 1.0 ,ug/ml BsAb, greater than 40% cytotoxicity was observedcompared to approximately 24% to 32% in Figure 7 in the absence of these factors.
S Figure 23 shows the results of pre-treating effector monocytes with IFN-~
(Panel B) or TNF (Panel C), using BsAb A77XH425, which combines the binding determinant for FcaR with the binding deterrninant for EGF-R. As described above, EGF receptors are known to be overexpressed on carcinomas of the breast~ skin~ head and neck and other tumor cells, so that BsAb A77XH425 comprises another embodiment of a multispecific composition for treatment of breast and other tumors. As shown in Figure 23, untreated monocytes caused substantial breast cancer cell cytotoxicity mediated by A77XH425 BsAb~ at BsAb concentrations of 0.1 ~glml and 1.0 llg/ml. Specifically~ BsAb A77XH425 caused greater than 60% cytolysis at 1.0 !lg/ml with untreated monocytes and with TNF treated monocytes, and greater than 40% at I .0 ~glml with IFN-y-treated monocytes.
Overall, these results demonstrate that each of the two BsAb preparations, A77X520C9 and A77XH425, bind to breast tumor cells or skin carcinoma cells, respectively, and mediate cytolysis of the cancer cells by neutrophils or monocytes without additional treatment of these effector cells by exogenous cytokines.
~xample 1~. The A77 FcocR Bindin~ Site is Different From That of FcaR for its Natural Ligand In design of a therapeutic BsAb directed to FcaRI, optimal functionality as a therapeutic agent in subjects would be achieved if binding of this entitv were independent of competition with endogenous molecular species, such as the natural ligand IgA. Thus a therapeutic BsAb or multispecific binding composition could be ~lministered, and not be blocked or inhibited, or not be substantially blocked or inhibited, by endogenous IgA.
Figure 24 shows that A77 antibody bound to the full extent to effector cells in the presence of IgA at 200 ~lg/ml, compared to control binding in the absence of IgA. Mean fluorescence intensity was unaffected by the presence of IgA. These results demonstrate that A77 mAb specifically binds an epitope on FcaRI that is different from the site for binding of IgA (Monteiro et al. 1992, J. Immunol. 1~8: 1764-177~).

CA 022~9371 l999-ol-o~

Exam~7le 13. FcaRI Modulation bY A77. and Absence of Modulation by A77X520C9 and A77 F(ab')~
Characterization of the effect of addition of A77 mAb to cells on FcaRI
regulation was achieved by studies in which modulation (decrease in receptor number) 5 of FcaRI from the cell surface was examined as a function of concentration of A77 mAb.
As shown in Figure 25, incubation of monocytes with various concentrations of A77 for 18h at 37~(~ caused modulation at 10 nanograms /ml, which reached a plateau at 55% to 60% of control (number of receptors in the absence of A77) at 1.0 to 10 llg/ml. In contrast. incubation of monocytes with antibody 520C9. whicll has the same isotype as A77 and which specifically binds the HER2/nell receptor that is not expressed on monocytes, had no effect on monocyte modulation of FcaR. Thus the A77 mAb functional determinant is capable of causing internalization and modulation of FcaR from the surface. Further, ability of the BsAb to bind HER2/neu by virtue of a binding deterrninant derived from 520C9 is independent of the FcaRI binding determinant. This result shows that down modulation of FcaR is achieved by incubation of cells bearing this receptor with antibody A77. Such modulation can be used for re~ulation of autoimmune disorders.
These f;n~lin~ are confirrned and contrasted with data for the BsAb A77X520C9? as well as A77 F(ab')2, which show no down modulation of FcaRI in monocytes, and little in PMN compared to that of A77. The modulation of FcaRI
expression upon BsAb or A77 mAb binding to monocytes or PMN was examined by flow cytometry. Figure 26 shows that 1 and 10 ~g/ml whole A77 mAb induced about 40%-50% reduction of FcaRI on PMN and monocytes after overnight incubation at 37~
C. This modulation activity did not require cross-linking of the bound A77 by an anti-murine antibody. However, 1 and 10 llg/ml of A77X520C9 BsAb or A77 F(ab')2 induced minimum or no modulation of FcaRI under the similar conditions, indicating that the Fc region of the A77 mAb may be required to down modulate FcaRI expression.
From the data of Figure 26 showing modulation by A77 mAb but not by BsAb or A77 F(ab')2~ it can be concluded that BsAb binding to monocytes and PMN
does not lead to cross-linking and subsequent down modulation of FcaRI in the absence of target antigen or cells. Therefore, the Fc~RI-directed BsAb can be used to arrn" the effector cells in a subject or a patient, without activation by receptor cross-linlcing, thus avoiding undesired systemic side effects. Such effector cells armed with multispecific antibodies of the invention can be activated locally only upon cross-linking of FcaRl by binding a target antigen, for example, an antigen on a tumor cell or on a pathogen.
Similar arming of monocytes by FcyRI-directed BsAb has been shown (Valone~ F.H.et CA 022~9371 1999-01-0 ~ X I - 06~ C P P C

al. 1995. J Clin Oncol. I 3 :~81-229'')~ however. for Fc~RI-directed BsAb arming tO
be effective~ pretreatment of the subject with G-GSF or IFN-y is required to engage the P.~l~! effector population in vivo (Repp, R.el al. 1991. Blood. 78:995; Weber J.S.et al. 1996. Proc. of ~SCO. I ~:354. (Abstr.) FcaRI-directed BsAb described here could S engage effector cells such as monocytes~ PMN and macrophages without cytokine pretreatment, and without down-modulation of the cognate receptor on the leukocytes.

Example 1~. Clonin~ and Sequencin~ A77 Variable Re ion Genes A77 RNA was prepared from A77 FcaR specific antibody producing hybridoma cells, and 33 ~g of total RNA was obtained from approximately 4 x 10 ~ A77 cells using the Rl\~Aeasy Total RNA kit (Qiagen). RT-PCR was then done on 200 ng of the total RNA preparation using the GeneArnp~ Therrnostable rTlh Reverse Transcriptase RNA PCR kit (Perkin Elmer). Ig V region cDNAs were made using primers CGlFOR. 5'-GG,4AGC17AGACAGATGGGGGTGTCGTTTTG~ SEQ ID NO:
I 5 I (encoding arnino acids 115-122 of the murine IgGl heavy chain CH I domain and a HindIII site) and CK2FOR?5'-GGAAGC7'TGAAGATGGATACAGTTGGTGCAGC.
SEQ ID NO: 2 (encoding amino acids 111 - 118 of the murine K light chain constant domain and a HindIII site).
The VH and VK cDNAs were arnplified by PCR using the cDNA primers ~0 along with SHlBACKBAM, 5'-GACTGGATCCATGGRATGGAGCTGGRTCWTBHTCTT, SEQ ID NO:, (encoding a consensus sequence of arnino acids -20 to - 12 of some VH signal peptides and a BamHI site) and VKlBACKBA~I, 5'-GACTGG,4TCCGACATTCAGCTGACCCAGTCTCCA, SEQ ID NO: 4 (encoding ~5 arnino acids -4 to - 1 of the signal peptide and residues 1 to 4 of some murine VK
domains and a BamHI site). The single-letter code for combinations of nucleotides, known to those of skill in the art, is given on p. l 74 of the 1996- 1997 New England Biolabs catalog (32 Tozer Rd., Beverly, MA).
Arnplified VH and VK DNA were purified using Wizard PCR Prep kit 30 (Promega), cloned into pUC19, and sequenced by the dideoxy method. DNA
sequencing was carried out by National Biosciences, Inc. At least 5 pUC 19 clones of each VK and VH were sequenced to obtain consensus sequences of these genes, which are shown in Figs. 27 (SEQ ID NO: 5) and 28 (SEQ ID NO: 7). The predicted arninoacid sequences are shown in Figs. 27 (SEQ ID NO: 6) and 28 (SEQ ID NO: 8) for VK35 and VH respectively.

SUBSTITUTE PAGE
AMENG~,D SHEE~

CA 022~9371 1999-ol-o~

These sequences are used to obtain recombinant h~]m~ni7ed A77 FcaR-binding determinants~ to produce single-chain antibodies and single-chain BsAbs. for engineering determinants with greater affinity using recombinant methods, for modeling studies to develop mimetic drugs using rational drug design. and for additional 5 applications described in the instant invention.

~quivalents Those skilled in the art will recognize, or be able to ascertain usin~ no more than routine experimentation, many equivalents to the specific embodiments of the 10 invention described herein. Such equivalents are intended to be encompassed bv the following claims.
The contents of all patents and publications referred to herein are hereby incorporated by reference.

CA 022~9371 l999-ol-o~

SEQUENCE LISTING

(1) GENERAL INFORMATION:

(i) APPLICANT:
(A~ NAME: MEDAREX, INC.
(B) STREET: 1545 ROUTE 22 EAST, P.O. BOX 992 (C) CITY: ANNANDALE
(D) STATE: NEW JERSEY
(E) COUNTRY: US
(F) POSTAL CODE: 08801-0992 (G) TELEPHONE:
(H) TELEFAX:
(ii) TITLE OF INVENTION: THERAPEUTIC MULTISPECIFIC COMPOUNDS
COMPRISED OF ANTI-FC~ RECEPTOR ANTI80DIES
(iii) NUMBER OF SEQUENCES: 8 (iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: LAHIVE & COCKFIELD, LLP
(B) STREET: 60 State Street, Suite 510 (C) CITY: Boston (D) STATE: Massachusetts (E) COUNTRY: US
(F) ZIP: 02109-1875 (v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk (B) COMPUTER: IBM PC compatible (C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: ASCII TEXT
(vi ) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: PCT/US97/
(B) FILING DATE: 10 JULY 1997 (C) CLASSIFICATION:
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 08/678,194 (B) FILING DATE: 11 JULY 1996 (viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: REMILLARD, JANE E.
(B) REGISTRATION NUMBER: 38,872 (C) REFERENCE/DOCKET NUMBER: MXI-064CPPC
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (617)227-7400 (B) TELEFAX: (617)227-5941 (2) INFORMATION FOR SEQ ID NO:l:

CA 022~9371 1999-ol-o~

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 31 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: slngle (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: misc_feature (B) LOCATION: 1..31 (D) OTHER INFORMATION: /note= "PCR primer"

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:1:

(2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 32 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA
(ix) FEATURE:
(A) NAME/KEY: misc_feature (B) LOCATION: 1..32 (D) OTHER INFORMATION: /note= "PCR prlmer"

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:

(2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 36 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA

(vi) FEATURE:
(A) NAME/KEY: misc_feature (B) LOCATION: 1..36 (D) OTHER INFORMATION: /note= "PCR primer"
5 5 ( vi i ) FEATURE:

.

CA 022~9371 l999-ol-o~

(A) NAME/KEY: misc_feature (B) LOCATION: 15,26 (D) OTHER INFORMATION: /note= "R is A or G"
5 (viii ) FEATURE:
(A) NAME/KEY: misc_feature (B) LOCATION: 29 (D) OTHER INFORMATION: /note= I'W is A or T~
(ix) FEATURE:
(A) NAME/KEY: misc_feature (B) LOCATION: 31 (D) OTHER INFORMATION: /note= "B is C or G or T~
(x) FEATURE:
(A) NAME/KEY: misc_feature (B) LOCATION: 32 (D) OTHER INFORMATION: /note= "H lS A or C or T"

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:

25 ( 2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 34 base pairs (B) TYPE: nucleic acid (c) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA
3 5 ( ix ) FEATURE:
(A) NAME/KEY: misc_feature (B) LOCATION: 1..34 (D) OTHER INFORMATION: /note= "PCR primer"

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:

45 ( 2) INFORMATION FOR SEQ ID NO:5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 336 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA

CA 022~9371 1999-01-0~
W O 98/02463 PCT~US97/12013 (lx) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..336 s (xi) SEQUENCE DESCXIPTION: SEQ ID NO:5:

Asp Ile Gln Leu Thr Gln Ser Pro Leu Thr Leu Ser Ile Thr Ile Gly Gln Pro Ala Ser Ile Ser Cys Lys Ser Ser Gln Ser Leu Leu Asp Ser Asp Gly Lys Thr Tyr Leu Asn Trp Leu Leu Gln Arg Pro Gly Gln Ser Pro Thr Arg Leu Ile Tyr Leu Val Ser Lys Leu Asp Ser Gly Val Pro Asp Arg Phe Thr Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile Ser Arg Val Glu Ala Glu Asp Leu Gly Ile Tyr Tyr Cys Trp Gln Gly Ala His Phe Pro Gln Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys (2) INFORMATION FOR SEQ ID NO:6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 112 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide ,. ._~

CA 022~9371 1999-01-0~
W O 98/02463 PCT~US97112013 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:6:
Asp Ile Gln Leu Thr Gln Ser Pro Leu Thr Leu Ser Ile Thr Ile Gly Gln Pro Ala Ser Ile Ser Cys Lys Ser Ser Gln Ser Leu Leu Asp Ser Asp Gly Lys Thr Tyr Leu Asn Trp Leu Leu Gln Arg Pro Gly Gln Ser Pro Thr Arg Leu Ile Tyr Leu Val Ser Lys Leu Asp Ser Gly Val Pro Asp Arg Phe Thr Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile Ser Arg Val Glu Ala Glu Asp Leu Gly Ile Tyr Tyr Cys Trp Gln Gly Ala His Phe Pro Gln Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys 25 ( 2) INFORMATION FOR SEQ ID NO:7:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 426 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA

(ix) FEATURE:
~A) NAME/KEY: CDS
(B) LOCATION: 1..426 , .

CA 0225937l l999-0l-0~
W O 98/02463 PCTrUS97/12013 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:7:

Met Gly Trp Ser Trp Val Ile Ile Phe Leu Leu Ser Gly Thr Ala Gly 51 5 lO 15 Ala His Ser Glu Ile Gln Leu Gln Gln Thr Gly Pro Glu Leu Val Lys Pro Gly Ala Ser Val Lys Ile Ser Cys Lys Ala Ser Gly Tyr Ser Phe ACT GAC TAC ATC ATA TTT TGG GTG AAG CAG AGC CAT GGA AAG AGC CTT 192Thr Asp Tyr Ile Ile Phe Trp Val Lys Gln Ser His Gly Lys Ser Leu Glu Trp Thr Gly Asn Ile Asn Pro Tyr Tyr Gly Ser Thr Ser Tyr Asn Leu Lys Phe Lys Gly Lys Ala Thr Leu Thr Val Asp Lys Ser Ser Ser Thr Ala Tyr Met Gln Leu Asn Ser Leu Thr Ser Glu Asp Ser Ala Val Tyr Tyr Cys Val Arg Gly Val Tyr Tyr Tyr Gly Ser Ser Tyr Glu Ala Phe Pro Tyr Trp Gly Gln Gly Thr Leu Val Thr Val Ser Ala 40 ( 2) INFORMATION FOR SEQ ID NO:8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGT~: 142 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:8:
Met Gly Trp Ser Trp Val Ile Ile Phe Leu Leu Ser Gly Thr Ala Gly Ala His Ser Glu Ile Gln ~eu Gln Gln Thr Gly Pro Glu Leu Val Lys .

CA 022~937l l999-Ol-0~

Pro Gly Ala Ser Val Lys Ile Ser Cys Lys Ala Ser Gly Tyr Ser Phe 5 Thr Asp Tyr Ile Ile Phe Trp Val Lys Gln Ser His Gly Lys Ser Leu Glu Trp Thr Gly Asn Ile Asn Pro Tyr Tyr Gly Ser Thr Ser Tyr Asn Leu Lys Phe Lys Gly Lys Ala Thr Leu Thr Val Asp Lys Ser Ser Ser Thr Ala Tyr Met Gln Leu Asn Ser Leu Thr Ser Glu Asp Ser Ala Val Tyr Tyr Cys Val Arg Gly Val Tyr Tyr Tyr Gly Ser Ser Tyr Glu Ala 20 Phe Pro Tyr Trp Gly Gln Gly Thr Leu Val Thr Val Ser Ala

Claims

What is claimed is:

1. A bispecific binding molecule. comprising a first binding determinant which binds to an Fc.alpha. receptor without being blocked by immunoglobulin A, and a second binding determinant which binds to a target antigen.

2. The bispecific binding molecule of claim 1, wherein the binding of the first binding determinant to Fc.alpha. receptor is not blocked by human immunoglobin A.

3. The bispecific binding molecule of claim 1, wherein the target antigen is a cancer cell antigen.

4. The bispecific binding molecule of claim 3, wherein the cancer cell antigen is selected from the group consisting of cancers of the breast, ovary testis, lung, colon, rectum, pancreas, liver, central nervous system, head and neck, kidney, bone, blood and lymphatic system.

5. The bispecific binding molecule of claim 1. wherein the target antigen is an infectious disease antigen from a pathogen or pathogen-infected cell.

6. The bispecific binding molecule of claim 3, wherein the cancer cell antigen is a member of the human EGF-like receptor family.

7. The bispecific binding molecule of claim 6, wherein the cancer cell antigen is an EGF receptor.

8. The bispecific binding molecule of claim 6, wherein the cancer cell antigen is HER-2/neu.

9. The bispecific binding molecule of claim 6, wherein the cancer cell antigen is selected from the group consisting of HER-3 and HER-4.

10. The bispecific binding molecule of claim 6, wherein the cancer cell antigen is a heterodimeric receptor comprised of at least one HER subunit.

11. The bispecific binding molecule of claim 3, wherein the cancer cell antigen is selected from the group consisting of carcinoembryonic antigen, gastrin releasing peptide receptor antigen. and mucine tumor antigen TAG 72.

12. The bispecific molecule of claim 1, wherein at least one of said binding determinants is an antibody or an antibody fragment.

13. The bispecific binding molecule of claim 12, wherein the antibody or antibody fragment is an IgG or IgG fragment.

14. The bispecific binding molecule of claim 12, wherein the antibody fragment is selected from the group consisting of an Fab, Fab', F(ab')2, Fv, and single chain Fv.

15. A bispecific binding molecule comprising a binding determinant which binds to an Fc.alpha. receptor without being blocked by immunoglobulin A and a binding determinant for a target selected from the group consisting of a cancer cell antigen and an antigen from a pathogen.

16. The bispecific binding molecule of claim 15, wherein the determinant which binds an Fc.alpha. receptor is humanized.

17. The bispecific binding molecule of claim 16, wherein the humanized determinant is derived from all or a portion of the nucleotide sequences encoding the Fv binding determinants of antibody A77 V K and V H regions shown in Figure 14 (SEQ ID
NO:5), and Figure 15 (SEQ ID NO:7), respectively.

18. The bispecific binding molecule of claim 16, wherein the humanized determinant is derived from all or a portion of the amino acid residue sequences of the Fv binding determinants of antibody A77 V K and V H regions shown in Figure 14 (SEQ
ID NO:6), and Figure 15 (SEQ ID NO:8), respectively.

19. The bispecific binding molecule of claim 18, wherein the humanized determinant is greater than 50% homologous to that of antibody A77 V K or V H regions shown in Figure 14 (SEQ ID NO:6), and Figure 15 (SEQ ID NO:8), respectively.

20. The bispecific binding molecule of claim 15, wherein the second binding determinant is selected from the group consisting of ATCC HB 8696 (antibody 520C9) ATCC HB 9459 (antibody CC49) and functional fragments thereof.

21. The bispecific binding molecule of claim 20 produced by chemical linkage of the antibody A77 fragment and the second binding determinant.

22. The bispecific binding molecule of claim 20 which is produced recombinantly in a host cell.

23. A multispecific binding molecule, comprising a first binding determinant which binds an Fc.alpha. receptor and a second binding determinant which binds one or more target antigens.

24. The multispecific binding molecule of claim 23, wherein the binding of the first binding determinant to Fc.alpha. receptor is not blocked by human immunoglobin A.

25. The multispecific binding molecule of claim 23 selected from the group consisting of a bispecific binding molecule and a trispecific binding molecule.

26. The multispecific binding molecule of claim 25, comprising a third binding determinant specific for an Fc receptor that is not an Fc.alpha. receptor.

27. The multispecific binding molecule of claim 26, wherein the third binding determinant binds an Fc receptor selected from the group consisting of Fc.gamma.
receptor, Fc.epsilon. receptor, Fc.delta. receptor and Fcµ receptor.

28. The multispecific binding molecule of claim 23, wherein binding of the Fc.gamma. receptor is not inhibited by human IgG.

29. The multispecific binding molecule of claim 28, wherein the second binding determinant binds to a target antigen selected from the group consisting of a cancer cell antigen, a pathogen antigen, and an antigen on a pathogen-infected cell.

30. The multispecific binding molecule of claim 28 produced by chemical linkage of said binding determinants.

31. The multispecific binding molecule of claim 23 produced recombinantlyin a host cell transfected with a nucleic acid encoding said binding determinants.

32. The multispecific molecule of claim 23, wherein at least one of said binding determinants is an antibody or an antibody fragment.

33. The multispecific molecule of claim 32, wherein at least one of said binding determinants is a humanized antibody or fragment thereof.

34. The multispecific binding molecule of claim 29, wherein the cancer cell antigen is from a cancer cell selected from the group consisting of cancer cells of the breast, ovary, testis, lung, colon, rectum, pancreas, liver, central nervous system, head and neck, kidney, bone, blood and lymphatic system.

35. The multispecific binding molecule of claim 29, wherein the pathogen antigen is selected from the group consisting of bacterial, fungal, protozoal, and viral antigens.

36. The multispecific binding molecule of claim 29, wherein the cancer cell antigen is a member of the human EGF-like receptor family.

37. The multispecific binding molecule of claim 36, wherein the cancer cell antigen is an EGF receptor.

38. The multispecific binding molecule of claim 36, wherein the cancer cell antigen is HER-2/neu.

39. The multispecific binding molecule of claim 36, wherein the cancer cell antigen is HER-3, HER-4, or a heteromultimeric receptor comprised of at least one HER
subunit.

40. The multispecific binding molecule of claim 29. wherein the cancer cell antigen is selected from the group consisting of carcinoembryonic antigen, gastrin releasing peptide receptor antigen, and mucine antigen TAG 72.

41. A multispecific binding molecule comprising a functional fragment of antibody A77 and a second binding determinant which binds an antigen selected from the group consisting of an antigen on a cancer cell, an antigen on a pathogen and an antigen on a pathogen-infected cell.

42. The multispecific binding molecule of claim 41, wherein the second binding determinant for an antigen on a cancer cell is selected from the group consisting of ATCC HB 8696 (antibody 520C9), ATCC HB 9459 (antibody CC49), and functional fragments thereof.

43. The multispecific binding molecule of claim 41, wherein the A77 antibody fragment binds to an IgA receptor on a white blood cell.

44. The multispecific binding molecule of claim 43. wherein the white blood cell is selected from the group consisting of a macrophage, monocyte, neutrophil, basophil, eosinophil, and lymphocyte.

44. The multispecific binding molecule of claim 41, wherein the pathogen is selected from the group consisting of bacteria, viruses, fungi and protozoans.

46. The multispecific binding molecule of claim 42. wherein the antibody or antibody fragment is an IgG or IgG fragment.

47. The multispecific binding molecule of claim 32, wherein the antibody fragment is selected from the group consisting of an Fab, Fab', F(ab')2, Fv, and single chain Fv.

48. The multispecific binding molecule of claim 23 comprising a third binding determinant which binds to an antigen of a target cell, wherein said second binding determinant binds to a different antigen on the same target cell.

49. The multispecific binding molecule of claim 23, comprising a third binding determinant which binds to an epitope of a target antigen, wherein said second binding determinant binds to a different epitope on the same target antigen.

50. The multispecific binding molecule of claim 45, wherein the pathogen is a fungus.

51. The multispecific binding molecule of claim 50, wherein the fungus is a species of the genus Candida.

52. The multispecific binding molecule of claim 51, wherein the species is C.
albicans.

53. The multispecific molecule of claim 50, wherein the binding determinant to Fca is not blocked by human immunoglobin A.

54. A method for eliminating or reducing an unwanted cell in a subject, comprising administering to the subject a therapeutically effective dose of a multispecific binding molecule, comprising a first binding determinant which binds an Fc.alpha. receptor and a second binding determinant which binds an antigen on the unwanted cell, in a pharmaceutically acceptable carrier.

55. The method of claim 54, wherein at least one of said binding determinants is humanized.

56. The method of claim 55, further comprising administering to the subject at least one agent that enhances the number or activity of Fc.alpha. receptors on an Fc.alpha.
receptor-bearing cell.

57. The method of claim 56, wherein the agent is a cytokine.

58. The method of claim 57, wherein the cytokine is selected from the group consisting of at least one of G-CSF, GM-CSF, IFN-.gamma., and TNF.

59. A method for treating a subject infected with a pathogen wherein the subject is administered a therapeutically effective dose of a multispecific binding molecule, said molecule comprising a first binding determinant which binds an Fc.alpha.
receptor and a second binding determinant which binds a target antigen of the pathogen or a pathogen-infected cell, in a pharmaceutically acceptable carrier.

60. A method for treating a subject to remove an unwanted cell in the subject, comprising obtaining a sample of blood or blood cells from the subject;
contacting said blood or blood cells ex vivo with a therapeutically effective dose of a multispecific binding molecule, said binding molecule comprising a first binding determinant which binds an Fc.alpha. receptor and a second binding determinant which binds one or more target antigens, in a pharmaceutically acceptable carrier; and returning said treated blood or blood cells to the subject.

61. The method of claim 60, wherein at least one of said determinants is humanized.
62. The method of claim 61, wherein said blood cells are isolated and expanded in culture.

63. The method of claim 61 for treating a subject for the presence of an unwanted cell, wherein said blood cells are treated with at least one agent that enhances the number or activity of Fc.alpha. receptors.

64. A method for vaccinating a subject against a pathogen or against a cancer, wherein the subject is administered a multispecific binding molecule, said molecule comprising a first binding determinant which binds an Fc.alpha. receptor and a second binding determinant which binds an antigen selected from the group consisting of an antigen on a cancer cell, an antigen on a pathogen and an antigen on a pathogen-infected cell, in a pharmaceutically acceptable carrier.

65. A method for arming effector cells of a subject against a pathogen oragainst a cancer, wherein the subject is administered a multispecific binding molecule, said molecule comprising a first binding determinant which binds an Fc.alpha. receptor and a second binding determinant which binds an antigen selected from the group consisting of an antigen on a cancer cell, an antigen on a pathogen and an antigen on a pathogen-infected cell, in a pharmaceutically acceptable carrier.

66. A method for modulating an Fc.alpha. receptor in a subject, comprising administering to the subject a composition comprising antibody A77 or a functional fragment of A77, in a pharmaceutically acceptable carrier.

67. A method for identifying for an agent which modulates Fc.alpha. receptors on the surface of cells, comprising contacting a sample of cells carrying Fc.alpha. receptors with the agent;
determining Fc.alpha. receptor activity in the sample with the agent, in a control sample with an antibody that modulates Fc.alpha. receptors, and in a control sample with cells not contacted with said agent or with antibody; and comparing Fc.alpha. receptor activities in the samples, such that a sample of cells contacted with said agent and having statistically significant less Fc.alpha.
receptor activity than control cells not contacted with agent, or having statistically significantly as low Fc.alpha. receptor activity as cells in a sample with an antibody, identifies an agent which modulates Fc.alpha. receptors on the surface of cells.

68 A method for designing an agent which modulates Fc.alpha. receptors for treatment of autoimmune disease by obtaining a three dimensional model of the A77 anti-Fc.alpha. receptor binding site using the sequence determinants of A77 heavy and light chain variable regions, comprising comparing the amino acid residues of the A77 variable region with that of heavy and light chain variable regions of antibodies of known threedimensional structure;
determining placement of non-homologous amino acid residues within the main peptide chain of the binding region of the V H and V K sites, such that the size, shape and charge of the A77 anti-Fc.alpha. receptor binding site is determined;
screening a library of molecules to obtain those of suitable size, shape and charge by computer modeling that are mimetics of the A77 binding site; and screening such candidates of appropriate size, shape and charge for activity as potential modulators of Fc.alpha. receptors, such that an agent which modulates Fc.alpha. receptors is designed.
CA002259371A 1996-07-11 1997-07-10 Therapeutic multispecific compounds comprised of anti-fc.alpha. receptor antibodies Abandoned CA2259371A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/678,194 1996-07-11
US08/678,194 US5922845A (en) 1996-07-11 1996-07-11 Therapeutic multispecific compounds comprised of anti-Fcα receptor antibodies

Publications (1)

Publication Number Publication Date
CA2259371A1 true CA2259371A1 (en) 1998-01-22

Family

ID=24721785

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002259371A Abandoned CA2259371A1 (en) 1996-07-11 1997-07-10 Therapeutic multispecific compounds comprised of anti-fc.alpha. receptor antibodies

Country Status (12)

Country Link
US (4) US5922845A (en)
EP (1) EP0914346B1 (en)
JP (1) JP3519415B2 (en)
CN (1) CN1230198A (en)
AT (1) ATE318845T1 (en)
CA (1) CA2259371A1 (en)
DE (1) DE69735367D1 (en)
HU (1) HUP9903951A3 (en)
IL (1) IL128003A (en)
PL (1) PL331091A1 (en)
RU (1) RU2201766C2 (en)
WO (1) WO1998002463A1 (en)

Families Citing this family (353)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6018031A (en) * 1989-10-20 2000-01-25 Trustees Of Dartmouth College Binding agents specific for IgA receptor
US7105159B1 (en) 1992-11-05 2006-09-12 Sloan-Kettering Institute For Cancer Research Antibodies to prostate-specific membrane antigen
US20040258688A1 (en) * 1995-01-31 2004-12-23 Daniel Hawiger Enhanced antigen delivery and modulation of the immune response therefrom
US20020187131A1 (en) * 1995-01-31 2002-12-12 Daniel Hawiger Enhanced antigen delivery and modulation of the immune response therefrom
US6685940B2 (en) * 1995-07-27 2004-02-03 Genentech, Inc. Protein formulation
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
US6136311A (en) 1996-05-06 2000-10-24 Cornell Research Foundation, Inc. Treatment and diagnosis of cancer
US7371376B1 (en) * 1996-10-18 2008-05-13 Genentech, Inc. Anti-ErbB2 antibodies
ZA9811162B (en) 1997-12-12 2000-06-07 Genentech Inc Treatment with anti-ERBB2 antibodies.
HUP0100929A3 (en) * 1998-02-17 2005-10-28 Celldex Therapeutics Inc Treating and diagnosing macrophage-mediated diseases using fc receptor ligands
EP2275540B1 (en) * 1999-04-09 2016-03-23 Kyowa Hakko Kirin Co., Ltd. Method for controlling the activity of immunologically functional molecule
US7041292B1 (en) 1999-06-25 2006-05-09 Genentech, Inc. Treating prostate cancer with anti-ErbB2 antibodies
DK2283866T3 (en) 1999-06-25 2015-05-18 Genentech Inc METHODS OF TREATMENT USING ANTI-ERBB ANTIBODY-MAYTANSINOID CONJUGATES
US6949245B1 (en) * 1999-06-25 2005-09-27 Genentech, Inc. Humanized anti-ErbB2 antibodies and treatment with anti-ErbB2 antibodies
US20030086924A1 (en) * 1999-06-25 2003-05-08 Genentech, Inc. Treatment with anti-ErbB2 antibodies
US20040013667A1 (en) * 1999-06-25 2004-01-22 Genentech, Inc. Treatment with anti-ErbB2 antibodies
EP1189641B1 (en) 1999-06-25 2009-07-29 Genentech, Inc. HUMANIZED ANTI-ErbB2 ANTIBODIES AND TREATMENT WITH ANTI-ErbB2 ANTIBODIES
JP4579471B2 (en) * 1999-06-25 2010-11-10 ジェネンテック, インコーポレイテッド Treatment of prostate cancer with anti-ErbB2 antibody
MXPA02000962A (en) * 1999-07-29 2002-07-02 Medarex Inc Human monoclonal antibodies to her2 neu.
ATE373017T1 (en) * 1999-07-30 2007-09-15 Medarex Inc THERAPEUTIC COMPOUNDS CONSISTING OF FC RECEPTOR-BINDING ACTIVE INGREDIENTS
EP2829609A1 (en) 1999-08-24 2015-01-28 E. R. Squibb & Sons, L.L.C. Human CTLA-4 antibodies and their uses
ES2330301T3 (en) 1999-08-27 2009-12-09 Genentech, Inc. DOSAGES FOR TREATMENT WITH ANTI-ERBB2 ANTIBODIES.
CA2385528C (en) 1999-10-01 2013-12-10 Immunogen, Inc. Compositions and methods for treating cancer using immunoconjugates and chemotherapeutic agents
DE10006432A1 (en) * 2000-02-14 2001-08-16 Ganzimmun Inst Fuer Ganzheitli Method for the detection of Helicobacter pylori in stool and saliva samples
US7097840B2 (en) 2000-03-16 2006-08-29 Genentech, Inc. Methods of treatment using anti-ErbB antibody-maytansinoid conjugates
US20010051147A1 (en) * 2000-03-27 2001-12-13 Van De Winkel Jan G.J. Methods for immunostimulation using binding agents for the Fc receptor of immunoglobulin A
ES2528794T3 (en) 2000-04-11 2015-02-12 Genentech, Inc. Multivalent antibodies and uses thereof
AU2001259271A1 (en) * 2000-04-28 2001-11-12 Millennium Pharmaceuticals, Inc. 14094, a novel human trypsin family member and uses thereof
CA2408594A1 (en) * 2000-05-08 2001-11-15 Medarex, Inc. Human monoclonal antibodies to dendritic cells
US7560534B2 (en) * 2000-05-08 2009-07-14 Celldex Research Corporation Molecular conjugates comprising human monoclonal antibodies to dendritic cells
CN102698265A (en) 2000-05-19 2012-10-03 杰南技术公司 Gene detection assay for improving the likelihood of an effective response to an erbb antagonist cancer therapy
US20030138870A1 (en) * 2000-06-27 2003-07-24 Megumi Tanaka Method of evaluating binding activity of ligand to lignad-binding protein
US6946292B2 (en) * 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
ES2390425T3 (en) 2000-12-22 2012-11-12 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Use of repulsive targeting molecules (RGM) and their modulators
US7754208B2 (en) 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
NZ527977A (en) 2001-02-12 2005-10-28 Medarex Inc Human monoclonal antibodies to FC alpha receptor (CD89)
US8163289B2 (en) * 2001-03-09 2012-04-24 Iterative Therapeutics, Inc. Methods and compositions involving polymeric immunoglobulin fusion proteins
EP1395605B8 (en) 2001-03-09 2014-12-17 Iterative Therapeutics, Inc. Polymeric immunoglobulin fusion proteins that target low-affinity fcgamma receptors
US20070160576A1 (en) 2001-06-05 2007-07-12 Genentech, Inc. IL-17A/F heterologous polypeptides and therapeutic uses thereof
EP1992643A3 (en) 2001-06-20 2008-12-10 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US20050271663A1 (en) * 2001-06-28 2005-12-08 Domantis Limited Compositions and methods for treating inflammatory disorders
WO2004058821A2 (en) * 2002-12-27 2004-07-15 Domantis Limited Dual specific single domain antibodies specific for a ligand and for the receptor of the ligand
DE60237282D1 (en) * 2001-06-28 2010-09-23 Domantis Ltd DOUBLE-SPECIFIC LIGAND AND ITS USE
US6997863B2 (en) * 2001-07-25 2006-02-14 Triton Biosystems, Inc. Thermotherapy via targeted delivery of nanoscale magnetic particles
US7731648B2 (en) * 2001-07-25 2010-06-08 Aduro Biotech Magnetic nanoscale particle compositions, and therapeutic methods related thereto
US7951061B2 (en) * 2001-07-25 2011-05-31 Allan Foreman Devices for targeted delivery of thermotherapy, and methods related thereto
US7074175B2 (en) 2001-07-25 2006-07-11 Erik Schroeder Handy Thermotherapy via targeted delivery of nanoscale magnetic particles
WO2003024392A2 (en) 2001-09-18 2003-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US8323903B2 (en) 2001-10-12 2012-12-04 Life Technologies Corporation Antibody complexes and methods for immunolabeling
AU2002365258A1 (en) * 2001-10-12 2003-09-02 University Of Vermont And State Agricultural College Binding peptides specific for the extracellular domain of erbb2 and uses therefor
US20050069962A1 (en) * 2001-10-12 2005-03-31 Archer Robert M Antibody complexes and methods for immunolabeling
WO2003088808A2 (en) 2002-04-16 2003-10-30 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
AU2002356844C1 (en) * 2001-10-23 2010-03-04 Amgen Fremont Inc. PSMA antibodies and protein multimers
US20050215472A1 (en) 2001-10-23 2005-09-29 Psma Development Company, Llc PSMA formulations and uses thereof
EP1575571A4 (en) 2002-01-02 2008-06-25 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor
US7691568B2 (en) 2002-04-09 2010-04-06 Kyowa Hakko Kirin Co., Ltd Antibody composition-containing medicament
CA2481515C (en) 2002-04-10 2013-10-01 Genentech, Inc. Anti-her2 antibody variants
US7799561B2 (en) * 2002-06-12 2010-09-21 Sigma-Aldrich, Co. Affinity peptides and method for purification of recombinant proteins
US7696320B2 (en) 2004-08-24 2010-04-13 Domantis Limited Ligands that have binding specificity for VEGF and/or EGFR and methods of use therefor
US9321832B2 (en) * 2002-06-28 2016-04-26 Domantis Limited Ligand
PT1517921E (en) * 2002-06-28 2006-09-29 Domantis Ltd DIFFERENTLY SPECIFIED LIGANDS WITH SEMI-LIFE IN THE INCREASED SERUM
PT1585966E (en) 2002-07-15 2012-02-20 Hoffmann La Roche Treatment of cancer with the anti-erbb2 antibody rhumab 2c4
US20080260744A1 (en) 2002-09-09 2008-10-23 Omeros Corporation G protein coupled receptors and uses thereof
JP2006513702A (en) * 2002-09-09 2006-04-27 ヌラ インコーポレーティッド G protein-coupled receptor and use thereof
EP1551876B1 (en) 2002-10-16 2011-03-16 Purdue Pharma L.P. Antibodies that bind cell-associated ca 125/0722p and methods of use thereof
US9701754B1 (en) 2002-10-23 2017-07-11 City Of Hope Covalent disulfide-linked diabodies and uses thereof
WO2004043375A2 (en) * 2002-11-08 2004-05-27 Bristol-Myers Squibb Company Pharmaceutical compositions and methods of using taxane derivatives
WO2004048525A2 (en) * 2002-11-21 2004-06-10 Genentech, Inc. Therapy of non-malignant diseases or disorders with anti-erbb2 antibodies
US20080014594A1 (en) * 2003-01-31 2008-01-17 Kevin Hestir Lung-Expressed Polypeptides
CA2514979A1 (en) * 2003-01-31 2004-09-02 Celldex Therapeutics, Inc. Antibody vaccine conjugates and uses therefor
US9259459B2 (en) * 2003-01-31 2016-02-16 Celldex Therapeutics Inc. Antibody vaccine conjugates and uses therefor
US20040156846A1 (en) * 2003-02-06 2004-08-12 Triton Biosystems, Inc. Therapy via targeted delivery of nanoscale particles using L6 antibodies
JP3803790B2 (en) * 2003-02-17 2006-08-02 株式会社東北テクノアーチ Novel diabody-type bispecific antibody
US8088387B2 (en) 2003-10-10 2012-01-03 Immunogen Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates, and methods of making said conjugates
SI1641822T1 (en) 2003-07-08 2013-08-30 Genentech, Inc. Il-17 a/f heterologous polypeptides and therapeutic uses thereof
WO2005048928A2 (en) * 2003-11-12 2005-06-02 George Mason Intellectual Property Methods for treating viral infection
EP1689432B1 (en) 2003-11-17 2009-12-30 Genentech, Inc. Compositions and methods for the treatment of tumor of hematopoietic origin
EP1570858A1 (en) * 2004-03-05 2005-09-07 Institut National De La Sante Et De La Recherche Medicale (Inserm) Monovalent ligand of the FCalphaRi receptor as an anti-inflammatory agent
MXPA06011199A (en) 2004-03-31 2007-04-16 Genentech Inc Humanized anti-tgf-beta antibodies.
US20150017671A1 (en) 2004-04-16 2015-01-15 Yaping Shou Methods for detecting lp-pla2 activity and inhibition of lp-pla2 activity
NZ551180A (en) 2004-06-01 2009-10-30 Genentech Inc Antibody drug conjugates and methods
JO3000B1 (en) 2004-10-20 2016-09-05 Genentech Inc Antibody Formulations.
US7964195B2 (en) 2005-01-07 2011-06-21 Diadexus, Inc. Ovr110 antibody compositions and methods of use
EP3698807A1 (en) 2005-01-21 2020-08-26 Genentech, Inc. Fixed dosing of her antibodies
CA2595395A1 (en) * 2005-02-09 2006-08-17 Genentech, Inc. Inhibiting her2 shedding with matrix metalloprotease antagonists
AU2006216732C1 (en) * 2005-02-23 2017-07-20 Genentech, Inc. Extending time to disease progression or survival in cancer patients using a HER dimerization inhibitor
WO2006096861A2 (en) * 2005-03-08 2006-09-14 Genentech, Inc. METHODS FOR IDENTIFYING TUMORS RESPONSIVE TO TREATMENT WITH HER DIMERIZATION INHIBITORS (HDIs)
LT2439273T (en) 2005-05-09 2019-05-10 Ono Pharmaceutical Co., Ltd. Human monoclonal antibodies to programmed death 1(PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
JP2006316040A (en) 2005-05-13 2006-11-24 Genentech Inc Herceptin(r) adjuvant treatment
US7741053B2 (en) * 2005-05-13 2010-06-22 Sigma-Aldrich Co. Processes for purification of recombinant proteins
US7858843B2 (en) 2005-06-06 2010-12-28 Genentech, Inc. Gene disruptions, compositions and methods relating thereto
CN105330741B (en) 2005-07-01 2023-01-31 E.R.施贵宝&圣斯有限责任公司 Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
EP1922412A4 (en) * 2005-07-15 2009-07-15 Siemens Healthcare Diagnostics Humanized antibody conjugates and related methods, assays, reagents, and kits
DK1912675T3 (en) 2005-07-25 2014-03-24 Emergent Product Dev Seattle B-cell reduction using specific and cd37-cd20-specific binding molecules
AU2006280321A1 (en) 2005-08-15 2007-02-22 Genentech, Inc. Gene disruptions, compositions and methods relating thereto
US8906864B2 (en) 2005-09-30 2014-12-09 AbbVie Deutschland GmbH & Co. KG Binding domains of proteins of the repulsive guidance molecule (RGM) protein family and functional fragments thereof, and their use
AU2006335053A1 (en) 2005-11-21 2007-07-19 Genentech, Inc. Novel gene disruptions, compositions and methods relating thereto
US8052971B2 (en) * 2005-11-21 2011-11-08 MG Biologics Oral use of specific antibodies for intestinal health
TW200730539A (en) * 2005-12-01 2007-08-16 Domantis Ltd Noncompetitive domain antibody formats that bind interleukin 1 receptor type 1
CA2638821A1 (en) 2006-02-17 2007-10-11 Genentech, Inc. Gene disruptons, compositions and methods relating thereto
EP1996716B1 (en) * 2006-03-20 2011-05-11 The Regents of the University of California Engineered anti-prostate stem cell antigen (psca) antibodies for cancer targeting
EP2614839A3 (en) 2006-04-05 2015-01-28 Genentech, Inc. Method for using BOC/CDO to modulate hedgehog signaling
US9044461B2 (en) 2006-04-07 2015-06-02 The Research Foundation Of State University Of New York Transcobalamin receptor polypeptides, nucleic acids, and modulators thereof, and related methods of use in modulating cell growth and treating cancer and cobalamin deficiency
EP2010226B1 (en) * 2006-04-07 2014-01-15 The Research Foundation of State University of New York Transcobalamin receptor polypeptides, nucleic acids, and modulators thereof, and related methods of use in modulating cell growth and treating cancer and cobalamin deficiency
EP2082645A1 (en) 2006-04-19 2009-07-29 Genentech, Inc. Novel gene disruptions, compositions and methods relating thereto
JP2009539413A (en) * 2006-06-12 2009-11-19 トゥルビオン・ファーマシューティカルズ・インコーポレーテッド Single-chain multivalent binding protein with effector function
US8586006B2 (en) 2006-08-09 2013-11-19 Institute For Systems Biology Organ-specific proteins and methods of their use
MX2009005189A (en) 2006-11-15 2009-06-30 Medarex Inc Human monoclonal antibodies to btla and methods of use.
JP5391073B2 (en) 2006-11-27 2014-01-15 ディアデクサス インコーポレーテッド Ovr110 antibody compositions and methods of use
WO2008109440A2 (en) 2007-03-02 2008-09-12 Genentech, Inc. Predicting response to a her dimerisation inhibitor based on low her3 expression
US7960139B2 (en) 2007-03-23 2011-06-14 Academia Sinica Alkynyl sugar analogs for the labeling and visualization of glycoconjugates in cells
KR20100040840A (en) * 2007-06-06 2010-04-21 도만티스 리미티드 Polypeptides, antibody variable domains and antagonists
US9551033B2 (en) * 2007-06-08 2017-01-24 Genentech, Inc. Gene expression markers of tumor resistance to HER2 inhibitor treatment
WO2008154249A2 (en) * 2007-06-08 2008-12-18 Genentech, Inc. Gene expression markers of tumor resistance to her2 inhibitor treatment
CN101990439A (en) * 2007-07-06 2011-03-23 特鲁比昂药品公司 Binding peptides having a c-terminally disposed specific binding domain
JP5469600B2 (en) 2007-07-16 2014-04-16 ジェネンテック, インコーポレイテッド Anti-CD79b antibody and immunoconjugate and method of use thereof
TW200918089A (en) 2007-07-16 2009-05-01 Genentech Inc Humanized anti-CD79b antibodies and immunoconjugates and methods of use
US20090304719A1 (en) 2007-08-22 2009-12-10 Patrick Daugherty Activatable binding polypeptides and methods of identification and use thereof
JP6126773B2 (en) 2007-09-04 2017-05-10 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア High affinity anti-prostatic stem cell antigen (PSCA) antibody for cancer targeting and detection
EP2209498A2 (en) * 2007-10-03 2010-07-28 Cornell University Treatment of proliferative disorders using antibodies to psma
NZ585556A (en) 2007-11-07 2012-07-27 Celldex Therapeutics Inc Antibodies that bind human dendritic and epithelial cell 205 (dec-205)
KR20100097691A (en) 2007-11-12 2010-09-03 테라클론 사이언시스, 아이엔씨. Compositions and methods for the therapy and diagnosis of influenza
TWI472339B (en) 2008-01-30 2015-02-11 Genentech Inc Composition comprising antibody that binds to domain ii of her2 and acidic variants thereof
SI2657253T1 (en) 2008-01-31 2017-10-30 Genentech, Inc. Anti-CD79b antibodies and immunoconjugates and methods of use
US8962803B2 (en) 2008-02-29 2015-02-24 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM A protein and uses thereof
AU2009223688B2 (en) 2008-03-10 2014-12-11 Theraclone Sciences, Inc. Compositions and methods for the therapy and diagnosis of cytomegalovirus infections
ES2368700T3 (en) 2008-04-11 2011-11-21 Emergent Product Development Seattle, Llc IMMUNOTHERAPEUTIC AGENT FOR CD37 AND COMBINATION WITH A BIFUNCTIONAL CHEMOTHERAPEUTIC AGENT OF THE SAME.
BRPI0812682A2 (en) 2008-06-16 2010-06-22 Genentech Inc metastatic breast cancer treatment
JP5986745B2 (en) 2008-07-15 2016-09-06 アカデミア シニカAcademia Sinica Glycan arrays on PTFE-like aluminum-coated glass slides and related methods
US20100069616A1 (en) * 2008-08-06 2010-03-18 The Regents Of The University Of California Engineered antibody-nanoparticle conjugates
AR072999A1 (en) 2008-08-11 2010-10-06 Medarex Inc HUMAN ANTIBODIES THAT JOIN GEN 3 OF LYMPHOCYTARY ACTIVATION (LAG-3) AND THE USES OF THESE
EP2727606A3 (en) 2008-09-08 2015-09-23 Psma Development Company, L.L.C. Compounds for killing psma-expressing, taxane-resistant cancer cells
WO2010062857A1 (en) 2008-11-26 2010-06-03 Allergan, Inc. Klk-13 antibody inhibitor for treating dry eye
RU2636046C2 (en) 2009-01-12 2017-11-17 Сайтомкс Терапьютикс, Инк Modified antibodies composition, methods of production and application
WO2010096486A1 (en) * 2009-02-17 2010-08-26 Cornell Research Foundation, Inc. Methods and kits for diagnosis of cancer and prediction of therapeutic value
BRPI1011384A2 (en) 2009-02-23 2016-03-15 Cytomx Therapeutics Inc proproteins and their methods of use
SI3260136T1 (en) 2009-03-17 2021-05-31 Theraclone Sciences, Inc. Human immunodeficiency virus (hiv) -neutralizing antibodies
ES2572728T3 (en) 2009-03-20 2016-06-02 F. Hoffmann-La Roche Ag Bispecific anti-HER antibodies
AU2010236787A1 (en) 2009-04-01 2011-11-10 Genentech, Inc. Anti-FcRH5 antibodies and immunoconjugates and methods of use
CA2759506A1 (en) 2009-04-23 2010-10-28 Theraclone Sciences, Inc. Granulocyte-macrophage colony-stimulating factor (gm-csf) neutralizing antibodies
AU2010249787A1 (en) 2009-05-20 2011-12-22 Theraclone Sciences, Inc. Compositions and methods for the therapy and diagnosis of influenza
JP5705836B2 (en) 2009-05-29 2015-04-22 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト Modulators for HER2 signaling in gastric cancer patients expressing HER2
EP2448966B1 (en) 2009-07-03 2018-11-14 Avipep Pty Ltd Immuno-conjugates and methods for producing them
US20110038871A1 (en) 2009-08-11 2011-02-17 Veena Viswanth Ccr2 inhibitors for treating conditions of the eye
BR112012009409A2 (en) 2009-10-22 2017-02-21 Genentech Inc method of identifying an inhibitory substance, antagonist molecule, isolated nucleic acid, vector, host cell, method of making the molecule, composition, article of manufacture, method of inhibiting a biological activity, method of treating a pathological condition, method for detect msp in a sample and method to detect hepsin in a sample
AR079217A1 (en) 2009-11-30 2012-01-04 Genentech Inc COMPOSITIONS AND METHODS FOR DIAGNOSIS AND TUMOR TREATMENT
US11377485B2 (en) 2009-12-02 2022-07-05 Academia Sinica Methods for modifying human antibodies by glycan engineering
US10087236B2 (en) 2009-12-02 2018-10-02 Academia Sinica Methods for modifying human antibodies by glycan engineering
CA2782333C (en) 2009-12-02 2019-06-04 Imaginab, Inc. J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (psma) and methods for their use
EP2507381A4 (en) 2009-12-04 2016-07-20 Hoffmann La Roche Multispecific antibodies, antibody analogs, compositions, and methods
SG181563A1 (en) 2009-12-08 2012-07-30 Abbott Gmbh & Co Kg Monoclonal antibodies against the rgm a protein for use in the treatment of retinal nerve fiber layer degeneration
CA2784610C (en) 2009-12-23 2020-07-14 Avipep Pty Ltd Immuno-conjugates and methods for producing them
US8900588B2 (en) * 2010-01-08 2014-12-02 Les Laboratories Servier Methods for treating breast cancer
JP5981853B2 (en) 2010-02-18 2016-08-31 ジェネンテック, インコーポレイテッド Neuregulin antagonists and their use in the treatment of cancer
MX2012009215A (en) 2010-02-23 2012-11-23 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor.
US9631018B2 (en) 2010-03-26 2017-04-25 The Trustees Of Dartmouth College Vista regulatory T cell mediator protein, vista binding agents and use thereof
US20150231215A1 (en) 2012-06-22 2015-08-20 Randolph J. Noelle VISTA Antagonist and Methods of Use
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
WO2011130332A1 (en) 2010-04-12 2011-10-20 Academia Sinica Glycan arrays for high throughput screening of viruses
EP2566510A1 (en) 2010-05-03 2013-03-13 F. Hoffmann-La Roche AG Compositions and methods useful for reducing the viscosity of protein-containing formulations
MA34291B1 (en) 2010-05-03 2013-06-01 Genentech Inc COMPOSITIONS AND METHODS FOR DIAGNOSING AND TREATING A TUMOR
CA2835489C (en) 2010-05-10 2018-03-06 Chi-Huey Wong Zanamivir phosphonate congeners with anti-influenza activity and determining oseltamivir susceptibility of influenza viruses
WO2011146568A1 (en) 2010-05-19 2011-11-24 Genentech, Inc. Predicting response to a her inhibitor
PT2580240T (en) 2010-06-14 2019-03-29 Lykera Biomed S A S100a4 antibodies and therapeutic uses thereof
US20110311527A1 (en) 2010-06-16 2011-12-22 Allergan, Inc. IL23p19 ANTIBODY INHIBITOR FOR TREATING OCULAR AND OTHER CONDITIONS
JP2013540701A (en) 2010-08-12 2013-11-07 セラクローン サイエンシーズ, インコーポレイテッド Anti-hemagglutinin antibody composition and method of use thereof
PT3556396T (en) 2010-08-31 2022-07-04 Scripps Research Inst Human immunodeficiency virus (hiv)-neutralizing antibodies
TW201302793A (en) 2010-09-03 2013-01-16 Glaxo Group Ltd Novel antigen binding proteins
US20130224116A1 (en) 2010-11-05 2013-08-29 TransBio Ltd. Markers of Endothelial Progenitor Cells and Uses Thereof
EP2643353A1 (en) 2010-11-24 2013-10-02 Novartis AG Multispecific molecules
EP2465536A1 (en) * 2010-12-14 2012-06-20 CSL Behring AG CD89 activation in therapy
SG191153A1 (en) 2010-12-23 2013-07-31 Hoffmann La Roche Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
AU2012217867A1 (en) 2011-02-14 2013-09-05 Theraclone Sciences, Inc. Compositions and methods for the therapy and diagnosis of influenza
WO2012119989A2 (en) 2011-03-04 2012-09-13 Oryzon Genomics, S.A. Methods and antibodies for the diagnosis and treatment of cancer
EP2685968A1 (en) 2011-03-15 2014-01-22 Theraclone Sciences, Inc. Compositions and methods for the therapy and diagnosis of influenza
EP2699598B1 (en) 2011-04-19 2019-03-06 Pfizer Inc Combinations of anti-4-1bb antibodies and adcc-inducing antibodies for the treatment of cancer
WO2012142662A1 (en) 2011-04-21 2012-10-26 Garvan Institute Of Medical Research Modified variable domain molecules and methods for producing and using them b
WO2012158948A1 (en) 2011-05-17 2012-11-22 The Rockefeller University Human immunodeficiency virus neutralizing antibodies adn methods of use thereof
WO2013015821A1 (en) 2011-07-22 2013-01-31 The Research Foundation Of State University Of New York Antibodies to the b12-transcobalamin receptor
US9120858B2 (en) 2011-07-22 2015-09-01 The Research Foundation Of State University Of New York Antibodies to the B12-transcobalamin receptor
MX2014001766A (en) 2011-08-17 2014-05-01 Genentech Inc Neuregulin antibodies and uses thereof.
US8822651B2 (en) 2011-08-30 2014-09-02 Theraclone Sciences, Inc. Human rhinovirus (HRV) antibodies
WO2013063229A1 (en) 2011-10-25 2013-05-02 The Regents Of The University Of Michigan Her2 targeting agent treatment in non-her2-amplified cancers having her2 expressing cancer stem cells
CA2857114A1 (en) 2011-11-30 2013-06-06 Genentech, Inc. Erbb3 mutations in cancer
WO2013083810A1 (en) 2011-12-09 2013-06-13 F. Hoffmann-La Roche Ag Identification of non-responders to her2 inhibitors
CA2855570A1 (en) 2011-12-14 2013-06-20 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
US10118958B2 (en) 2011-12-14 2018-11-06 AbbVie Deutschland GmbH & Co. KG Composition and method for the diagnosis and treatment of iron-related disorders
MY176695A (en) 2012-01-27 2020-08-19 Abbvie Inc Composition and method for the diagnosis and treatment of diseases associated with neurite degeneration
RU2627185C1 (en) * 2012-03-16 2017-08-03 Коваген Аг New binding molecules with anti-tumour activity
JP2015514710A (en) 2012-03-27 2015-05-21 ジェネンテック, インコーポレイテッド Diagnosis and treatment of HER3 inhibitors
US10130714B2 (en) 2012-04-14 2018-11-20 Academia Sinica Enhanced anti-influenza agents conjugated with anti-inflammatory activity
WO2013169693A1 (en) 2012-05-09 2013-11-14 Bristol-Myers Squibb Company Methods of treating cancer using an il-21 polypeptide and an anti-pd-1 antibody
JP6267689B2 (en) 2012-05-10 2018-01-24 バイオアトラ、エルエルシー Multispecific monoclonal antibody
WO2014039983A1 (en) 2012-09-07 2014-03-13 The Trustees Of Dartmouth College Vista modulators for diagnosis and treatment of cancer
US9890215B2 (en) 2012-06-22 2018-02-13 King's College London Vista modulators for diagnosis and treatment of cancer
AR091649A1 (en) 2012-07-02 2015-02-18 Bristol Myers Squibb Co OPTIMIZATION OF ANTIBODIES THAT FIX THE LYMPHOCYTE ACTIVATION GEN 3 (LAG-3) AND ITS USES
AU2013306098A1 (en) 2012-08-18 2015-02-12 Academia Sinica Cell-permeable probes for identification and imaging of sialidases
WO2014031762A1 (en) 2012-08-21 2014-02-27 Academia Sinica Benzocyclooctyne compounds and uses thereof
PL2908912T3 (en) 2012-10-18 2021-05-17 The Rockefeller University Broadly-neutralizing anti-hiv antibodies
MX363188B (en) 2012-11-30 2019-03-13 Hoffmann La Roche Identification of patients in need of pd-l1 inhibitor cotherapy.
US9469686B2 (en) 2013-03-15 2016-10-18 Abbott Laboratories Anti-GP73 monoclonal antibodies and methods of obtaining the same
US20140283157A1 (en) 2013-03-15 2014-09-18 Diadexus, Inc. Lipoprotein-associated phospholipase a2 antibody compositions and methods of use
US9308236B2 (en) 2013-03-15 2016-04-12 Bristol-Myers Squibb Company Macrocyclic inhibitors of the PD-1/PD-L1 and CD80(B7-1)/PD-L1 protein/protein interactions
ES2753419T3 (en) 2013-06-07 2020-04-08 Univ Duke Complement factor H inhibitors
US10086054B2 (en) 2013-06-26 2018-10-02 Academia Sinica RM2 antigens and use thereof
EP3013347B1 (en) 2013-06-27 2019-12-11 Academia Sinica Glycan conjugates and use thereof
CN105682666B (en) 2013-09-06 2021-06-01 中央研究院 Activation of human iNKT cells using glycolipids
TWI728373B (en) 2013-12-23 2021-05-21 美商建南德克公司 Antibodies and methods of use
PL3712174T3 (en) 2013-12-24 2022-07-04 Janssen Pharmaceutica Nv Anti-vista antibodies and fragments
US11014987B2 (en) 2013-12-24 2021-05-25 Janssen Pharmaceutics Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
WO2015108719A1 (en) 2014-01-14 2015-07-23 The Medical College Of Wisconsin, Inc. Targeting clptm1l for treatment and prevention of cancer
US10150818B2 (en) 2014-01-16 2018-12-11 Academia Sinica Compositions and methods for treatment and detection of cancers
US9982041B2 (en) 2014-01-16 2018-05-29 Academia Sinica Compositions and methods for treatment and detection of cancers
TWI687428B (en) 2014-03-27 2020-03-11 中央研究院 Reactive labelling compounds and uses thereof
US9850225B2 (en) 2014-04-14 2017-12-26 Bristol-Myers Squibb Company Compounds useful as immunomodulators
CN106573971A (en) 2014-05-27 2017-04-19 中央研究院 Anti-CD20 glycoantibodies and uses thereof
EP3149161B1 (en) 2014-05-27 2021-07-28 Academia Sinica Fucosidase from bacteroides and methods using the same
AU2015267045B2 (en) 2014-05-27 2021-02-25 Academia Sinica Anti-HER2 glycoantibodies and uses thereof
US10118969B2 (en) 2014-05-27 2018-11-06 Academia Sinica Compositions and methods relating to universal glycoforms for enhanced antibody efficacy
TWI732738B (en) 2014-05-28 2021-07-11 中央研究院 Anti-tnf-alpha glycoantibodies and uses thereof
CN113549153A (en) 2014-05-29 2021-10-26 宏观基因有限公司 Trispecific binding molecules and methods of use thereof
EA037006B1 (en) 2014-06-06 2021-01-26 Бристол-Майерс Сквибб Компани Antibodies against glucocorticoid-induced tumor necrosis factor receptor (gitr) and uses thereof
MX2016016310A (en) 2014-06-11 2017-10-20 A Green Kathy Use of vista agonists and antagonists to suppress or enhance humoral immunity.
TWI693232B (en) 2014-06-26 2020-05-11 美商宏觀基因股份有限公司 Covalently bonded diabodies having immunoreactivity with pd-1 and lag-3, and methods of use thereof
WO2016040369A2 (en) 2014-09-08 2016-03-17 Academia Sinica HUMAN iNKT CELL ACTIVATION USING GLYCOLIPIDS
NZ730348A (en) 2014-09-26 2024-03-22 Macrogenics Inc Bi-specific monovalent diabodies that are capable of binding cd19 and cd3, and uses thereof
US9732119B2 (en) 2014-10-10 2017-08-15 Bristol-Myers Squibb Company Immunomodulators
CA2961439A1 (en) 2014-11-05 2016-05-12 Genentech, Inc. Anti-fgfr2/3 antibodies and methods using same
US9856292B2 (en) 2014-11-14 2018-01-02 Bristol-Myers Squibb Company Immunomodulators
RS60631B1 (en) 2014-11-21 2020-09-30 Bristol Myers Squibb Co Antibodies against cd73 and uses thereof
EP3227341A1 (en) 2014-12-02 2017-10-11 CeMM - Forschungszentrum für Molekulare Medizin GmbH Anti-mutant calreticulin antibodies and their use in the diagnosis and therapy of myeloid malignancies
EP3226900A4 (en) 2014-12-05 2018-09-19 Immunext, Inc. Identification of vsig8 as the putative vista receptor and its use thereof to produce vista/vsig8 modulators
US9861680B2 (en) 2014-12-18 2018-01-09 Bristol-Myers Squibb Company Immunomodulators
US9944678B2 (en) 2014-12-19 2018-04-17 Bristol-Myers Squibb Company Immunomodulators
TWI708786B (en) 2014-12-23 2020-11-01 美商必治妥美雅史谷比公司 Antibodies to tigit
PT3240801T (en) 2014-12-31 2021-02-18 Checkmate Pharmaceuticals Inc Combination tumor immunotherapy
US9975965B2 (en) 2015-01-16 2018-05-22 Academia Sinica Compositions and methods for treatment and detection of cancers
US10495645B2 (en) 2015-01-16 2019-12-03 Academia Sinica Cancer markers and methods of use thereof
EP3248005B1 (en) 2015-01-24 2020-12-09 Academia Sinica Novel glycan conjugates and methods of use thereof
AU2015380455A1 (en) 2015-01-26 2017-08-03 Macrogenics, Inc. Multivalent molecules comprising DR5-binding domains
US20160222060A1 (en) 2015-02-04 2016-08-04 Bristol-Myers Squibb Company Immunomodulators
CN107580500B (en) 2015-02-19 2023-05-30 康姆普根有限公司 anti-PVRIG antibodies and methods of use
US9809625B2 (en) 2015-03-18 2017-11-07 Bristol-Myers Squibb Company Immunomodulators
CA2987410A1 (en) 2015-05-29 2016-12-08 Bristol-Myers Squibb Company Antibodies against ox40 and uses thereof
TWI773646B (en) 2015-06-08 2022-08-11 美商宏觀基因股份有限公司 Lag-3-binding molecules and methods of use thereof
JP7026509B2 (en) 2015-06-24 2022-02-28 ヤンセン ファーマシューティカ エヌブイ Anti-VISTA antibody and fragment
AU2016285920A1 (en) 2015-06-29 2018-02-01 Bristol-Myers Squibb Company Antibodies to CD40 with enhanced agonist activity
KR20180034588A (en) 2015-07-30 2018-04-04 마크로제닉스, 인크. PD-1-binding molecules and methods for their use
NZ739721A (en) 2015-08-07 2019-09-27 Imaginab Inc Antigen binding constructs to target molecules
EP3352760A4 (en) 2015-09-21 2019-03-06 Aptevo Research and Development LLC Cd3 binding polypeptides
RU2638457C2 (en) 2015-09-28 2017-12-13 Общество С Ограниченной Ответственностью "Онкомакс" Antibodies specifically binding type 1 receptor of fibroblast growth factor, antibodies application for oncological disease treatment, method for antibodies production
WO2017062619A2 (en) 2015-10-08 2017-04-13 Macrogenics, Inc. Combination therapy for the treatment of cancer
US10745382B2 (en) 2015-10-15 2020-08-18 Bristol-Myers Squibb Company Compounds useful as immunomodulators
JP6998869B2 (en) 2015-11-08 2022-02-04 ジェネンテック, インコーポレイテッド Screening method for multispecific antibody
AU2016356780A1 (en) 2015-11-19 2018-06-28 Bristol-Myers Squibb Company Antibodies against glucocorticoid-induced tumor necrosis factor receptor (GITR) and uses thereof
CN106729743B (en) 2015-11-23 2021-09-21 四川科伦博泰生物医药股份有限公司 anti-ErbB 2 antibody-drug conjugate, and composition, preparation method and application thereof
CR20180318A (en) 2015-12-14 2018-09-19 Macrogenics Inc BISPECIFIC MOLECULES THAT HAVE IMMUNORREACTIVITY WITH PD-1 AND CTLA-4, AND METHODS OF USE OF THE SAME
CN116920085A (en) 2016-02-12 2023-10-24 詹森药业有限公司 anti-VISTA (B7H 5) antibodies
UY37127A (en) 2016-02-17 2017-08-31 Macrogenics Inc MOLECULES OF UNION TO ROR1, AND METHODS OF USE OF THE SAME
US10143746B2 (en) 2016-03-04 2018-12-04 Bristol-Myers Squibb Company Immunomodulators
EA201891983A8 (en) 2016-03-04 2020-05-28 Бристол-Майерс Сквибб Компани COMBINED THERAPY BY ANTIBODIES TO CD73
SG11201807677YA (en) 2016-03-04 2018-10-30 Univ Rockefeller Antibodies to cd40 with enhanced agonist activity
TW201808978A (en) 2016-03-08 2018-03-16 中央研究院 Methods for modular synthesis of N-glycans and arrays thereof
WO2017165681A1 (en) 2016-03-24 2017-09-28 Gensun Biopharma Inc. Trispecific inhibitors for cancer treatment
US10358463B2 (en) 2016-04-05 2019-07-23 Bristol-Myers Squibb Company Immunomodulators
RU2021111187A (en) 2016-04-15 2021-04-29 Янссен Фармасьютикалз, Инк. ANTIBODIES AGAINST HUMAN VISTA AND THEIR APPLICATION
MY198114A (en) 2016-04-15 2023-08-04 Macrogenics Inc Novel b7-h3-binding molecules, antibody drug conjugates thereof and methods of use thereof
MA44723A (en) 2016-04-18 2019-02-27 Celldex Therapeutics Inc HUMAN CD40 BINDING AGONIST ANTIBODIES AND THEIR USES
EP3454863A1 (en) 2016-05-10 2019-03-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Combinations therapies for the treatment of cancer
CA3029991A1 (en) 2016-07-08 2018-01-11 Bristol-Myers Squibb Company 1,3-dihydroxy-phenyl derivatives useful as immunomodulators
JP7027401B2 (en) 2016-07-14 2022-03-01 ブリストル-マイヤーズ スクイブ カンパニー Antibodies to TIM3 and its use
JP7213549B2 (en) 2016-08-22 2023-01-27 シーエイチオー ファーマ インコーポレイテッド Antibodies, Binding Fragments, and Methods of Use
US10144706B2 (en) 2016-09-01 2018-12-04 Bristol-Myers Squibb Company Compounds useful as immunomodulators
JP7083819B2 (en) * 2016-09-30 2022-06-13 セントレ ナショナル デ ラ レセルシュ シャンティフィク Cell marker
EP3519824A1 (en) 2016-10-03 2019-08-07 Abbott Laboratories Improved methods of assessing uch-l1 status in patient samples
CN110267971B (en) 2016-11-07 2023-12-19 百时美施贵宝公司 Immunomodulators
JP7106572B2 (en) 2016-12-20 2022-07-26 ブリストル-マイヤーズ スクイブ カンパニー Compounds Useful as Immunomodulators
EP3558368A4 (en) 2016-12-23 2020-12-30 MacroGenics, Inc. Adam9-binding molecules, and methods of use thereof
EP3565839A4 (en) 2017-01-05 2021-04-21 Gensun Biopharma Inc. Checkpoint regulator antagonists
EP3565845A4 (en) 2017-01-06 2020-10-07 Biosion, Inc. Erbb2 antibodies and uses therefore
US11377497B2 (en) 2017-01-23 2022-07-05 Suzhou Alphamab Co., Ltd. PD-L1 binding polypeptide or composite
WO2018147960A1 (en) 2017-02-08 2018-08-16 Imaginab, Inc. Extension sequences for diabodies
WO2018152496A1 (en) 2017-02-17 2018-08-23 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Compositions and methods for the diagnosis and treatment of zika virus infection
US11459394B2 (en) 2017-02-24 2022-10-04 Macrogenics, Inc. Bispecific binding molecules that are capable of binding CD137 and tumor antigens, and uses thereof
US11016092B2 (en) 2017-03-23 2021-05-25 Abbott Laboratories Methods for aiding in the diagnosis and determination of the extent of traumatic brain injury in a human subject using the early biomarker ubiquitin carboxy-terminal hydrolase L1
ES2961550T3 (en) 2017-03-27 2024-03-12 Bristol Myers Squibb Co Substituted isoquinoline derivatives as immunomodulators
TWI788340B (en) 2017-04-07 2023-01-01 美商必治妥美雅史谷比公司 Anti-icos agonist antibodies and uses thereof
US10877048B2 (en) 2017-04-15 2020-12-29 Abbott Laboratories Methods for aiding in the hyperacute diagnosis and determination of traumatic brain injury in a human subject using early biomarkers
EP3615569A1 (en) 2017-04-25 2020-03-04 The U.S.A. As Represented By The Secretary, Department Of Health And Human Services Antibodies and methods for the diagnosis and treatment of epstein barr virus infection
US10877038B2 (en) 2017-04-28 2020-12-29 Abbott Laboratories Methods for aiding in the hyperacute diagnosis and determination of traumatic brain injury using early biomarkers on at least two samples from the same human subject
US10865238B1 (en) 2017-05-05 2020-12-15 Duke University Complement factor H antibodies
AU2018272054A1 (en) 2017-05-25 2019-09-26 Abbott Laboratories Methods for aiding in the determination of whether to perform imaging on a human subject who has sustained or may have sustained an injury to the head using early biomarkers
JP7269183B2 (en) 2017-05-30 2023-05-08 アボット・ラボラトリーズ Methods for Aiding in Diagnosing and Assessing Mild Traumatic Brain Injury in Human Subjects Using Cardiac Troponin I
KR20200020858A (en) 2017-06-23 2020-02-26 브리스톨-마이어스 스큅 컴퍼니 Immunomodulators Acting as Antagonists of PD-1
WO2019010131A1 (en) 2017-07-03 2019-01-10 Abbott Laboratories Improved methods for measuring ubiquitin carboxy-terminal hydrolase l1 levels in blood
CN111094334A (en) 2017-07-19 2020-05-01 美国卫生与公众服务部 Antibodies and methods for diagnosis and treatment of hepatitis B virus infection
US11492375B2 (en) 2017-10-03 2022-11-08 Bristol-Myers Squibb Company Cyclic peptide immunomodulators
WO2019089921A1 (en) 2017-11-01 2019-05-09 Bristol-Myers Squibb Company Immunostimulatory agonistic antibodies for use in treating cancer
BR112020010085A2 (en) 2017-12-09 2020-10-13 Abbott Laboratories methods to assist in the diagnosis and assessment of a traumatic brain injury in a human subject using a combination of gfap and uch-l1
US11022617B2 (en) 2017-12-09 2021-06-01 Abbott Laboratories Methods for aiding in the diagnosis and evaluation of a subject who has sustained an orthopedic injury and that has or may have sustained an injury to the head, such as mild traumatic brain injury (TBI), using glial fibrillary acidic protein (GFAP) and/or ubiquitin carboxy-terminal hydrolase L1 (UCH-L1)
RU2020122822A (en) 2017-12-12 2022-01-13 Макродженикс, Инк. BISPECIFIC CD16 BINDING MOLECULES AND THEIR USE IN THE TREATMENT OF DISEASES
CR20200313A (en) 2017-12-19 2020-12-01 Univ Rockefeller HUMAN IgG Fc DOMAIN VARIANTS WITH IMPROVED EFFECTOR FUNCTION
EP3732198A1 (en) 2017-12-27 2020-11-04 Bristol-Myers Squibb Company Anti-cd40 antibodies and uses thereof
JP7358361B2 (en) 2018-01-12 2023-10-10 ブリストル-マイヤーズ スクイブ カンパニー Antibodies against TIM3 and their uses
US11414418B2 (en) 2018-01-23 2022-08-16 Bristol-Myers Squibb Company Compounds useful as immunomodulators
US11685781B2 (en) 2018-02-15 2023-06-27 Macrogenics, Inc. Variant CD3-binding domains and their use in combination therapies for the treatment of disease
WO2019169123A1 (en) 2018-03-01 2019-09-06 Bristol-Myers Squibb Company Compounds useful as immunomodulators
TW202003565A (en) 2018-03-23 2020-01-16 美商必治妥美雅史谷比公司 Antibodies against MICA and/or MICB and uses thereof
EA202092316A1 (en) 2018-03-28 2021-05-25 Бристол-Маерс Сквибб Компани FUSION PROTEINS OF INTERLEUKIN-2 / ALPHA-RECEPTOR OF INTERLEUKIN-2 AND METHODS OF APPLICATION
CA3096674A1 (en) 2018-04-12 2019-10-17 Bristol-Myers Squibb Company Anticancer combination therapy with cd73 antagonist antibody and pd-1/pd-l1 axis antagonist antibody
WO2019213416A1 (en) 2018-05-02 2019-11-07 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Antibodies and methods for the diagnosis, prevention, and treatment of epstein barr virus infection
EP3569618A1 (en) 2018-05-19 2019-11-20 Boehringer Ingelheim International GmbH Antagonizing cd73 antibody
WO2020006511A1 (en) 2018-06-29 2020-01-02 Gensun Biopharma, Inc. Trispecific antagonists
US20200109200A1 (en) 2018-10-09 2020-04-09 Genentech, Inc. Methods and systems for determining synapse formation
PE20211284A1 (en) 2018-11-16 2021-07-19 Bristol Myers Squibb Co ANTI-NKG2A ANTIBODIES AND USES OF THEM
WO2020115223A1 (en) 2018-12-05 2020-06-11 Katholieke Universiteit Leuven S100a4 as a marker of treatment with spironolactone, pioglitazone and metformin
CN109929037B (en) 2019-04-01 2023-03-17 华博生物医药技术(上海)有限公司 Conjugates to programmed death ligands and uses thereof
US10851157B2 (en) 2019-07-01 2020-12-01 Gensun Biopharma, Inc. Antagonists targeting the TGF-β pathway
JP2022541652A (en) 2019-07-26 2022-09-26 ヴァンダービルト ユニバーシティ Human monoclonal antibody against enterovirus D68
US20240124417A1 (en) 2019-10-04 2024-04-18 Bristol-Myers Squibb Company Compounds useful as immunomodulators
IL295310A (en) 2020-02-11 2022-10-01 Univ Vanderbilt Human monoclonal antibodies to severe acute respiratory syndrome coronavirus 2 (sars-cov- 2)
US20230140384A1 (en) 2020-03-09 2023-05-04 Bristol-Myers Squibb Company Antibodies to cd40 with enhanced agonist activity
WO2021195385A1 (en) 2020-03-26 2021-09-30 Vanderbilt University HUMAN MONOCLONAL ANTIBODIES TO SEVERE ACUTE RESPIRATORY SYNDROME CORONAVIRUS 2 (SARS-GoV-2)
PL4045533T3 (en) 2020-03-26 2024-04-15 Vanderbilt University Human monoclonal antibodies to severe acute respiratory syndrome coronavirus 2 (sars-cov-2)
WO2021211331A1 (en) 2020-04-13 2021-10-21 Abbott Point Of Care Inc. METHODS, COMPLEXES AND KITS FOR DETECTING OR DETERMINING AN AMOUNT OF A ß-CORONAVIRUS ANTIBODY IN A SAMPLE
WO2021231732A1 (en) 2020-05-15 2021-11-18 Bristol-Myers Squibb Company Antibodies to garp
EP4175650A1 (en) 2020-07-06 2023-05-10 Kiromic BioPharma, Inc. Mesothelin isoform binding molecules and chimeric pd1 receptor molecules, cells containing the same and uses thereof
US20220043000A1 (en) 2020-08-04 2022-02-10 Abbott Laboratories Methods and kits for detecting sars-cov-2 protein in a sample
US20220043003A1 (en) 2020-08-04 2022-02-10 Abbott Rapid Diagnostics International Unlimited Company Assays for detecting sars-cov-2
DE102020125457A1 (en) 2020-09-29 2022-03-31 Immatics Biotechnologies Gmbh Amidated peptides and their deamidated counterparts presented by HLA-A*02 molecules for use in immunotherapy against various types of cancer
TW202229312A (en) 2020-09-29 2022-08-01 德商英麥提克生物技術股份有限公司 Amidated peptides and their deamidated counterparts displayed by non-hla-a*02 for use in immunotherapy against different types of cancers
DE102020125465A1 (en) 2020-09-29 2022-03-31 Immatics Biotechnologies Gmbh Amidated peptides and their deamidated counterparts presented by non-HLA-A*02 molecules for use in immunotherapy against various types of cancer
US20230399407A1 (en) * 2020-11-06 2023-12-14 Korea University Research And Business Foundation Fc alpha receptor binding antibody
WO2022108627A1 (en) 2020-11-18 2022-05-27 Kiromic Biopharma, Inc.Kiromic Biopharma, Inc. Gamma-delta t cell manufacturing processes and chimeric pd1 receptor molecules
WO2023102384A1 (en) 2021-11-30 2023-06-08 Abbott Laboratories Use of one or more biomarkers to determine traumatic brain injury (tbi) in a subject having received a head computerized tomography scan that is negative for a tbi
CA3198161A1 (en) 2020-12-01 2022-06-09 Beth MCQUISTON Use of one or more biomarkers to determine traumatic brain injury (tbi) in a subject having received a head computerized tomography scan that is negative for a tbi
WO2022147147A1 (en) 2020-12-30 2022-07-07 Abbott Laboratories Methods for determining sars-cov-2 antigen and anti-sars-cov-2 antibody in a sample
TW202241925A (en) 2021-01-15 2022-11-01 德商英麥提克生物技術股份有限公司 Peptides displayed by hla for use in immunotherapy against different types of cancers
EP4314068A1 (en) 2021-04-02 2024-02-07 The Regents Of The University Of California Antibodies against cleaved cdcp1 and uses thereof
KR20240004801A (en) 2021-05-05 2024-01-11 브리스톨-마이어스 스큅 컴퍼니 Lactone and lactam-containing compounds useful as immunomodulators
AU2022279156A1 (en) 2021-05-18 2023-11-02 Abbott Laboratories Methods of evaluating brain injury in a pediatric subject
EP4352077A1 (en) 2021-06-09 2024-04-17 Bristol-Myers Squibb Company Cyclic peptide immunomodulators
CA3222291A1 (en) 2021-06-14 2022-12-22 Jaime MARINO Methods of diagnosing or aiding in diagnosis of brain injury caused by acoustic energy, electromagnetic energy, an over pressurization wave, and/or blast wind
WO2023283523A1 (en) 2021-07-06 2023-01-12 Bristol-Myers Squibb Company 2,3-dihydrobenzo[b][l,4]dioxin-6-yl containing compounds useful as immunomodulators
WO2023034777A1 (en) 2021-08-31 2023-03-09 Abbott Laboratories Methods and systems of diagnosing brain injury
AU2022354059A1 (en) 2021-09-30 2024-03-28 Abbott Laboratories Methods and systems of diagnosing brain injury
WO2023089131A1 (en) 2021-11-19 2023-05-25 Lykera Biomed, S.A. Treatment and diagnosis of diseases associated to pathogenic fibrosis
WO2023114978A1 (en) 2021-12-17 2023-06-22 Abbott Laboratories Systems and methods for determining uch-l1, gfap, and other biomarkers in blood samples
WO2023129942A1 (en) 2021-12-28 2023-07-06 Abbott Laboratories Use of biomarkers to determine sub-acute traumatic brain injury (tbi) in a subject having received a head computerized tomography (ct) scan that is negative for a tbi or no head ct scan
WO2023150652A1 (en) 2022-02-04 2023-08-10 Abbott Laboratories Lateral flow methods, assays, and devices for detecting the presence or measuring the amount of ubiquitin carboxy-terminal hydrolase l1 and/or glial fibrillary acidic protein in a sample
GB202204813D0 (en) 2022-04-01 2022-05-18 Bradcode Ltd Human monoclonal antibodies and methods of use thereof
WO2024006876A1 (en) 2022-06-29 2024-01-04 Abbott Laboratories Magnetic point-of-care systems and assays for determining gfap in biological samples
WO2024059708A1 (en) 2022-09-15 2024-03-21 Abbott Laboratories Biomarkers and methods for differentiating between mild and supermild traumatic brain injury

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4954617A (en) * 1986-07-07 1990-09-04 Trustees Of Dartmouth College Monoclonal antibodies to FC receptors for immunoglobulin G on human mononuclear phagocytes
DE69029036T2 (en) * 1989-06-29 1997-05-22 Medarex Inc SPECIFIC REAGENTS FOR AIDS THERAPY
CA2067244C (en) * 1989-10-20 2000-12-19 Li Shen Monoclonal antibody specific for iga receptor
ES2129029T5 (en) * 1990-10-05 2005-10-16 Celldex Therapeutics, Inc. DIRECT IMMUNOSTIMULATION WITH BISPECIFIC REAGENTS.
JPH07501451A (en) 1991-11-25 1995-02-16 エンゾン・インコーポレイテッド Multivalent antigen binding protein
AU5322494A (en) * 1992-10-02 1994-04-26 Trustees Of Dartmouth College Bispecific reagents for redirected targeting of low density lipoprotein
ATE187494T1 (en) 1992-12-11 1999-12-15 Dow Chemical Co MULTIVALENT SINGLE CHAIN ANTIBODIES
IT1271461B (en) * 1993-12-01 1997-05-28 Menarini Ricerche Sud Spa ANTI-CD3 / ANTI-EGFR MONOCLONAL ANTI-BODY ANTIBODY, PROCESS FOR PRODUCTION AND ITS USE.
AU692239B2 (en) * 1994-03-07 1998-06-04 Medarex, Inc. Bispecific molecules having clinical utilities
JPH11507057A (en) * 1995-06-07 1999-06-22 メダレツクス・インコーポレーテツド Antiallergic bispecific molecule

Also Published As

Publication number Publication date
DE69735367D1 (en) 2006-04-27
JP3519415B2 (en) 2004-04-12
EP0914346B1 (en) 2006-03-01
US6193966B1 (en) 2001-02-27
WO1998002463A1 (en) 1998-01-22
AU3723397A (en) 1998-02-09
RU2201766C2 (en) 2003-04-10
IL128003A0 (en) 1999-11-30
CN1230198A (en) 1999-09-29
AU705643B2 (en) 1999-05-27
HUP9903951A3 (en) 2002-11-28
US6303755B1 (en) 2001-10-16
PL331091A1 (en) 1999-06-21
EP0914346A1 (en) 1999-05-12
US5922845A (en) 1999-07-13
ATE318845T1 (en) 2006-03-15
JP2000505091A (en) 2000-04-25
US20010014328A1 (en) 2001-08-16
HUP9903951A2 (en) 2000-03-28
IL128003A (en) 2003-06-24

Similar Documents

Publication Publication Date Title
EP0914346B1 (en) THERAPEUTIC MULTISPECIFIC COMPOUNDS COMPRISED OF ANTI-FCalpha RECEPTOR ANTIBODIES
US5837243A (en) Therapeutic compounds comprised of anti-Fc receptor antibodies
US6365161B1 (en) Therapeutic compounds comprised of anti-FC receptor binding agents
US6018031A (en) Binding agents specific for IgA receptor
JP2009516513A (en) Composition and production method of hybrid antigen binding molecule and use thereof
CN109069620B (en) Humanized antibodies to the CCR7 receptor
AU779116B2 (en) Therapeutic compounds comprised of anti-Fc receptor binding agents
US8697079B2 (en) IgE antibodies for the treatment of cancer
AU705643C (en) Therapeutic multispecific compounds comprised of anti-FCalpha receptor antibodies
CN115916839A (en) Bispecific antibodies against PDL1 x EGFR
Eccles c-erbB-2 as a target for immunotherapy
AU2003200884B2 (en) Therapeutic compounds comprised of anti-Fc receptor antibodies
TW202235436A (en) Siglec-15 binding protein and preparation and use thereof
AU5337300A (en) Therapeutic compounds comprised of anti-Fc receptor antibodies
ZA200201314B (en) Therapeutic compounds comprised of anti-Fc receptor binding agents.

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued