WO2010056399A1 - Method and composition for modulating the immune system and various inflammatory conditions comprising complement depletors - Google Patents

Method and composition for modulating the immune system and various inflammatory conditions comprising complement depletors Download PDF

Info

Publication number
WO2010056399A1
WO2010056399A1 PCT/US2009/046331 US2009046331W WO2010056399A1 WO 2010056399 A1 WO2010056399 A1 WO 2010056399A1 US 2009046331 W US2009046331 W US 2009046331W WO 2010056399 A1 WO2010056399 A1 WO 2010056399A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
protein
human
complement
certain instances
Prior art date
Application number
PCT/US2009/046331
Other languages
French (fr)
Inventor
Carl-Wilhelm Vogel
Paul W. Finnegan
William D. St. John
Kevin L. Stark
David C. Fritzinger
Original Assignee
Incode Biopharmaceutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Incode Biopharmaceutics, Inc. filed Critical Incode Biopharmaceutics, Inc.
Publication of WO2010056399A1 publication Critical patent/WO2010056399A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2839Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39541Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • 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
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • monoclonal antibodies bind to an antigen on a target cell.
  • an immune system effector cell e.g. a NK cell
  • the methods and compositions disclosed herein comprise co-administering an antibody and a complement modulating agent.
  • the complement modulating agent is an agent that depletes the complement cascade without producing (or only minimally producing) C5a.
  • the complement modulating agent is an agent that inhibits the formation and/or activity of C3b without producing (or only minimally producing) C5a.
  • a method of treating an inflammatory disorder comprising co-administering to an individual in need thereof a synergistically-effective amount of (a) an antibody or fusion protein comprising an Fc domain; and (b) a second agent selected from: a complement depleting agent, an agent that inhibits the formation and/or activity of C3b, or a combination thereof; and wherein the production of C5a is not stimulated, or is only minimally stimulated.
  • the IgG is IgGl, IgG2, IgG3, IgG4, or combinations thereof.
  • the antibody is a monoclonal antibody, a polyclonal antibody, or a combination thereof.
  • the antibody is a monoclonal antibody.
  • the antibody is an anti-CD3 antibody, an anti- CD 1 la ⁇ antibody, an anti-CD-20 antibody, an anti-CD22 antibody, an anti-CD-25 antibody, an anti-CD52 antibody, anti- TNF antibody, anti- IgE antibody, an anti- ⁇ 4-integrin antibody, or combinations thereof.
  • the antibody is an anti-CD3 antibody.
  • the antibody is muromonab-CD3.
  • the antibody is an anti- CDl la ⁇ antibody.
  • the antibody is efalizumab.
  • the antibody is an anti-CD20 antibody.
  • the antibody is GAlOl (R7159, Genentech), ocrelizumab (PRO70769), ofatumumab (HuMax-CD20 human IgGl monoclonal antibody or 2F2), PRO 131921 (Genentech), rituximab, veltuzumab (IMMU- 106 or hA20), or combinations thereof.
  • the antibody is an anti-CD22 antibody.
  • the antibody is epratuzumab.
  • the antibody is an anti-CD25 antibody.
  • the antibody is daclizumab, basiliximab, or a combination thereof.
  • the antibody is an anti-CD52 antibody.
  • the antibody is alemtuzumab. In some embodiments, the antibody is an anti- TNF antibody. In some embodiments, the antibody is infliximab, adalimumab, or a combination thereof. In some embodiments, the antibody is an anti- IgE antibody. In some embodiments, the antibody is omalizumab. In some embodiments, the antibody is an anti- ⁇ 4-integrin antibody. In some embodiments, the antibody is natalizumab.
  • the complement depleting agent is GR-2II, AGIIa, AGIIb-I, AR-2IIa, AR-2IIb, AR-2IIc, AR-2IId, CVF, a hybrid human C3-CVF protein, rC3, HC3-1496, HC3-1496-2, HC3-1496-3, HC3-1496-4, HC3-1496/1617, HC3-1496-8, HC3-1496-9, HC3-1496-10, HC3-1496-11, HC3-1496-12, HC3-1496-13, HC3- 1496-14, HC3-1496-15, HC3-1496-16, HC3-1496-17, or combinations thereof.
  • the agent that inhibits the formation and/or activity of C3b is complement receptor 1, sCRl, APT070, TPlO, TP20, sCRl [desLHR-A]), sCRl-SLe x , Crry, Crry-Ig, fucan, BS8, complestatin, Ecb, Efb, compstatin, rosmarinic acid, CRIT, CRIT-H17, glycyrrhetinic acid, CD55, sCD55, a CD55/MCP fusion protein, BCX- 1470, FUT- 175, Factor I, MCP, sMCP, heparin, an anti-properdin antibody, or combinations thereof.
  • the inflammatory disorder is Atopic dermatitis; Acute disseminated encephalomyelitis; Acute leukocyte -mediated lung injury; Addison's disease; AIDS dementia; Allergic rhinitis; Ankylosing spondylitis; Antiphospholipid antibody syndrome; Alzheimer's disorder; Asthma; Adult respiratory distress syndrome; Autoimmune hemolytic anemia; Autoimmune hepatitis; Autoimmune inner ear disease; Behcet's syndrome; Bronchitis; Bullous pemphigoid; Chagas disease; Chronic obstructive pulmonary disease; Coagulative Necrosis; Coeliac disease; Crohn's disorder; Collagenous colitis; Conjunctivitis; Cystic fibrosis; Dermatomyositis; Dermatitis; Diabetes mellitus type 1; Diabetes mellitus type 2; Distal proctitis; Diversion colitis; Encephalitis; Endometriosis; Endotoxin shock
  • a method of enhancing an immune response against a pathogen comprising co-administering to an individual in need thereof a synergistically-effective amount of (a) an antibody or fusion protein comprising an Fc domain; and (b) a second agent selected from: a complement depleting agent, an agent that inhibits the formation and/or activity of C3b, or a combination thereof; and wherein the production of C5a is not stimulated, or is only minimally stimulated.
  • the pathogen is a bacterium, a virus, a fungus, a prion, a protozoan, or combinations thereof.
  • the pathogen is RSV, Vibrio cholerae, Plasmodium falciparum, Plasmodium vivax, Plasmodium ovale, Plasmodium malariae, Mycobacterium tuberculosis, a influenza, HIV, E. coli O157:H7, a hepatitis virus, Poliovirus, Mycobacterium leprae, Yersinia pestis, Rickettsia prowazekii, Borrelia burgdorferi, Borrelia afzelii, Borrelia garinii, or combinations thereof.
  • the IgG is IgGl, IgG2, IgG3, IgG4, or combinations thereof.
  • the antibody is a monoclonal antibody, a polyclonal antibody, or a combination thereof. In some embodiments, the antibody is a monoclonal antibody. In some embodiments, the antibody is an anti- RSV gpF antibody. In some embodiments, the antibody is palivizumab. In some embodiments, the antibody is palivizumab. In some embodiments, the antibody is an anti- Pfs25 (Plasmodium falciparum) antibody, an anti- Pfs28 (Plasmodium falciparum), or combinations thereof. In some embodiments, the antibody is an anti- phenolic glycolipid TB (PGL-Tb) antibody. In some embodiments, the antibody is an anti- influenza matrix 2 protein (M2e) antibody. In some embodiments, the antibody is an anti- Y. pestis capsular Fl (Cafl) antibody.
  • M2e influenza matrix 2 protein
  • Cafl Y. pestis capsular Fl
  • the complement depleting agent is GR-2II, AGIIa, AGIIb- 1 , AR-2IIa, AR-2IIb, AR-2IIc, AR-2IH, CVF, a hybrid human C3-CVF protein, rC3, HC3-1496, HC3- 1496-2, HC3-1496-3, HC3-1496-4, HC3-1496/1617, HC3-1496-8, HC3-1496-9, HC3-1496-10, HC3-1496-11, HC3-1496-12, HC3-1496-13, HC3-1496-14, HC3-1496-15, HC3-1496-16, HC3- 1496-17, or combinations thereof.
  • the agent that inhibits the formation and/or activity of C3b is complement receptor 1, sCRl, APT070, TPlO, TP20, sCRl [desLHR- A]), sCRl-SLe x , Crry, Crry-Ig, fucan, BS8, complestatin, Ecb, Efb, compstatin, rosmarinic acid, CRIT, CRIT-H17, glycyrrhetinic acid, CD55, sCD55, a CD55/MCP fusion protein, BCX- 1470, FUT- 175, Factor I, MCP, sMCP, heparin, an anti-properdin antibody, or combinations thereof.
  • a modified human complement C3 protein comprising a human C3 protein, wherein one or more amino acid residues in the human C3 protein are substituted with a corresponding portion of a CVF protein; and wherein the protein does not comprise C3a.
  • the amino acid residues in the human C3 protein corresponding to amino acid residues 1496 to 1663 of SEQ ID NO :1 are substituted with a corresponding portion of a CVF protein.
  • the protein is at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, or at least about 95% homologous with a human C3 sequence.
  • one or more amino acid residues in the CVF portion is modified.
  • one or more amino acid residues in the CVF portion is modified, and the modification is selected from the group consisting of: (a) T1499D and L1501K; (b) I1507R, G1508D, N1509E, and V1510L; (c) S1519F, S1520I, L1521Q, N1522K, H1523S, and Q1524D; (d) D1528T, V1529L, P1530E, L1531E, and Q1532R; (e) 1519-1550 replaced with corresponding amino acid residues of SEQ ID NO: 1; (f) Q 157 IS, T1573S, N1576V, P1577Q, and R1578V; (g) 1596-1617 replaced with corresponding amino acid residues of SEQ ID NO: 1 ; (h) 1596- 1611 replaced with corresponding amino acid residues of SEQ ID NO:l; (i) V1598L, N1599D, D1600N, S1607L,
  • an amino acid of human C3 is substituted or deleted in a domain selected from: MGl, MG2, MG6, alpha-NT', TED, MG7, or combinations thereof.
  • Glutamine (Q) 163 of human C3 is substituted.
  • Glutamine (Q) 163 of human C3 is deleted.
  • Glutamine (Q) 163 of human C3 is deleted, the amino acid residue immediately preceding Glutamine 163 is deleted, and the three amino acid residues immediately following Glutamine 163 are deleted.
  • Serine (S) 1075 of human C3 is substituted.
  • Serine (S) 1075 of human C3 is substituted with Histidine (H). In some embodiments, Serine (S) 1075 of human C3 is substituted with Histidine (H) and the two preceding amino acid residues are deleted. In some embodiments, Asparagine (N) 738 of human C3 is substituted. In some embodiments, Asparagine (N) 738 of human C3 is substituted with Aspartic Acid. In some embodiments, Asparagine (N) 738 of human C3 is substituted with Aspartic Acid and the two preceding Glutamic acid residues (EE) are substituted with aspartic acid and serine (DS).
  • Asparagine (N) 738 of human C3 is substituted with Aspartic acid and the immediately preceding Glutamic acid (E) is substituted with Serine (S).
  • the Glutamine (Q) at position 1139 of human C3 is substituted with a Lysine (K) or a Tyrosine (Y).
  • FIG. 1 depicts the chain structures of human C3 and CVF with shaded portions present in the mature proteins.
  • FIGS. 2A-2D provide an alignment between prepro-CVF (SEQ ID NO:2) from Naja naja
  • FIGS. 3A-3H show nucleic acid and amino acid sequence of human prepro-C3. The NH2- and C-termini of the ⁇ - and ⁇ -chains, functionally important regions, and ligand binding sites are indicated. Amino acid residue numbering starts at the NH2 -terminus of the prepro-C3 molecule.
  • FIG. 4 shows survival curves of mice treated with CVF with or without monoclonal antibody
  • FIG. 5 shows survival curves of mice treated with HC3- 1496 with or without mAb.
  • complement C3 protein that modulates the innate immune system that includes the complement cascade as well as the immune system as a whole.
  • Complement C3 is the center piece that enables further complement activation from the three known key pathways that initially activate the complement cascade: (1) the classical pathway, (2) the alternative pathway, and (3) mannose binding lectin pathway.
  • the modulation of the innate immune system by depletion of complement C3 may alone alter the dynamics of the entire immune system including the adaptive immune system and also affect the interactions among the circulating blood cells and their secreted products as well as therapeutics such as monoclonal antibodies that is introduced into the circulatory system. When monoclonal antibodies are introduced into circulation, the antibodies bind to certain antigens on a target cell.
  • an immune system effector cell binds to the antibody and initiates apoptosis of the antibody-bound cell.
  • complement depletion through C3 depletion alters the interaction among immune system and inflammatory proteins in vivo (e.g., within any organism) as well as ex vivo (e.g., within blood products that in certain instances contain activatable complement proteins).
  • leukocyte comprises, by way of non- limiting example, lymphocytes, monocytes, macrophages, eosinophils, neutrophils and basophils.
  • Leukocytes include, but are not limited to, to hematopoietic stem cells and all myeloid and lymphoid lineages that arise from hematopoietic stem cells.
  • Leukocytes further include, but are not limited to, all immature, mature, undifferentiated and differentiated white blood cell populations including tissue specific and specialized varieties.
  • lymphocyte encompasses, by way of non- limiting example, B-cells, T-cells, NKT cells, and NK cells.
  • lymphocytes refers to all immature, mature, undifferentiated and differentiated white lymphocyte populations including tissue specific and specialized varieties. Lymphocytes include all B-cell lineages including pre-B-cells, Progenitor B cells, Early Pro-B cells, Late Pro-B cells, Large Pre-B cells, Small Pre-B cells, Immature B cells, Mature B cells, plasma B-cells, memory B-cells, B-I cells, B-2 cells and anergic AN1/T3 cell populations.
  • B-cell refers to, by way of non- limiting example, a pre-B-cell, Progenitor B cell, Early Pro-B cell, Late Pro-B cell, Large Pre-B cell, Small Pre-B cell, Immature B cell, Mature B cell, plasma B-cell, memory B-cell, B-I cell, B-2 cells and anergic AN1/T3 cell populations.
  • B-cell includes a B-cell that expresses an immunoglobulin heavy chain and/or light chain on its cells surface. Further, the term B-cell includes a B-cell that expresses and secretes an immunoglobulin heavy chain and/or light chain.
  • B-cell includes a cell that binds an antigen on its cell-surface.
  • B-cells or AN1/T3 cells are utilized in the processes described.
  • such cells are optionally substituted with any animal cell suitable for expressing, capable of expressing (e.g., inducible expression), or capable of being differentiated into a cell suitable for expressing an antibody including, e.g., a hematopoietic stem cell, a B-cell, a pre-B-cell, a Progenitor B cell, a Early Pro-B cell, a Late Pro-B cell, a Large Pre-B cell, a Small Pre-B cell, an Immature B cell, a Mature B cell, a plasma B-cell, a memory B-cell, a B-I cell, a B-2 cell, an anergic B-cell, or an anergic AN1/T3 cell.
  • immunological refers to the introduction of an antigen into an animal by any route.
  • routes are by way of intradermal injection, intravenous injection, intraocular administration, subcutaneous injection, intraperitoneal injection, oral administration, or topical administration.
  • antigen refers to a substance that is capable of inducing the production of an antibody. Further, antigen refers to a substance that binds to an antibody variable region.
  • antibody refers to monoclonal antibodies, polyclonal antibodies, bi-specific antibodies, multispecific antibodies, grafted antibodies, human antibodies, humanized antibodies, synthetic antibodies, chimeric antibodies, camelized antibodies, single- chain Fvs (scFv), single chain antibodies, Fab fragments, F(ab') fragments, disulfide-linked Fvs (sdFv), intrabodies, and anti-idiotypic (anti-Id) antibodies and antigen-binding fragments of any of the above.
  • antibodies include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules, i.e., molecules that contain an antigen binding site.
  • Immunoglobulin molecules are of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG i ; IgG 2 , IgG 3, IgG A, IgA 1 and IgA 2 ) or subclass.
  • the terms "antibody” and “immunoglobulin” are used interchangeably in the broadest sense.
  • an antibody is part of a larger molecule, formed by covalent or non-covalent association of the antibody with one or more other proteins or peptides.
  • derivative in the context of a polypeptide or protein, e.g. an antibody, refers to a polypeptide or protein that comprises an amino acid sequence which has been altered by the introduction of amino acid residue substitutions, deletions or additions.
  • derivative also refers to a polypeptide or protein which has been modified, i.e., by the covalent attachment of any type of molecule to the antibody.
  • a polypeptide or protein is modified, e.g., by glycosylation, acetylation, pegylation, phosphorylation, amidation, derivatization by protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc.
  • derivatives, polypeptides or proteins are produced by chemical modifications using suitable techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, metabolic synthesis of tunicamycin, etc.
  • a derivative a polypeptide or protein possesses a similar or identical function as the polypeptide or protein from which it was derived.
  • full length antibody “intact antibody” and “whole antibody” are used herein interchangeably, to refer to an antibody in its substantially intact form, and not antibody fragments as defined below. These terms particularly refer to an antibody with heavy chains contains Fc regions.
  • an antibody variant provided herein is a full length antibody.
  • the full length antibody is human, humanized, chimeric, and/or affinity matured.
  • An "affinity matured" antibody is one having one or more alteration in one or more CDRs thereof which result in an improvement in the affinity of the antibody for antigen, compared to a parent antibody which does not possess those alteration(s).
  • Preferred affinity matured antibodies will have nanomolar or even picomolar affinities for the target antigen.
  • Affinity matured antibodies are produced by suitable procedures. See, for example, Marks et al., (1992) Biotechnology 10:779-783 that describes affinity maturation by variable heavy chain (VH) and variable light chain (VL) domain shuffling. Random mutagenesis of CDR and/or framework residues is described in: Barbas, et al. (1994) Proc. Nat. Acad.
  • binding fragment means a portion or fragment of an intact antibody molecule, preferably wherein the fragment retains antigen-binding function.
  • antibody fragments include Fab, Fab', F(ab') 2 , Fd, Fd' and Fv fragments, diabodies, linear antibodies (Zapata et al. (1995) Protein Eng. 10: 1057), single-chain antibody molecules, single-chain binding polypeptides, scFv, bivalent scFv, tetravalent scFv, and bispecific or multispecific antibodies formed from antibody fragments.
  • Fab fragments are typically produced by papain digestion of antibodies resulting in the production of two identical antigen-binding fragments, each with a single antigen-binding site and a residual "Fc" fragment. Pepsin treatment yields a F(ab')2 fragment that has two antigen- combining sites capable of cross-linking antigen.
  • An "Fv” is the minimum antibody fragment that contains a complete antigen recognition and binding site. In a two-chain Fv species, this region consists of a dimer of one heavy- and one light-chain variable domain in tight, non- covalent association.
  • scFv single-chain Fv
  • one heavy- and one light-chain variable domain are covalently linked by a flexible peptide linker such that the light and heavy chains associate in a "dimeric" structure analogous to that in a two-chain Fv species. It is in this configuration that the three CDRs of each variable domain interact to define an antigen-binding site on the surface of the VH — VL dimer.
  • the six CDRs confer antigen-binding specificity to the antibody.
  • a single variable domain or half of an Fv comprising only three CDRs specific for an antigen
  • the Fab fragment also contains the constant domain of the light chain and the first constant domain (C H 1) of the heavy chain.
  • Fab fragments differ from Fab' fragments by the addition of a few residues at the carboxy terminus of the heavy-chain C H 1 domain including one or more cysteines from the antibody hinge region.
  • Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear a free thiol group.
  • F(ab') 2 antibody fragments originally were produced as pairs of Fab' fragments that have hinge cysteines between them. Other chemical couplings of antibody fragments are also suitable. Methods for producing the various fragments from monoclonal Abs include, e.g., Pluckthun, 1992, Immunol. Rev. 130: 152-188.
  • hypervariable region refers to the amino acid residues of an antibody which are responsible for antigen-binding.
  • the CDRs comprise amino acid residues from three sequence regions which bind in a complementary manner to an antigen and are known as CDRl, CDR2, and CDR3 for each of the VH and VL chains.
  • the CDRs typically correspond to approximately residues 24-34 (CDRLl), 50-56 (CDRL2) and 89-97 (CDRL3), and in the heavy chain variable domain the CDRs typically correspond to approximately residues 31-35 (CDRHl), 50-65 (CDRH2) and 95- 102 (CDRH3) according to Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)). It is understood that the CDRs of different antibodies may contain insertions, thus the amino acid numbering may differ.
  • the Kabat numbering system accounts for such insertions with a numbering scheme that utilizes letters attached to specific residues (e.g., 27A, 27B, 27C, 27D, 27E, and 27F of CDRLl in the light chain) to reflect any insertions in the numberings between different antibodies.
  • the CDRs typically correspond to approximately residues 26-32 (CDRLl), 50-52 (CDRL2) and 91-96 (CDRL3)
  • the CDRs typically correspond to approximately residues 26-32 (CDRHl), 53-55 (CDRH2) and 96-101 (CDRH3) according to Chothia and Lesk, J. MoI. Biol., 196: 901-917 (1987)).
  • monoclonal antibody refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that are present in minor amounts.
  • monoclonal antibodies are made, for example, by the hybridoma method first described by K ⁇ hler and Milstein (1975) Nature 256:495, or are made by recombinant methods, e.g., as described in U.S. Pat. No. 4,816,567.
  • monoclonal antibodies are isolated from phage antibody libraries using the techniques described in Clackson et al, Nature 352:624-628 (1991), as well as in Marks et al, J. MoI. Biol. 222:581- 597 (1991).
  • the antibodies herein include monoclonal, polyclonal, recombinant, chimeric, humanized, bi- specific, grafted, human, and fragments thereof including antibodies altered by any means to be less immunogenic in humans.
  • the monoclonal antibodies and fragments, etc., herein include “chimeric” antibodies and "humanized” antibodies.
  • chimeric antibodies include a portion of the heavy and/or light chain that is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567); Morrison et al Proc. Natl Acad. ScL 81:6851-6855 (1984).
  • a chimeric antibody contains variable regions derived from a mouse and constant regions derived from human in which the constant region contains sequences homologous to both human IgG2 and human IgG4.
  • "Humanized" forms of non-human (e.g., murine) antibodies or fragments are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab') 2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin.
  • Humanized antibodies include, grafted antibodies or CDR grafted antibodies wherein part or all of the amino acid sequence of one or more complementarity determining regions (CDRs) derived from a non-human animal antibody is grafted to an appropriate position of a human antibody while maintaining the desired binding specificity and/or affinity of the original non-human antibody.
  • CDRs complementarity determining regions
  • corresponding non-human residues replace Fv framework residues of the human immunoglobulin.
  • humanized antibodies comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications are made to further refine and optimize antibody performance.
  • the humanized antibody comprises substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • a humanized antibody also includes antibodies in which part, or all of the CDRs of the heavy and light chain are derived from a non-human monoclonal antibody, substantially all the remaining portions of the variable regions are derived from human variable region (both heavy and light chain), and the constant regions are derived from a human constant region.
  • the CDRl, CDR2 and CDR3 regions of the heavy and light chains are derived from a non-human antibody.
  • at least one CDR (e.g., a CDR3) of the heavy and light chains is derived from a non- human antibody.
  • Various combinations of CDRl, CDR2, and CDR3 can be derived from a non-human antibody and are contemplated herein.
  • the specified antibodies or binding molecules bind to a particular polypeptide, protein or epitope yet does not bind in a significant or undesirable amount to other molecules present in a sample.
  • the specified antibody or binding molecule does not undesirably cross-react with non-target antigens and/or epitopes.
  • a variety of immunoassay formats are used to select antibodies or other binding molecule that are immunoreactive with a particular polypeptide and have a desired specificity.
  • solid-phase ELISA immunoassays, BIAcore, flow cytometry and radioimmunoassays are used to select monoclonal antibodies having a desired immunoreactivity and specificity. See, Harlow, 1988, ANTIBODIES, A LABORATORY MANUAL, Cold Spring Harbor Publications, New York (hereinafter, "Harlow”), for a description of immunoassay formats and conditions that are used to determine or assess immunoreactivity and specificity.
  • “Selective binding”, “selectivity”, and the like refer the preference of an antibody to interact with one molecule as compared to another.
  • interactions between antibodies, particularly modulators, and proteins are both specific and selective.
  • an antibody is designed to "specifically bind” and “selectively bind” two distinct, yet similar targets without binding to other undesirable targets.
  • polypeptide peptide
  • protein protein
  • polypeptide peptide
  • the terms apply to naturally occurring amino acid polymers as well as amino acid polymers in which one or more amino acid residues is a non-naturally occurring amino acid, e.g., an amino acid analog.
  • the terms encompass amino acid chains of any length, including full length proteins (i.e., antigens), wherein the amino acid residues are linked by covalent peptide bonds.
  • isolated and purified refer to a material that is substantially or essentially removed from or concentrated in its natural environment.
  • an isolated nucleic acid is one that is separated from at least some of the nucleic acids that normally flank it or other nucleic acids or components (proteins, lipids, etc..) in a sample.
  • a polypeptide is purified if it is substantially removed from or concentrated in its natural environment. Methods for purification and isolation of nucleic acids and proteins are documented methodologies. Embodiments of "substantially” include at least 20%, at least 40%, at least 50%, at least 75%, at least 85%, at least 90%, at least 95%, or at least 99%.
  • the terms "individual,” “subject,” or “patient” are used interchangeably. As used herein, they mean any mammal (i.e. species of any orders, families, and genus within the taxonomic classification animalia: chordata: vertebrata: mammalia). In some embodiments, the mammal is a human. In some embodiments, the mammal is a non-human. In some embodiments, the mammal is a member of the taxonomic orders: primates (e.g. lemurs, lorids, galagos, tarsiers, monkeys, apes, and humans); rodentia (e.g.
  • mice, rats, squirrels, chipmunks, and gophers mice, rats, squirrels, chipmunks, and gophers); lagomorpha (e.g. hares, rabbits, and pika); erinaceomorpha (e.g. hedgehogs and gymnures); soricomorpha (e.g. shrews, moles, and solenodons); chiroptera (e.g., bats); cetacea (e.g. whales, dolphins, and porpoises); carnivora (e.g. cats, lions, and other feliformia; dogs, bears, weasels, and seals); perissodactyla (e.g.
  • artiodactyla e.g. pigs, camels, cattle, and deer
  • proboscidea e.g. elephants
  • sirenia e.g. manatees, dugong, and sea cows
  • cingulata e.g. armadillos
  • pilosa e.g. anteaters and sloths
  • didelphimorphia e.g. american opossums
  • paucituberculata e.g. shrew opossums
  • microbiotheria e.g. Monito del Monte
  • notoryctemorphia e.g.
  • the animal is a reptile (i.e. species of any orders, families, and genus within the taxonomic classification animalia: chordata: vertebrata: reptilia). In some embodiments, the animal is a bird (i.e. animalia: chordata: vertebrata: aves).
  • a health care worker e.g. a doctor, a registered nurse, a nurse practitioner, a physician's assistant, an orderly, or a hospice worker.
  • treat include alleviating, inhibiting or reducing symptoms, reducing or inhibiting severity of, reducing incidence of, prophylactic treatment of, reducing or inhibiting recurrence of, preventing, delaying onset of, delaying recurrence of, abating or ameliorating a disease or condition symptoms, ameliorating the underlying metabolic causes of symptoms, inhibiting the disease or condition, e.g., arresting the development of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a condition caused by the disease or condition, or stopping the symptoms of the disease or condition.
  • the terms further include achieving a therapeutic benefit.
  • therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated, and/or the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the individual.
  • compositions are optionally administered to an individual at risk of developing a particular disease, to an individual reporting one or more of the physiological symptoms of a disease, or to an individual at risk of reoccurrence of the disease.
  • the agents described herein be limited by the particular nature of the combination.
  • the agents described herein are optionally administered in combination as simple mixtures as well as chemical hybrids.
  • An example of the latter is where the agent is covalently linked to a targeting carrier or to an active pharmaceutical.
  • Covalent binding can be accomplished in many ways, such as, though not limited to, the use of a commercially available cross-linking agent.
  • combination treatments are optionally administered separately or concomitantly.
  • the terms “pharmaceutical combination”, “administering an additional therapy”, “administering an additional therapeutic agent” and the like refer to a pharmaceutical therapy resulting from the mixing or combining of more than one active ingredient and includes both fixed and non- fixed combinations of the active ingredients.
  • the term “fixed combination” means that at least one of the agents described herein, and at least one co-agent, are both administered to an individual simultaneously in the form of a single entity or dosage.
  • non- fixed combination means that at least one of the agents described herein, and at least one co-agent, are administered to an individual as separate entities either simultaneously, concurrently or sequentially with variable intervening time limits, wherein such administration provides effective levels of the two or more agents in the body of the individual.
  • the co-agent is administered once or for a period of time, after which the agent is administered once or over a period of time. In other instances, the co-agent is administered for a period of time, after which, a therapy involving the administration of both the co-agent and the agent are administered. In still other embodiments, the agent is administered once or over a period of time, after which, the co-agent is administered once or over a period of time.
  • cocktail therapies e.g. the administration of three or more active ingredients.
  • the terms "co-administration”, “administered in combination with” and their grammatical equivalents are meant to encompass administration of the selected therapeutic agents to a single individual, and are intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different times.
  • the agents described herein will be co-administered with other agents.
  • These terms encompass administration of two or more agents to an animal so that both agents and/or their metabolites are present in the animal at the same time. They include simultaneous administration in separate compositions, administration at different times in separate compositions, and/or administration in a composition in which both agents are present.
  • the agents described herein and the other agent(s) are administered in a single composition.
  • the agents described herein and the other agent(s) are admixed in the composition.
  • the terms "effective amount” or "therapeutically effective amount” as used herein, refer to a sufficient amount of at least one agent being administered which achieve a desired result, e.g., to relieve to some extent one or more symptoms of a disease or condition being treated.
  • the result is a reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system.
  • the result is a decrease in the growth of, the killing of, or the inducing of apoptosis in at least one abnormally proliferating cell, e.g., a cancer cell or a cancer stem cell.
  • an "effective amount” for therapeutic uses is the amount of the composition comprising an agent as set forth herein required to provide a clinically significant decrease in a disease.
  • An appropriate "effective” amount in any individual case is determined using any suitable technique, such as a dose escalation study.
  • the terms “administer,” “administering”, “administration,” and the like, as used herein, refer to the methods that are used to enable delivery of agents or compositions to the desired site of biological action. These methods include, but are not limited to oral routes, intraduodenal routes, parenteral injection (including intravenous, subcutaneous, intraperitoneal, intramuscular, intravascular or infusion), topical and rectal administration.
  • Administration techniques that are optionally employed with the agents and methods described herein, include e.g., as discussed in Goodman and Gilman, The Pharmacological Basis of Therapeutics, current ed.; Pergamon; and Remington's, Pharmaceutical Sciences (current edition), Mack Publishing Co., Easton, Pa.
  • the agents and compositions described herein are administered orally.
  • pharmaceutically acceptable refers to a material that does not abrogate the biological activity or properties of the agents described herein, and is relatively nontoxic (i.e., the toxicity of the material significantly outweighs the benefit of the material). In some instances, a pharmaceutically acceptable material is administered to an individual without causing significant undesirable biological effects or significantly interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • carrier refers to relatively nontoxic chemical agents that, in certain instances, facilitate the incorporation of an agent into cells or tissues.
  • compositions for increasing the efficacy of monoclonal antibody therapies without inducing inflammation are disclosed herein. Further disclosed herein, in certain instances, is a method of treating an immune disorder. In some embodiments, the methods described herein comprise co-administering an antibody and a complement modulating agent.
  • the antibody is a monoclonal antibody, a polyclonal antibody, or a combination thereof. In some embodiments, the antibody is a monoclonal antibody.
  • the antibody is a murine antibody. In some embodiments, the antibody is a rabbit antibody. In some embodiments, the antibody is a chimeric antibody. In some embodiments, the antibody is a humanized antibody. In some embodiments, the antibody comprises a murine variable region and a human Fc region.
  • the antibody is conjugated to a cytotoxic agent. In some embodiments, the antibody is conjugated to a chemotherapeutic agent. In some embodiments, the antibody is conjugated to a drug-activating enzyme. In certain instances, the drug-activating enzyme converts a non-toxic agent into a toxic agent. In some embodiments, the antibody is conjugated to a liposome. In some embodiments, the antibody is covalently bound to a radioactive isotope
  • the antibody binds to a specific antigen (e.g., CD20; CD25, IgE, TNF, flagella, fimbrae, LPS, toxins) differentially expressed and/or overexpressed on a target cell.
  • a specific antigen e.g., CD20; CD25, IgE, TNF, flagella, fimbrae, LPS, toxins
  • the binding of the antibody to the antigen results (either partially or fully) in an immune response against the cell.
  • the antibody comprises at least a portion of an Immunoglobulin G
  • the IgG antibody is IgGl, IgG2, IgG3, or IgG4. In some embodiments, the antibody is an IgGl antibody. In some embodiments, the antibody is an IgG2 antibody. In some embodiments, the antibody comprises at least a portion of an IgGl or IgG2Fc region. In certain instances, the Fc portion of an IgGl or IgG2 antibody interacts with (e.g., binds to) cell surface receptors called Fc receptors. In certain instances, an Fc receptor is expressed on NK cells, macrophages, neutrophils, and mast cells.
  • the methods described herein induce ADCC by natural killer (NK) cells or related cellular killing mechanisms.
  • NK natural killer
  • the binding of the Fc region of an antibody to an Fc receptor on a NK cell results in (either partially or fully) the NK cell releasing cytotoxic molecules (IFN- ⁇ ) and cytotoxic granules containing perforin and proteases.
  • IFN- ⁇ cytotoxic molecules
  • perforin forms pores in the membrane of an antigen presenting cell.
  • the cytotoxic molecules and proteases enter the antigen presenting cell and induce apoptosis. This process is called antibody dependent cell-mediated cytotoxicity (ADCC).
  • ADCC antibody dependent cell-mediated cytotoxicity
  • the methods described herein are used to treat an immune-mediated inflammatory disorder (e.g., by administering an antibody to an antigen expressed on a leukocyte).
  • an immune -mediated inflammatory disorder results from (either partially or fully) a derangement of an immune system whereby the immune system (e.g., via leukocytes) attacks self antigens.
  • the methods and compositions disclosed herein treat an immune-mediated inflammatory disorder by initiating an immune response (e.g., ADCC or related cellular killing mechanisms) against leukocytes.
  • the methods described herein are used to treat an infectious disorder (e.g., by administering an antibody to an antigen expressed on a pathogen).
  • the methods and compositions disclosed herein treat an infectious disorder by initiating an immune response (e.g., ADCC or related cellular killing mechanisms) against an infectious agent.
  • compositions for increasing the efficacy of monoclonal antibody therapies without inducing inflammation are disclosed herein. Further disclosed herein, in certain instances, is a method of activating an immune system, a method of inducing apoptosis in a cell, and a method of treating a neoplasia. In some embodiments, the methods described herein comprise co-administering an antibody and a complement modulating agent.
  • the complement modulating agent is a complement depleting agent and/or an agent that inhibits the formation and/or activity of C3b.
  • the complement system is part of the innate immune system. It attacks pathogens in a nonspecific manner (i.e., in a non-adaptive manner).
  • the complement system functions by recruiting immune system cells (e.g. macrophages and neutrophils) to the site of an infection by chemotaxis.
  • the complement system also utilizes the complement cascade to attack pathogens and recruit immune system cells.
  • the complement system also removes foreign substances by action of white blood cells (e.g. neutrophils and macrophages).
  • the inactive complement system comprises over 20 proteins and enzymes, most of which are present in an inactive form.
  • activation of complement e.g., by the presence of antibodies, the presence of antigens, or the spontaneous hydrolysis of C3 activates the inactive proteases in the system.
  • the proteases cleave targets (e.g., C3).
  • targets e.g., C3
  • the initial cleavage of the targets results (either partially or fully) in a cascade of cleavages.
  • an active complement system comprises anaphylatoxins (e.g. C3a and C5a), the membrane attack complex (MAC), and proteins that facilitate opsonization (e.g. C3b).
  • the complement system is activated by three pathways; the classical pathway, the alternative pathway, and the mannose-binding lectin pathway.
  • the CIq subunit of this enzyme either directly binds to an antigen or it binds to an antibody bound to an antigen.
  • the binding of CIq leads to a conformational change in CIq.
  • the conformational change in CIq leads to the activation of the two CIr subunits (CIr*) and CIs subunits (CIs*).
  • the activation of the CIr and CIs subunits results in an active Cl enzyme (Clq2Clr*2Cls*).
  • activated C 1 cleaves the protein complement component 4 (C4) into C4a and C4b.
  • C4b binds to the plasma membrane of a pathogen or a host cell.
  • activated C 1 also cleaves the protein complement component 2 (C2) into C2a and C2b.
  • C2a binds to C4b forming a C3 convertase (C4bC2a).
  • C4bC2a cleaves the protein C3 into C3a and C3b.
  • C3b binds to the membrane of a pathogen or host cell facilitating opsonization (e.g.
  • C3b binds to C4bC2a forming a C5 convertase (C4bC2aC3a) which cleaves complement component 5 (C5) into C5a and C5b.
  • the alternative pathway begins with the spontaneous hydrolysis of protein C3 (complement component 3), forming C3(H 2 O).
  • the hydrolysis of C3 causes a conformational change that allows Factor B to bind to C3(H 2 O).
  • Factor D cleaves Factor B into Ba and Bb.
  • Bb remains bound to C3(H 2 O) forming the complex C3 (H 2 O)Bb (the fluid phase C3 convertase).
  • the fluid phase C3 convertase cleaves C3 into C3a and C3b.
  • C3b binds to the plasma membrane of a pathogen or a host cell where it facilitates opsonization of the host cell or pathogen.
  • the C3b is bound by Factor B.
  • Factor B when bound to C3b, Factor B is cleaved by Factor D into Ba and Bb.
  • Bb remains bound to C3b forming an unstable C3 protease (C3Bb).
  • the unstable C3bBb protease is stabilized by the binding of the protein properdin (P) forming a more stable C3 convertase (C3bBbP).
  • the C3bBbP upon the binding of a second C3b component, the C3bBbP becomes a C5 convertase (C3bBbC3bP).
  • C3b binds to the membrane of an antigen presenting cell. In certain instances, the binding of C3b to an antigen presenting cell facilitates opsonization of the antigen presenting cell. In certain instances, the binding of C3b to an antigen presenting cell interferes with ADCC.
  • the methods described herein comprise depleting and/or inhibiting the activity of a C3 convertase (e.g., C4bC2a, C3(H 2 O)Bb, C3bBb, C3bBbP). In some embodiments, the methods described herein comprise inhibiting the formation of a C3 convertase.
  • depleting and/or inhibiting the activity of a C3 convertase, or inhibiting the formation of a C3 convertase comprises inhibiting the expression of C3, inhibiting the expression of any of the subunits of Cl, inhibiting the activity of Cl, inhibiting the expression of C4, inhibiting the expression of C2, inhibiting the expression of Factor B, increasing the expression of Factor I, administering exogenous Factor I, administering exogenous CRl.
  • the Membrane Attack Complex comprises five protein sub-units: C5b, C6, C7, C8, and C9.
  • C5b is produced by the cleavage of C5.
  • C5b binds C6.
  • C5bC6 is then bound by C7.
  • the binding of C7 induces a conformational change in C7, exposing a hydrophobic domain.
  • the hydrophobic domain enables C7 to insert itself into the plasma membrane of a pathogen or host cell.
  • C8 binds to the C5bC6C7 complex.
  • the binding of C8 also induces a conformational change in C8, exposing a hydrophobic domain that enables C8 to insert itself into the plasma membrane.
  • the C5bC6C7C8 complex induces the polymerization of multiple C9 proteins.
  • the C9 proteins form a pore in a plasma membrane.
  • the pore allows the free diffusion of fluids, ions, and proteins into and out of the cell; a process that ultimately leads to the death of a cell.
  • C3a, C4a, and C5a are anaphylatoxins.
  • anaphylatoxins are fluid phase proteins that bind to receptors on mast cells.
  • anaphylatoxins regulate smooth muscle spasms (e.g. bronchospasms), increase in the permeability of capillaries, and are chemotactic targets of leukocytes (e.g. the follow the increasing concentration gradient of an anaphylatoxin).
  • C3a and C5a are the most potent anaphylatoxins.
  • C3a regulates degranulation of Mast-cells and serves as a chemotactic target for eosinophile granulocytes.
  • C5a serves as a chemotactic target for granulocytes and macrophages, and regulates vascular permeability, smooth muscle spasms and mast cell degranulation.
  • C5a accelerates the growth of a neoplasm.
  • C5a recruits myeloid-derived suppressor cells (MDSC).
  • MDSC inhibit (either partially or fully) the activity of CD8 + T-cells.
  • antagonizing C5a and/or C5aR inhibits (either partially or fully) the growth of a neoplasm.
  • the methods described herein deplete and/or inhibit the complement cascade without or only minimally cleaving C5 (i.e., producing C5a).
  • the methods and compositions described herein treat inflammation (e.g., acute or chronic).
  • inflammation results from (either partially or fully) an infection.
  • inflammation results from (either partially or fully) damage to a tissue (e.g., by a burn, by frostbite, by exposure to a cytotoxic agent, or by trauma).
  • inflammation results from (either partially or fully) an autoimmune disorder.
  • inflammation results from (either partially or fully) the presence of a foreign body (e.g., a splinter).
  • inflammation results from exposure to a toxin and/or chemical irritant.
  • acute inflammation refers to inflammation characterized in that it develops over the course of a few minutes to a few hours, and ceases once the stimulus has been removed (e.g., an infectious agent has been killed by an immune response or administration of a therapeutic agent, a foreign body has been removed by an immune response or extraction, or damaged tissue has healed).
  • the short duration of acute inflammation results from the short half- lives of most inflammatory mediators.
  • acute inflammation begins with the activation of leukocytes (e.g., dendritic cells, endothelial cells and mastocytes).
  • leukocytes e.g., dendritic cells, endothelial cells and mastocytes.
  • the leukocytes release inflammatory mediators (e.g., histamines, proteoglycans, serine proteases, eicosanoids, and cytokines).
  • inflammatory mediators result in (either partially or fully) the symptoms associated with inflammation.
  • an inflammatory mediator dilates post capillary venules, and increases blood vessel permeability.
  • the increased blood flow that follows vasodilation results in (either partially or fully) rubor and calor.
  • permeability of the blood vessels results in an exudation of plasma into the tissue leading to edema. In certain instances, the latter allows leukocytes to migrate along a chemotactic gradient to the site of the inflammatory stimulant.
  • structural changes to blood vessels e.g., capillaries and venules
  • the structural changes are induced (either partially or fully) by monocytes and/or macrophages.
  • the structural changes include, but are not limited to, remodeling of vessels, and angiogenesis.
  • angiogenesis contributes to the maintenance of chronic inflammation by allowing for increased transport of leukocytes.
  • histamines and bradykinin irritate nerve endings leading to itching and/or pain.
  • chronic inflammation results from the presence of a persistent stimulant (e.g., persistent acute inflammation, bacterial infection (e.g., by Mycobacterium tuberculosis), prolonged exposure to chemical agents (e.g., silica, or tobacco smoke) and autoimmune reactions (e.g., rheumatoid arthritis)).
  • a persistent stimulant e.g., persistent acute inflammation, bacterial infection (e.g., by Mycobacterium tuberculosis), prolonged exposure to chemical agents (e.g., silica, or tobacco smoke) and autoimmune reactions (e.g., rheumatoid arthritis)
  • the persistent stimulant results in continuous inflammation (e.g., due to the continuous recruitment of monocytes, and the proliferation of macrophages).
  • the continuous inflammation further damages tissues which results in the additional recruitment of mononuclear cells thus maintaining and exacerbating the inflammation.
  • physiological responses to inflammation further include angiogenesis and fibrosis.
  • Inflammatory disorders include, but are not limited to, Atopic dermatitis; Acute disseminated encephalomyelitis; Acute leukocyte-mediated lung injury; Addison's disease; AIDS dementia; Allergic rhinitis; Ankylosing spondylitis; Antiphospholipid antibody syndrome; Alzheimer's disorder; Asthma; Adult respiratory distress syndrome; Autoimmune hemolytic anemia; Autoimmune hepatitis; Autoimmune inner ear disease; Behcet's syndrome; Bronchitis; Bullous pemphigoid; Chagas disease; Chronic obstructive pulmonary disease; Coagulative Necrosis; Coeliac disease; Crohn's disorder; Collagenous colitis; Conjunctivitis; Cystic fibrosis; Dermatomyositis; Dermatitis; Diabetes mellitus type 1; Diabetes mellitus type 2; Distal proctitis; Diversion colitis; Encephalitis; Endometriosis; Endotoxi
  • a T-cell mediated autoimmune disorder is characterized by a T-cell mediated immune response against self (e.g., native cells and tissues).
  • T-cell mediated autoimmune disorders include, but are not limited to colitis, multiple sclerosis, arthritis, rheumatoid arthritis, osteoarthritis, juvenile arthritis, psoriatic arthritis, acute pancreatitis, chronic pancreatitis, diabetes, insulin-dependent diabetes mellitus (IDDM or type I diabetes), insulitis, inflammatory bowel disease, Crohn's disease, ulcerative colitis, autoimmune hemolytic syndromes, autoimmune hepatitis, autoimmune neuropathy, autoimmune ovarian failure, autoimmune orchitis, autoimmune thrombocytopenia, reactive arthritis, ankylosing spondylitis, silicone implant associated autoimmune disease, Sjogren's syndrome, systemic lupus erythematosus (SLE), vasculitis syndromes (e.g., giant cell arteritis, Behcet's disease & Wegener's granulomatosis), vitiligo, secondary hematologic manifestation of autoimmune diseases (e.g.,
  • Pain includes, but is not limited to acute pain, acute inflammatory pain, chronic inflammatory pain and neuropathic pain.
  • hypersensitivity refers to an undesirable immune system response. Hypersensitivity is divided into four categories. Type I hypersensitivity includes allergies (e.g., Atopy, Anaphylaxis, or Asthma). Type II hypersensitivity is cytotoxic/antibody mediated (e.g., Autoimmune hemolytic anemia, Thrombocytopenia, Erythroblastosis fetalis, or Goodpasture's syndrome). Type III is immune complex diseases (e.g., Serum sickness, Arthus reaction, or SLE).
  • allergies e.g., Atopy, Anaphylaxis, or Asthma
  • Type II hypersensitivity is cytotoxic/antibody mediated (e.g., Autoimmune hemolytic anemia, Thrombocytopenia, Erythroblastosis fetalis, or Goodpasture's syndrome).
  • Type III is immune complex diseases (e.g., Serum sickness, Arthus reaction, or SLE).
  • Type IV is delayed-type hypersensitivity (DTH), Cell-mediated immune memory response, and antibody-independent (e.g., Contact dermatitis, Tuberculin skin test, or Chronic transplant rejection).
  • allergy means a disorder characterized by excessive activation of mast cells and basophils by IgE.
  • the excessive activation of mast cells and basophils by IgE results (either partially or fully) in an inflammatory response.
  • the inflammatory response is local.
  • the inflammatory response results in the narrowing of airways (i.e., bronchoconstriction).
  • the inflammatory response results in inflammation of the nose (i.e., rhinitis).
  • the inflammatory response is systemic (i.e., anaphylaxis).
  • non-cancer-based angiogenesis refers to the formations of new blood vessels that arises from non-cancer conditions.
  • angiogenesis occurs with chronic inflammation.
  • angiogenesis is induced by monocytes and/or macrophages.
  • the methods and compositions described herein treat obesity.
  • obesity means an accumulation of adipose tissue with a BMI of greater than or equal to 30kg/m .
  • obesity is characterized a proinflammatory state, increasing the risk of thrombosis.
  • obesity is associated with a low-grade inflammation of white adipose tissue (WAT).
  • WAT associated with obesity is characterized by an increased production and secretion of a wide range of inflammatory molecules including TNF-alpha and interleukin-6 (IL-6).
  • WAT is infiltrated by macrophages, which produce pro-inflammatory cytokines.
  • TNF-alpha is overproduced in adipose tissue.
  • IL-6 production increases during obesity.
  • metabolic syndrome is associated with fasting hyperglycemia; high blood pressure; central obesity; decreased HDL levels; elevated triglyceride levels; systemic inflammation; or combinations thereof.
  • metabolic syndrome is characterized by an increase in the levels of C-reactive protein, fibrinogen, (IL-6), and TNF ⁇ .
  • transplant complications means an immune-mediated complication resulting from an organ, a plurality of cells, and/or a tissue transplant.
  • a "transplant complication” can result from the recipient's immune system mounting an immune response against the transplanted organ, tissue, and/or plurality of cells.
  • a "transplant complication” also includes an immune response mounted against the recipient by the immune cells from the transplanted organ, plurality of cells, and/or tissue.
  • Transplant complications includes hyperacute transplant rejection, acute transplant rejection, chronic transplant rejection, acute graft-versus-host disease, chronic graft-versus-host disease, or combinations thereof.
  • ccines [0083] Disclosed herein, in certain embodiments, is a method of manufacturing a vaccine.
  • a pathogen e.g., a virus
  • complement is needed to mount a successful immune reaction.
  • inhibiting or depleting the complement system in an organism with active infection facilitates replication of the pathogen.
  • the method comprises administering a complement depletor (e.g., CVF, hybrid human C3-CVF protein, or a modified human complement C3 protein) to a fertilized egg or a live chick embryos that has been inoculated (or challenged) with a pathogen.
  • a complement depletor e.g., CVF, hybrid human C3-CVF protein, or a modified human complement C3 protein
  • the complement depletor depletes C3 (i.e., removes the process of amplification of complement activation as well as activation of the terminal complement pathway).
  • the complement depletor removes the process of amplification of complement activation as well as activation of the terminal complement pathway.
  • depleting or inhibiting complement increases pathogen replication.
  • increasing pathogen replication increases production of a vaccine.
  • the method applies to any pathogen used in vaccine production.
  • the method applies to any in- vivo system, including certain cellular systems, used in vaccine production. ood Storage
  • a method of collecting and storing whole blood or blood components In certain instances, collecting blood (and the necessary exposure to collection tubes and bags), separating blood into components (e.g., plasma) and storing whole blood or blood components activate the complement cascade. In certain instances, activation of the complement cascade is amplified due to the activation of C3. In certain instances, the activation of C3 leads to the production of and then the release of C3a and C3b. In certain instances, the production of and then the release of C3a and C3b lead further to the activation of the terminal complement cascade and production of C5a and C5b-9.
  • the activation of C3 and any or all of the downstream complement proteins induces and/or exacerbates the degradation of whole blood (i.e., induces the degradation of components of whole blood), or lyses blood cells and/or blood components.
  • transfusing an organism with whole blood comprising activated C3 with or without activated C5 and related downstream proteins further results (partially or fully) in organ dysfunction or systemic inflammatory response.
  • depleting the complement system in whole blood increases: (a) the shelf-life of blood and/or blood components and (b) the safety of infusion of whole blood, packed red blood cells and/or human blood components.
  • the method comprises contacting a complement depletor (e.g., CVF, recombinant CVF, hybrid human C3-CVF protein, a modified human complement C3 protein which depletes complement at C3 without activating C5) with whole blood or blood components.
  • a complement depletor e.g., CVF, hybrid human C3-CVF protein, a modified human complement C3 protein
  • the method comprises contacting an HC3-1496 protein, an analogue thereof, or combinations thereof with whole blood.
  • the method comprises contacting an HC3-1496 protein, an analogue thereof, or combinations thereof with whole blood before storage of the blood or separation of the blood into components.
  • contacting a complement depletor e.g., HC3-1496
  • an analogue thereof, or combinations thereof with whole blood removes the process of amplification of complement activation as well as activation of the terminal complement pathway.
  • depleting complement decreases the concentration of C3a and/or C3b and/or C5a and/or C5b, including C5b-9 complex.
  • decreasing the concentration of C3a and/ or C3b and/or C5a and/or C5b, including C5b-9 complex increases the amount of whole blood (or blood components) that can be transfused, increases the storage time for whole blood (or blood components), protects organs, reduces systemic inflammatory response, allows for reinfusion of shed blood which has been decomplemented by a complement depletor, or combinations thereof.
  • the antibody is an anti-CD3 antibody, an anti-CD 11 a ⁇ antibody, an anti-CD-20 antibody, an anti-CD22 antibody, an anti-CD-25 antibody, an anti-CD52 antibody, anti- TNF antibody, anti- IgE antibody, an anti- ⁇ 4-integrin antibody, anti- RSV gpF antibody, or combinations thereof.
  • the antibody is an anti-CD3 antibody.
  • CD3 is expressed on T-cells.
  • the binding of an antibody to a T-cell induces an immune response against the T-cell.
  • antagonizing CD3 prevents T cells from replicating, and from activating B cells.
  • B-cells bind to antigens (e.g., those expressed by transplanted organs and stimulate an immune response against the antigen presenting cell.
  • an anti-CD3 antibody is administered to treat an immune disorder.
  • an anti-CD3 antibody is administered to treat transplant rejection.
  • an anti-CD3 antibody is administered to treat kidney transplant rejection.
  • the antibody is muromonab-CD3. [0089] In some embodiments, the antibody is an anti-CD 11a antibody. In certain instances, CD 11 a is the ⁇ subunit of leukocyte function antigen- 1 (LFA-I). In certain instances, LFA-I is expressed by T-cells. Further, in certain instances, the binding of an antibody to a T-cell induces an immune response against the T-cell. In certain instances, LFA-I binds to intercellular adhesion molecule (ICAM)- 1. In certain instances, ICAM- 1 modulates the migration of T cells to sites of inflammation. In some embodiments, an anti- CD 11a antibody is administered to treat an immune disorder. In some embodiments, an anti- CD 11a antibody is administered to treat plaque psoriasis. In some embodiments, the antibody is efalizumab.
  • the monoclonal antibody is an anti-CD20 antibody.
  • CD20 is expressed on about 90% of mature B-cells.
  • CD20 is absent (either partially or fully) on progenitor B-cells.
  • an anti-CD20 antibody is administered to treat a B-cell mediated disorder.
  • an anti- CD20 antibody is administered to treat an autoimmune disorder.
  • an anti- CD20 antibody is administered to treat rheumatoid arthritis, a transplant complication, a transplant rejection (e.g., hyperacute, actue, and chronic), graft-versus-host disease (e.g., actue, and chronic), idiopathic autoimmune hemolytic anemia, pure red cell aplasia, idiopathic thrombocytopenic purpura, Evans syndrome, vasculitis, multiple sclerosis, bullous skin disorders (e.g., pemphigus, pemphigoid), type 1 diabetes mellitus, Sjogren's syndrome, Devic's Syndrome, systemic lupus erythematosus, or a combination thereof.
  • a transplant rejection e.g., hyperacute, actue, and chronic
  • graft-versus-host disease e.g., actue, and chronic
  • idiopathic autoimmune hemolytic anemia pure red cell aplasia
  • the anti-CD20 antibody is epratuzumab, GAlOl (R7159, Genentech), ocrelizumab (PRO70769), ofatumumab (HuMax-CD20 human IgGl monoclonal antibody or 2F2), PRO 131921 (Genentech), rituximab, veltuzumab (IMMU- 106 or hA20), or combinations thereof.
  • the monoclonal antibody is an anti-CD22 antibody.
  • CD22 present or all or most mature B-lineage cells. In certain instances, CD22 is not expressed (or only minimally expressed) on hematopoietic stem cells.
  • an anti-CD22 antibody is administered to treat an inflammatory disorder. In some embodiments, an anti-CD22 antibody is administered to treat lupus (SLE). In some embodiments, the anti-CD22 antibody is epratuzumab.
  • the monoclonal antibody is an anti-CD25 antibody.
  • CD25 is expressed on T-cells.
  • the binding of an antibody to a T- cell induces an immune response against the T-cell.
  • antagonizing CD25 prevents T cells from replicating, and from activating B cells.
  • B-cells bind to antigens (e.g., those expressed by transplanted organs and stimulate an immune response against the antigen presenting cell.
  • an anti-CD25 antibody is administered to treat an immune disorder.
  • an anti-CD3 antibody is administered to treat transplant rejection.
  • an anti-CD3 antibody is administered to treat kidney transplant rejection.
  • an anti-CD3 antibody is administered to treat graft-versus-host disease (e.g., acute, and chronic). In some embodiments, an anti-CD3 antibody is administered to treat multiple sclerosis. In some embodiments, the monoclonal antibody is daclizumab, basiliximab, or a combination thereof.
  • the monoclonal antibody is an anti-CD52 antibody.
  • CD52 is expressed on the surface of mature lymphocytes (e.g., expressed by lymphocytes, especially T cells, monocytes, macrophages, and monocyte-derived dendritic cells). In certain instances, CD52 is absent (either partially or fully) from hematopoietic progenitor cells.
  • an anti-CD52 antibody is administered to condition an individual for a transplant.
  • an anti-CD52 antibody is administered to condition an individual for a bone marrow transplant and/or kidney transplant.
  • an anti-CD52 antibody is administered to treat an immune disorder.
  • an anti-CD52 antibody is administered to treat multiple sclerosis and/or graft- versus-host disease (e.g., acute, and chronic).
  • the anti-CD52 antibody is alemtuzumab.
  • the monoclonal antibody is an anti- TNF antibody.
  • TNF- ⁇ mediates inflammation.
  • TNF- ⁇ levels are elevated in individuals with Crohn's Disease.
  • TNF- ⁇ levels are elevated in sera and synovial fluid of individuals with rheumatoid arthritis.
  • the binding of an antibody to a TNF- ⁇ molecule initiates an immune response against the TNF- ⁇ molecule.
  • an immune response against a TNF- ⁇ molecule inhibits (either partially or fully) the ability of the TNF- ⁇ molecule to bind to TNF receptors.
  • inhibiting the ability of the TNF- ⁇ molecule to bind to TNF receptors inhibits (either partially or fully) the production of proinflammatory cytokines (e.g., IL-6, IL-I), the production of proteases (e.g., the proteases that render blood vessels permeable to leukocytes), and the release of adhesion molecules.
  • an anti- TNF antibody is administered to treat an immune disorder.
  • an anti- TNF antibody is administered to treat rheumatoid arthritis, psoriatic arthritis, ulcerative colitis, Crohn's Disease, ankylosing spondylitis, or a combination thereof.
  • the monoclonal antibody is infliximab, adalimumab, or a combination thereof.
  • the monoclonal antibody is an anti- IgE antibody.
  • IgE mediates immune response to allergens.
  • the binding of an antibody to a IgE cell induces an immune response against the IgE cell.
  • IgE bound by an antibody cannot bind to a basophil.
  • IgE bound by an antibody attenuates degranulation.
  • an anti- IgE antibody is administered to treat an immune disorder.
  • an anti- IgE antibody is administered to treat an allergic reaction and/or hypersensitivity.
  • an anti- IgE antibody is administered to treat asthma.
  • the monoclonal antibody is omalizumab.
  • the monoclonal antibody is an anti- RSV gpF antibody. In some embodiments, an anti- RSV antibody is administered to treat an RSV infection. In some embodiments, the monoclonal antibody is palivizumab.
  • the monoclonal antibody is an anti- ⁇ 4-integrin (also known as VLA-4) antibody.
  • VLA-4 is a receptor found on leukocytes that mediates adhesion and migration of immune cells through interaction with its ligand, (VCAM)-I.
  • VCAM ligand
  • VLA-4 is expressed by leukocytes infiltrating the central nervous system and the gut.
  • the binding of an antibody to a leukocyte induces an immune response against the leukocyte.
  • VLA-4 mediates Th- 1 cell migration in animal models of MS, and experimental autoimmune encephalomyelitis.
  • the binding of an antibody to a Th- 1 cell induces an immune response against the Th- 1 cell.
  • an anti- ⁇ 4-integrin antibody is administered to treat an immune disorder.
  • an anti- ⁇ 4-integrin antibody is administered to multiple sclerosis, Crohn's disease, rheumatoid arthritis, allergic encephalomyelitis, adjuvant induced arthritis, diabetes mellitus, graft-versus-host disease (e.g., acute, and chronic), allograft rejection and/or immediate hypersensitivity reactions.
  • the monoclonal antibody is natalizumab.
  • the methods described herein comprising co-administering to an individual in need thereof a synergistically-effective amount of (a) an antibody or fusion protein comprising an Fc domain; and (b) a complement modulating agent.
  • the complement modulating agent is a complement depleting agent, an agent that inhibits the formation and/or activity of C3b, or a combination thereof.
  • the complement modulating agent depletes and/or inhibits the activity of a C3 convertase; or inhibits the formation of a C3 convertase (e.g., by inhibiting the expression of C3, inhibiting the expression of any of the subunits of Cl, inhibiting the activity of Cl, inhibiting the expression of C4, inhibiting the expression of C2, inhibiting the expression of Factor B, or increasing the expression of Factor I).
  • the methods described herein comprising co-administering to an individual in need thereof a synergistically-effective amount of (a) an antibody or fusion protein comprising an Fc domain; and (b) a complement modulating agent.
  • the complement modulating agent is a complement enhancing (i.e., agonizing) agent.
  • a complement modulating agent increases the expression of a complement protein.
  • a complement modulating agent catalyzes the cleavage of a complement component (e.g., C3 or C5) by a complement convertase (e.g., a C3 convertase, or C5 convertase).
  • an anaphylatoxin modulator is administered to an individual in need thereof.
  • the anaphylatoxin modulator is an antagonist of C5a.
  • the C5a antagonist is chemotaxis inhibitory protein of S.
  • a C5a antagonist binds to a C5aR and thereby antagonizes the binding of C5a.
  • CHIPS binds to the C5a receptor (C5aR) on a macrophage thereby inhibiting the C5a induced chemotaxis of a macrophage.
  • C5aRAM and C5aRAD are derived from the modification of the C terminus of C5a.
  • the antagonist of C5aR activation is an antisense peptide.
  • the antisense peptide of C5a is PR226-MAP (LRTWSRRATRSTKTLKVV), PL37-MAP (RAARISLGPRCIKAFTE), or combinations thereof.
  • a C5a antagonist binds to a C5aR and thereby antagonizes the binding of C5a.
  • an anaphylatoxin modulator is administered to an individual in need thereof.
  • the anaphylatoxin modulator is an antagonist of C3a.
  • the C3a antagonist is SB-290157 (N(2)-[(2,2-diphenylethoxy)acetyl]-L-arginine).
  • SB-290157 binds to the C3a receptor (C3aR) thereby blocking the binding of C3a.
  • a complement activator is administered to an individual in need thereof.
  • the complement activator is GR-2II, a pectic arabinogalactan (e.g. AGIIa, and AGIIb-I), a pectin (e.g. AR-2IIa, AR-2IIb, AR-2IIc, and AR-2IId), CVF, a hybrid human C3-CVF protein, or combinations thereof.
  • AR-2IIa, AR- 21Ib, and AR-2IIc activate the complement system via the classical pathway and not the alternative pathway.
  • Cobra Venom Factor is a three-chain ( ⁇ -chain, ⁇ -chain, and ⁇ -chain) glycoprotein extracted from the reptile Naja sp.
  • CVF is a human complement system activating protein. It is structurally homologous to C3b. In certain instances, CVF binds to Factor B which is then cleaved by Factor D. The resulting complex, CVFBb, functions as a C3 convertase and a C5 convertase. At 7.5 hours CVFBb exhibits a longer half-life than C3bBb (1.5 minutes). Further, CVFBb is resistant to disassembly by Factor H and CVF is resistant to inactivation by Factor I.
  • CVFBb will continuously hydro lyze C3 and C5.
  • the continuous hydro lyzation of C3 and C5 results in the depletion (or exhaustion) of the complement system within several hours.
  • resynthesis of the components of the complement system begins quickly and the entire system is reconstituted with 5-10 days.
  • CVF is highly antigenic in vivo.
  • humanized CVF also known as, “humanized CVF” and “hybrid human C3-CVF protein”
  • these derivatives exhibit similar activity to natural CVF (e.g. 50-97% of the activity of natural CVF); however, they do not or have a reduced capability of activating an immune response in vivo.
  • several amino acids from the ⁇ -chain of the CVF polypeptide are removed.
  • the CVF polypeptide is conjugated to a human antibody (e.g.
  • a human C3 derivative and/or analog e.g. recombinant C3, rC3, humanized CVF
  • the human C3 derivative and/or analog comprises a portion of a CVF polypeptide sequence.
  • portions of a human C3 polypeptide e.g. the ⁇ -chain; or portions of the carboxy terminal
  • CVF polypeptide e.g. the ⁇ -chain; or portions of the carboxy terminal
  • the ⁇ -chain of the human C3 is replaced by the corresponding carboxy terminal amino acids of the CVF polypeptide.
  • the CVF analog and/or derivative is HC3-1496, HC3-1496-2, HC3-1496-3, HC3-1496-4, HC3- 1496/1617, HC3-1496-8, HC3-1496-9, HC3-1496-10, HC3-1496-11, HC3-1496-12, HC3- 1496-13, HC3-1496-14, HC3-1496-15, HC3-1496-16, HC3-1496-17, or combinations thereof.
  • hybrid human C3-CVF protein that has one or more of the following characteristics: (a) ability to deplete a complement component; (b) ability to mediate the cleavage of factor B; (c) ability to form C3 convertase; (d) ability to cleave C3 and/or C5 upon activation; (e) increased resistance to the regulatory actions of factors H and/or I as compared to native human C3 protein; (f) binding affinity to factor D; (g) increased intrinsic half-life than native human C3 protein; and (h) less immunogenicity than CVF.
  • the hybrid human C3-CVF protein has sequence changes that have C3 complement depleting characteristics similar or superior to that of recombinant or natural cobra venom factor, but without activation of C5.
  • a hybrid human C3-CVF protein has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, or at least about 95% identity with the human C3 sequence.
  • Two polynucleotide or polypeptide sequences are said to be “identical” if the sequence of nucleotides or amino acids in the two sequences is the same when aligned for maximum correspondence as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity.
  • a “comparison window” as used herein refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence is compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • optimal alignment of sequences for comparison is conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, WI), using default parameters.
  • This program embodies several alignment schemes described in the following references: Dayhoff, M.O. (1978) A model of evolutionary change in proteins - Matrices for detecting distant relationships. In Dayhoff, M.O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington DC Vol. 5, Suppl. 3, pp. 345-358; Hein J., 1990, Unified Approach to Alignment and Phylogenes pp. 626- 645 Methods in Enzymology vol.
  • the "percentage of sequence identity” is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window comprises additions or deletions (i.e. gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the percentage is calculated by determining the number of positions at which the identical nucleic acid bases or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e. the window size) and multiplying the results by 100 to yield the percentage of sequence identity.
  • the substituted portion of the CVF is within the alpha chain of C3. In some embodiments, the substituted portion of the CVF is a C-terminal portion of the alpha chain of C3. In some embodiments, only the C-terminal portion of the alpha chain of C3 is substituted with the corresponding region of CVF protein. In some embodiments, the substituted portion of CVF is within the beta chain of C3. In some embodiments, the substituted C-terminal portion includes amino acid 1663 of SEQ ID NO: 1 (amino acid sequence of prepro- human C3). In some embodiments, the substituted C-terminal portion is an internal portion that does not extend through the entire C-terminus of the human C3 protein.
  • the modified human C3 protein is a single chain protein. In some embodiments, the modified human C3 protein is cleaved into at least two chains in a form that resembles native human C3. In some embodiments, the modified human C3 protein is proteolytic cleaved to release a portion therefrom (e.g., a portion like a C3a). In some embodiments, the modified human C3 protein is a mature protein. In some embodiments, the modified human C3 protein has 1 to about 19 amino acids at the N-terminus that are not part of human C3 or CVF. In some embodiments, the modified human C3 protein contains the signal sequence.
  • the signal sequence is a non-human C3 signal peptide, such as a Drosophila signal sequence.
  • the modified human C3 protein has an affinity for factor B and supports formation of a convertase.
  • the resulting convertase cleaves C3 and not C5.
  • the modified C3 protein has modified affinity for factor B and/or factor D.
  • the modified human C3 protein shows partial or complete resistance to Factor H and/or Factor I.
  • the modified human C3 protein is substantially non-immunogenic.
  • the convertase has an intrinsic half-life between about 1.5 minutes and about 7 hours at 37°C. In some embodiments, the resulting convertase has an intrinsic half- life of at least about 7 hours at 37°C.
  • the methods described herein comprise administering to an individual in need thereof a modified human C3 protein, comprising a human C3 protein, wherein some or all amino acid residues in the human C3 protein corresponding to amino acid residues 700- 1663 of SEQ ID NO: 1 are substituted with a corresponding portion of a CVF protein, including but not limited to regions of from 20 to about 1000 amino acids, including but not limited to: 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 150, 175, 180, 190, 200, 250, 275, 300, 350, 375, 400, 450, 475, 500, 550, 575, 600, 650, 675, 700, 750, 775, 800, 850, 875, 900, 950 975, 1000.
  • the beta chain of C3 is intact, that is, is the same as in natural C3.
  • the invention provides a modified human C3 protein, comprising human C3 protein, wherein amino acid residues in the human C3 protein corresponding to amino acid residues 1550-1663, 1504-1663, 1348-1663, 1550-1617, 1504-1617, 1470-1663, 1348-1617, 1470-1617, 1264-1324, 1348-1386, 749-784, 874-921, 994-1663, 994-1550, 1496-1663, 1496- 1617, or 936-970 of SEQ ID NO:1 are substituted with a corresponding portion of CVF protein.
  • still smaller substitutions in these areas corresponds to smaller regions that result in a C3 with the desired CVF functions or qualities.
  • the methods described herein comprise administering to an individual in need thereof a modified human C3 protein, comprising a human C3 protein, wherein the amino acid residues in the human C3 protein corresponding to amino acid residues 1550-1663, 1504-1663, 1496-1663 or 1348-1663 of SEQ ID NO: 1 are substituted with a corresponding portion of CVF protein.
  • the modified human C3 proteins are referred to as HC3-1550, HC3- 1504, HC3-1496 and HC3-1348, respectively.
  • these proteins comprise a modified human C3 protein, comprising a human C3 protein, wherein the amino acid residues in the human C3 protein corresponding to amino acid residues 1550-1617 of SEQ ID NO: 1 are substituted with a corresponding protein of CVF protein. In some embodiments, these proteins comprise a modified human C3 protein, comprising a human C3 protein, wherein the amino acid residues in the human C3 protein corresponding to amino acid residues 1496-1663 of SEQ ID NO: 1 are substituted with a corresponding protein of CVF protein.
  • the methods described herein comprise administering to an individual in need thereof a modified human C3 protein, comprising a human C3 protein, wherein at least 68 amino acid residues in the human C3 protein corresponding to amino acid residues 1596 to 1638 of SEQ ID NO: 1 (amino acid sequence of prepro-human C3) are substituted with the corresponding portion of CVF.
  • the amino acid residues in the human C3 protein corresponding to amino acid residues 1596 to 1638 of SEQ ID NO: 1 (amino acid sequence of prepro-human C3) are substituted with amino acid residues 1575 to 1617 of SEQ ID NO:2 (amino acid sequence of prepro-CVF).
  • the modified human C3 protein is any as described in US 2005/0079585, filed 07/02/2004 and published 04/14/2005.
  • the methods described herein comprise administering to an individual in need thereof a proteins comprising a human C3 protein, wherein the amino acid residues in the human C3 protein corresponding to amino acid residues 1496 to 1663 of SEQ ID NO:1 (amino acid sequence of pro-human C3) are substituted with a corresponding portion of a CVF protein, and wherein one or more amino acid residues in the CVF portion of the modified human complement C3 protein are further modified.
  • the one or more amino acids in one or more of the following regions is modified: 1499-1501, 1507-1510, 1519- 1550, 1519-1524, 1528-1532, 1571-1578, 1596-1617, 1596-1611, and 1598-1608.
  • At least one, at least two, at least three, at least four, at least five, at least six, or more amino acids in one or more of these regions are modified. In some embodiments, any of one, two, three, four, five, six or more amino acids in one or more of these regions are modified. In some embodiments, the amino acids in one or more of these regions are substituted into corresponding amino acids in a human C3 protein, a cobra C3 protein, or other mammalian C3 proteins.
  • Examples of the modifications of amino acid residues in the CVF portion of the modified human complement C3 protein include: 1) T1499D and L1501K; 2) I1507R, G1508D, N1509E, and V1510L; 3) S1519F, S 15201, L1521Q, N1522K, H1523S, and Q1524D; 4) D1528T, V1529L, P 1530E, L 153 IE, and Q1532R; 5) 1519-1550 replaced with corresponding amino acid residues of SEQ ID NO: 1; 6) Q1571S, T1573S, N1576V, P1577Q, and R1578V; 7) 1596-1617 replaced with corresponding amino acid residues of SEQ ID NO: 1; 8) 1596-1611 replaced with corresponding amino acid residues of SEQ ID NO: l; 9) V1598L, N1599D, D1600N, S1607L, and R1608S; 10) V1598L, N1599D, and
  • amino acid positions indicated herein correspond to the amino acid positions in SEQ ID NO: 1, are in the actual position(s) in the modified human C3 protein. In some embodiments, the amino acid positions indicated herein correspond to the amino acid positions in SEQ ID NO: 1, are not in the actual position(s) in the modified human C3 protein.
  • the methods described herein comprise administering to an individual in need thereof a modified human complement C3 proteins comprising a human C3 protein, wherein the amino acid residues in the human C3 protein corresponding to amino acid residues 1496 to 1617 of SEQ ID NO: 1 are substituted with a corresponding portion of a CVF protein, and wherein amino acid residues in the CVF portion of the modified human complement C3 protein are modified with E1654Y and V1658E.
  • the modified human C3 protein is any as described in WO 05/107785, filed 04/29/2005 and published 11/17/2005, and US 60/859,330, filed on 11/15/2006.
  • the modified human C3 protein is selected from Table 1.
  • the modified human C3 proteins in Table 1 contain specific CVF sequences so as to create modified human C3 proteins with CVF function.
  • the portions of human C3 that are replaced with corresponding CVF sequences are indicated in the table. This table should in no way be construed as limiting. Other possible modifications are possible and are intended to be included within the rubric of "hybrid human C3-CVF protein" or "modified CVF".
  • Table 1 Exemplary Modified Human C3 Proteins
  • the modified human C3 protein is substantially the same number of amino acid residues as an unmodified human C3 protein. In some embodiments, the modified human C3 protein has additional non-C3 and non-CVF amino acids attached to the carboxy terminus of the protein.
  • the modified human C3 proteins of the present invention are in any form.
  • the modified human C3 protein has additional non-C3 and non-CVF amino acids attached to the carboxy terminus of the protein.
  • the modified C3 protein is in a single chain form comprising the signal sequence or in a single chain form with the signal sequence being cleaved.
  • the modified C3 protein is in a two-chain form that resembles the structure of human C3 as shown in Figure 1.
  • the modified human C3 protein is in a form that resembles human C3b, wherein the C3a portion is cleaved.
  • the cleavage sites are not the cleavage sites for human C3 proteins (e.g., depending on the host cells). For example, one, two, or more amino acids on either end of the cleavage site of human C3 are cleaved for the modified C3 protein.
  • the catalytic activity of a convertase containing the modified human C3 protein is at least 50% that of the convertase containing CVF. In some embodiments, the catalytic activity of a convertase containing the modified human C3 protein is greater than that of the convertase containing native human C3. In some embodiments, the catalytic activity is about any of 60%, 70%, 80% 90% or 100% that of the CVF convertase. In some embodiments, the invention provides convertases containing the modified human C3 protein having a catalytic activity that falls between the two, or that exceeds the activity of the convertase containing native human C3.
  • the invention additionally provides convertases containing the modified human C3 protein having catalytic activity from about 10% to about 1000%, or more, that of the convertase containing CVF, including but not limited to about any of 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 110%, 135%, 150%, 200%, 300%, 400%, 500%, 750%, 1000% and more.
  • the catalytic efficiency is approximately eight-fold greater for C3bBb compared to CVFBb when cleaving C3.
  • the catalytic efficiency of a convertase containing the modified human C3 protein is at least 50% that of the convertase containing CVF, and is greater than that of the convertase containing native human C3b.
  • the invention also provides convertases containing the modified human C3 protein having a catalytic efficiency that falls between the two, or that exceeds the efficiency of the convertase containing native human C3b.
  • the invention additionally provides convertases containing the modified human C3 protein having catalytic efficiency from about 10% to about 1000%, or more, that of the convertase containing CVF, including but not limited to about any of 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 110%, 135%, 150%, 200%, 300%, 400%, 500%, 750%, 1000% and more.
  • the C5 cleaving activity of the modified human C3 proteins is decreased as compared to native human C3.
  • the C5 cleaving activity is from non-detectable to about 50% of the activity native human C3, including but not limited to about any of 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, and 50%.
  • the binding affinity of the modified human C3 proteins to Factor B and/or its subsequent cleavage by factor D is reduced.
  • the modified human C3 protein is useful.
  • the methods described herein comprise administering to an individual in need thereof a convertases having the modified human C3 proteins exhibiting substantially the same complement-activating activity of those containing natural CVF.
  • the term "exhibit substantially the same complement-activating activity of natural CVF” means that the modified human C3 protein of the present invention have from about 5% to about 100%, from about 50 to about 97%, from about 80 to about 97% of the level of the complement activating activity of natural CVF as measured by the method of Cochran et al., (1970) J. Immunol. 105(1), 55-69.
  • the modified human C3 proteins disclosed herein have immunogenicity less than that of CVF.
  • the modified human C3 protein is substantially non-immunogenic.
  • the modified human C3 protein are as non-immunogenic as C3, or is at least about any of 50%, 60%, 70%, 80%, 90% less immunogenic than CVF.
  • immunogenicity is measured by any suitable method.
  • the intrinsic half- life of the convertase formed with the modified human C3 protein is greater than about 1.5 minutes or greater than about 10 minutes. In some embodiments, the intrinsic half-life is between that of the CVF-containing convertase (7 hours or longer) and that of native human C3 (1.5 minutes), including but not limited to about any of: 2 minutes, 10 minutes, 20 minutes, 30 minutes, 40 minutes, 50 minutes, 60 minutes, 90 minutes, 2 hours, 2.5 hours, 3 hours, 3.5 hours, 4 hours, 4.5 hours, 5 hours, 5.5 hours, 6 hours, 6.5 hours, 7 hours, 8 hours, 9 hours, 10 hours, 10.5 hours, or more.
  • the resistance of the modified human C3 proteins to factors H and/or I is greater than that for native human C3. In some embodiments, the modified human C3 proteins have about the same level of resistance to factors H and/or I as that of CVF.
  • a polynucleotide containing the nucleic acid sequence encoding of any of the modified human C3 proteins described herein is cloned into a vector.
  • suitable cloning vectors are constructed by any suitable manner, or are selected from a large number of cloning vectors commercially available.
  • cloning vectors have the ability to self-replicate, possess a single target for a particular restriction endonuclease, and/or carry genes for a marker for use in selecting clones containing the vector.
  • Suitable examples include plasmids and bacterial viruses, e.g., pUC18, pUC19, Bluescript (e.g., pBS SK+) and its derivatives, mpl8, mpl9, pBR322, pMB9, CoIEl, pCRl, RP4, phage DNAs, and shuttle vectors such as pSA3 and pAT28.
  • vectors for expression in Drosophila cells are pMT/BiP-V5-HisA, pMT/BiP-V5-HisB, and pMT/BiP-V5-HisC.
  • Example of vectors for expression in Pichia pastoris are pPICZA, pPICZB, pPICZC, pPICZf -A, pPICZf -B, and pPICZf -C. These and many other cloning vectors are available from commercial vendors such as BioRad, Strategene, and Invitrogen. Suitable vectors also include mammalian expression vectors (e.g., pCDNA2, as pcDNA3.1, or pSecTag2 cloning vectors).
  • mammalian expression vectors e.g., pCDNA2, as pcDNA3.1, or pSecTag2 cloning vectors.
  • Expression vectors generally are replicable polynucleotide constructs that contain a polynucleotide according to the invention. It is implied that an expression vector must be replicable in the host cells either as episomes or as an integral part of the chromosomal DNA. Suitable expression vectors include but are not limited to plasmids, viral vectors, including adenoviruses, adeno-associated viruses, retroviruses, cosmids, and expression vector(s) disclosed in PCT Publication No. WO 87/04462.
  • vector components include, but are not limited to, one or more of the following: a signal sequence; an origin of replication; one or more marker genes; suitable transcriptional controlling elements (such as promoters, enhancers and terminator).
  • suitable transcriptional controlling elements such as promoters, enhancers and terminator.
  • one or more translational controlling elements are also usually required, such as ribosome binding sites, translation initiation sites, and stop codons.
  • the vectors containing the polynucleotides of interest are introduced into the host cell by any by any suitable method, including electroporation, transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; and infection (e.g., where the vector is an infectious agent such as vaccinia virus).
  • electroporation employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances
  • microprojectile bombardment e.g., where the vector is an infectious agent such as vaccinia virus.
  • infection e.g., where the vector is an infectious agent such as vaccinia virus.
  • the choice of introducing vectors or polynucleotides will often depend on features of the host cell.
  • the host cell is any suitable host cells (i.e., a cell capable of over- expressing heterologous DNAs).
  • suitable host cells include prokaryotes (such as E. coli or B. subtillis) and yeast (such as S. cerevisae, S. pombe, P. pastoris, or K. lactis).
  • Other examples of host cells include, but are not limited to, Drosophila S2 cell, a Sf9 cell, a HiFive cell, a BHK cell, and an HEK293 cell.
  • a modified human C3 protein is produced by culturing the host cell comprising the polynucleotides under a condition that the modified human C3 protein is expressed; and purifying the modified human C3 protein.
  • the modified human C3 protein is purified from the culture medium of the host cell.
  • the invention also provides modified human C3 proteins produced by the method.
  • the modified C3 protein produced is in more than one form (e.g., the protein produced is a mixture containing both single and double forms).
  • an HC3-1496 protein contains 19 additional amino acid residues on the N-terminus of the ⁇ -chain that are an artifact of the cloning procedure.
  • the coding sequence for the protein was engineered so that the signal sequence was removed, and a unique Afel site was engineered in to facilitate cloning of the insert into blunt-ended cloning sites.
  • an EcoRV site in the multiple cloning site region of the vector is used.
  • the cleavage site for the signal protease is coded for by the vector, resulting in an HC3-1496 protein as expressed in S2 cells having an additional 19 amino acids on the N-terminus of the protein, coded for by the vector between the signal cleavage site and the cloning site.
  • the same expression cassette is cloned into pSecTag2/Hygro, in certain instances, there are 27 vector-coded residues on the N-terminus of the protein.
  • an HC3- 1496 protein with a mature, C3-like N-terminus is obtained by utilizing a version of an HC3-1496 protein that is expressed with the native, human C3 signal sequence.
  • the mammalian C3 is produced and exported efficiently in mammalian cells in vivo.
  • a construct containing the native signal sequence is exported in mammalian expression systems (e.g., CHO cells).
  • a plasmid used in a method disclosed herein encodes (a) HC3-1496 and (b) the native signal sequence.
  • the coding sequence further comprises a restriction site on the 5' end (e.g., to facilitate easy transfer to an expression vector).
  • the coding sequence is engineered to comprise a restriction site on the 5' end by any suitable method (e.g., PCR).
  • an HC3-1496 protein is produced as two forms: (a) a two-chain form resembling mature C3 or "C3-like” form; and (a) a form resembling an activated C3 or "C3b- like” form, in which 77 amino acid residues (the C3a domain) are removed from the N-terminus of the larger ( ⁇ ) chain of the protein.
  • the C3b-likeform accounts for about 40% to about 70% of the protein.
  • a small amount of a single chain form is present.
  • the growth conditions of the CHO cells are optimized for the production of one form of the protein.
  • the C3a domain is removed and the construct comprises the mature N-terminus of the ⁇ ' -chain directly after the four arginines that constitute a furin-cleavage site (e.g., the furin site that is efficiently cleaved both in S2 cells and in vivo).
  • the site where S2 cells cleave the protein is modified by any suitable technique. Increasing the stability of human C3/CVF hybrid proteins
  • CVF cardiovascular disease 2019
  • injection of CVF into animals results in relatively long-term serum complement depletion (three to four days, depending on species).
  • the complement depletion caused by several hybrid human C3-CVF proteins is much shorter in duration (e.g., 24 hours or less).
  • hybrid human C3-CVF protein is any protein disclosed herein (e.g., HC3-1496 or analogues thereof).
  • the hybrid human C3-CVF protein is more resistant to dissociation of the C3b-like moiety and Bb.
  • the increased stability affects a longer biological life in depleting complement.
  • hybrid human C3-CVF protein-containing convertase that is nearly as stable as the CVF-containing enzyme.
  • the hybrid human C3-CVF protein has reduced affinity for factor H.
  • reducing the affinity of hybrid human C3-CVF protein for factor H increases the stability of the convertase and of the protein itself.
  • a hybrid human C3-CVF protein with increased stability is prepared by any suitable method (e.g., modification of one or more amino acids (e.g., by PCR)).
  • an amino acid is modified in a domain selected from: MGl, MG2, MG6, alpha- NT', TED, MG7, or combinations thereof.
  • the amino acid that is modified is Glutamine (Q) 163 of human C3.
  • Glutamine (Q) 163 of human C3 is deleted.
  • Glutamine (Q) 163 of human C3 is deleted, the amino acid residue immediately preceding Glutamine 163 is deleted, and the three amino acid residues immediately following Glutamine 163 are deleted.
  • the amino acid that is modified is Serine (S) 1075 of human C3.
  • Serine (S) 1075 of human C3 is replaced with Histidine (H).
  • Serine (S) 1075 of human C3 is replaced with Histidine (H) and the two preceding amino acid residues are removed.
  • the amino acid that is modified is Asparagine (N) 738 of human C3.
  • Asparagine (N) 738 of human C3 is replaced with Aspartic Acid.
  • Asparagine (N) 738 of human C3 is replaced with Aspartic Acid and the two preceding Glutamic acid residues (EE) are replaced with aspartic acid and serine (DS).
  • Asparagine (N) 738 of human C3 is replaced with Aspartic acid and the immediately preceding Glutamic acid (E) is replaced with Serine (S).
  • a Glutamine (Q) at position 1139 of human C3 is modified into a Lysine or to a Tyrosine. The aforementioned amino acids are identified according to mature C3b numbering.
  • a hybrid human C3-CVF protein is modified such that some of the residues of C3b that interact with factor H are replaced with sequences from CVF that interact with factor H by any suitable method (e.g., by analyzing a crystal structure).
  • the hybrid protein is more stable to cleavage in the presence of factors H and I.
  • the hybrid protein has an increased life in vivo and in vitro.
  • a complement component 1 (Cl) modulator is administered to an individual in need thereof.
  • the complement Cl modulator is a Cl inhibitor.
  • the C 1 inhibitor prevents fluid-phase C 1 activation.
  • administration of C 1 inhibitor prevents reperfusion injury.
  • dextran sulfate is administered to an individual in need thereof.
  • Cl inhibitor is administered before, after, or simultaneous with dextran sulfate.
  • dextran sulfate potentiates C 1 inhibitor.
  • a complement component Iq receptor (CIqR) is administered to an individual in need thereof.
  • CIq regulates the presentation of adhesion molecules on endothelial cells.
  • a CIq receptor e.g. cClqR, ClqR p , and gClqR
  • a CIq receptor prevents complement-mediated lysis of CIq sensitized erythrocytes.
  • administration of Cl inhibitor prevents reperfusion injury.
  • an antagonist of CIq binding is administered to an individual in need thereof.
  • the antagonist of CIq binding is CIq inhibitor, decorin, CSPG (chondroitin sulfate proteoglycan), CBP2 (complement binding peptide T), or combinations thereof.
  • CSPG partially or fully inhibits the binding of CIq to CIs and CIr, thereby interfering with the formation of the enzyme Cl.
  • CBP2 interferes with the binding of CIq to an antigen or antigen-bound antibody.
  • a complement receptor 1 is administered to an individual in need thereof.
  • CRl complement receptor 1
  • By binding to C3b and C4b CRl promotes phagocytosis and clearance of antigen- antibody complexes. Further, it inhibits both the classic and alternative pathways.
  • CRl acts as a decay-accelerator for both C3 and C5. Additionally, in certain instances, CRl acts as a Factor I cofactor.
  • a soluble CRl is administered to an individual in need thereof.
  • Soluble CRl lacks the transmembrane and cytoplasmic domains of CRl.
  • sCRl decrease the amount of MAC produced by the complement system.
  • sCRI ameliorates ischemic/reperfusion injuries.
  • sCRl reduces cellular and tissue injuries in animal models with acute or chronic inflammatory disorders.
  • the sCRl is APT070 (Mirococept), TPlO (Avant Immunotherapeutics), TP20 (Avant Immunotherapeutics), or combinations thereof.
  • a soluble CRl lacking the long homologous repeat A (LHR-A) domain (sCRl [desLHR-A]) is administered to an individual in need thereof.
  • sCRl [desLHR-A] lacks the transmembrane and cytoplasmic domains of CRl and the C4b binding domain.
  • sCRl [desLHR-A] inhibits the alternative pathway but exhibits a diminished ability to inhibit the classical pathway as compared to sCRl.
  • a soluble CRl (sCRl) bound by SLe x moieties is administered to an individual in need thereof.
  • SLe x is a carbohydrate ligand for selectins that, in certain instances, inhibits E-selectin and P-selectin mediated neutrophil adhesion.
  • sCRl-SLe x inhibits complement activation and inhibits the recruitment of neutrophils to the site of inflammation.
  • a complement receptor 1 -related gene/protein y is administered to an individual in need thereof.
  • a recombinant Crry (Crry-Ig) is administered to an individual in need thereof. Crry inhibits both the classic and alternative pathways. In certain instances, Crry acts as a decay-accelerator for both C3 and C5. Additionally, in certain instances, Crry acts as a Factor I cofactor. Complement Component 3 Convertase Modulators
  • a modulator of C3 convertase is administered to an individual in need thereof.
  • the C3 convertase modulator is a fucan.
  • a fucan is extracted from brown seaweed (e.g. Phaeophyceae, Ascophyllum nodosum, and Ecklonia kurome).
  • brown seaweed e.g. Phaeophyceae, Ascophyllum nodosum, and Ecklonia kurome.
  • a fucan partially or fully suppresses the classical pathway.
  • a fucan partially or fully suppresses the alternative pathway.
  • the fucan is BS8. In certain instances, BS8 partially or fully inhibited formation of C4bC2a by interfering with Cl activation.
  • BS8 partially or fully inhibited formation of C4bC2a by interfering C4 cleavage. In certain instances, BS8, partially or fully inhibits C3Bb by interfering with the binding of Factor B to C3b and by interfering with the binding of properdin.
  • a modulator of C3 convertase is administered to an individual in need thereof.
  • the C3 convertase modulator is complestatin.
  • complestatin interferes with the binding of C4b and C2b, and thus antagonizes the formation of the classical C3 convertase (C4bC2b).
  • a modulator of C3 convertase is administered to an individual in need thereof.
  • the C3 convertase modulator is extracellular complement- binding protein (Ecb).
  • Ecb is isolated from S. aureus.
  • it modulates C3b containing molecules (e.g. the alternative C3 convertase C3bB3, and the C5convertases C4bC2aC3b and C3bBbC3b) by blocking the ability of C3b containing molecules to cleave their substrates (e.g. C3 and C5).
  • a modulator of C3 convertase is administered to an individual in need thereof.
  • the C3 convertase modulator is extracellular fibrinogen-binding protein (Efb).
  • Efb is isolated from S. aureus.
  • Efb modulates C3b containing molecules (e.g. the alternative C3 convertase C3bB3) by blocking the ability of C3bBb to cleave C3.
  • a modulator of C3 convertase is administered to an individual in need thereof.
  • the C3 convertase modulator is compstatin.
  • compstatin antagonizes C3 convertases by binding to C3 and partially or fully inhibiting the ability for a C3 convertase to bind to and cleave C3.
  • a modulator of C3 convertase is administered to an individual in need thereof.
  • the C3 convertase modulator is rosmarinic acid.
  • rosmarinic acid reacts with the activated thioester of metastable C3b.
  • the reaction of rosmarinic acid and the activated thioester of metastable C3b results in covalent attachment of rosmarinic acid to a C3 convertase.
  • the covalent attachment of rosmarinic acid to a C3 convertase prevents the binding of a C3 convertase to a host cell or pathogen.
  • a modulator of C3 convertase is administered to an individual in need thereof.
  • the C3 convertase modulator is CRIT.
  • the C3 convertase modulator is a peptide sequence comprising the C-terminal 11 -amino-acid of the first CRIT-extracellular domain (CRIT-H 17).
  • CRIT inhibits the formation of C3 convertase.
  • CRIT binds to C2, thus inhibiting C4b from binding to C2 and forming C3 convertase.
  • a modulator of C3 convertase is administered to an individual in need thereof.
  • the C3 convertase modulator is glycyrrhetinic acid.
  • glycyrrhetinic acid modulates C2 and thus modulates the formation of the classical pathway C3 convertase.
  • a modulator of C5 convertase is administered to an individual in need thereof.
  • the C5 convertase modulator is an anti-complement component 5 (C5) murine monoclonal.
  • an anti-C5 mAb partially or fully inhibits the cleavage of C5 by C5 convertase.
  • an anti-C5 mAB inhibits the formation of C5a.
  • an anti-C5 mAb partially or fully inhibits the formation of C5b and thus the formation of a MAC.
  • an anti-C5 mAB does not inhibit the cleavage of a C3.
  • an anti-C5 mAB is derived from the variable region of the N 19/8 mAb. In certain instances, administration of an anti-C5 mAB ameliorates an autoimmune disease. In certain instances, an anti-C5 mAB partially or fully inhibits CDl Ib up- regulation. In certain instances, decreases the number of P-selectin presenting platelets. In certain instances, an anti-C5 mAB reduces the formation of leukocyte -platelet aggregates. In some embodiments, the C5 antibody is pexelizumab.
  • an anti-C5 murine single-chain antibody is administered to an individual in need thereof.
  • an anti-C5 murine single-chain antibody is derived from the variable region of the N 19/8 mAb.
  • an anti-C5 murine single-chain antibody is inhibits the cleavage of C5 and the production of C5a.
  • an anti-C5 murine single-chain antibody is partially or fully inhibits C5b-9-mediated hemolysis of erythrocytes.
  • an anti-C5 humanized single-chain antibody e.g. 5Gl.1-SC is administered to an individual in need thereof.
  • a modulator of C5 convertase is administered to an individual in need thereof.
  • the C5 convertase modulator is K76 (6,7-diformyl- 3',4',4a',5',6',7',8',8a'-octahydro-4,6',7'-trihydroxy-2',5',5',8a'-tetrame thyl spiro[l'(2'H)- naphthalene-2(3M)-benzofuran]), or a derivative thereof (e.g. TKIXc, and K76 COOH).
  • K76 antagonizes C5 convertase by interfering with the ability of C5 convertase to bind to and/or cleave C5.
  • a modulator of C5 convertase is administered to an individual in need thereof.
  • the C5 convertase modulator is a staphylococcal complement inhibitor (e.g. SCIN, SCIN-B, and SCIN-C).
  • a staphylococcal complement inhibitor is isolated from S. aureus.
  • an SCIN binds to and stabilizes a C3 convertase (e.g. C4bC2a and C3bBb).
  • the binding of an SCIN prevents the binding of a C3b subunit to the complex; thus, preventing the formation of a C5 convertase from a C3 convertase.
  • a CD55 is administered to an individual in need thereof.
  • CD55 also known as Decay Accelerating Factor (DAF)
  • DAF Decay Accelerating Factor
  • the binding of CD55 to C4b disassociates the C3 convertase of the classical pathway and thus also inhibits the formation of the classical C5 convertase.
  • the binding of CD55 to C3b disassociates the C3 and C5 convertases of the alternative pathway.
  • a CD55 protein is a soluble protein (sCD55).
  • sCD55 is administered to an individual in need thereof.
  • a CD59 protein is administered to an individual in need thereof.
  • CD59 inhibits the formation of a MAC by binding to C8 and C9 and thereby preventing their binding to the C5bC6C7 complex.
  • a soluble CD59 (sCD59) protein is administered to an individual in need thereof.
  • a CD59/CD55 fusion protein is administered to an individual in need thereof.
  • the CD59 subunit inhibits the formation of a MAC by binding to C8 and C9 and thereby preventing their binding to the C5bC6C7 complex.
  • a CD59/CD55 fusion protein prevents the formation of a MAC, and prevents the formation of or inhibits the activity of a C5 convertase.
  • the CD55 subunit binds to C4b thereby disassociating the C3 convertase of the classical pathway and inhibiting the formation of the classical C5 convertase.
  • the CD55 subunit binds to C3b thereby disassociating the C5 convertase of the alternative pathway.
  • a CD55/MCP fusion protein is administered to an individual in need thereof.
  • the CD55 subunit binds to C4b thereby disassociating the C3 convertase of the classical pathway and inhibiting the formation of the classical C5 convertase.
  • the CD55 subunit binds to C3b thereby disassociating the C5 convertase of the alternative pathway.
  • the MCP (Membrane Cofactor Protein, or CD46) subunit is a co-factor of Factor I.
  • the MCP subunit activates Factor I leading to the inactivation of a C3 convertase of the classical pathway and/or a C3 convertase of the alternative pathway.
  • the CD55/MCP fusion protein is a soluble protein sCD55/MCP (Complement Activation Blocker-2, CAB-2).
  • CAB-2 exhibits greater antagonism of convertases (e.g. C3 and C5) as compared to either CD55 administered alone, MCP administer alone, or CD55 and MCP administered in combination.
  • CAB-2 inhibits complement activation in vivo.
  • a Factor D modulator is administered to an individual in need thereof.
  • the Factor D modulator is a Factor D antagonist.
  • the Factor D antagonist is BCX- 1470 (2-amidino-6-(2-thiophene carboxy)benzothiophene methanesulfonate); FUT- 175 (6-amidino-2-naphthyl p-guanidinobenzoate dimethane- sulphonate); or combinations thereof.
  • Factor D antagonists inhibit the formation of the alternative pathway fluid phase C3 convertase by antagonizing Factor D's ability to bind to and cleave Factor B.
  • a Factor I protease and a co-factor thereof are administered to an individual in need thereof.
  • Factor I when bound to a co-factor, cleaves C3b and/or C4b; thus, inactivating them.
  • the inactivation of C4b iC4b
  • C3 convertase of the classical pathway and thus also inhibits the formation of the classical C5 convertase.
  • the inactivation of C3b iC3b
  • a Membrane Cofactor Protein (MCP OR CD46) is administered to an individual in need thereof.
  • MCP is a co-factor of Factor I.
  • MCP is administered to an individual in need thereof in soluble form (sMCP).
  • sMCP and/or MCP is administered before, after, or simultaneously with Factor I.
  • sMCP and/or MCP is administered with CD55.
  • administration of sMCP inhibits complement-mediated inflammation.
  • administration of MCP activates Factor I leading to the inactivation of a C3 convertase of the classical pathway and/or a C3 convertase of the alternative pathway.
  • administration of MCP activates Factor I leading to a decrease in the production of C5 convertase the classical pathway and/or inactivation of a C5 convertase of the alternative pathway.
  • a Factor H modulator is administered to an individual in need thereof.
  • Factor H serves as a co-factor for Factor I which cleaves C3b and/or C4b; thus, inactivating them.
  • the inactivation of C4b (iC4b) inhibits the activity of a C3 convertase of the classical pathway and thus also inhibits the formation of the classical C5 convertase.
  • the inactivation of C3b iC3b
  • the Factor H modulator is a Factor H mimic, an agent that that increases the concentration of Factor H, or a combination thereof.
  • the Factor H modulator is TT30 (Taligen Therapeutics).
  • a Factor B modulator is administered to an individual in need thereof.
  • the C3b is bound by Factor B.
  • Factor B when bound to C3b, Factor B is cleaved by Factor D into Ba and Bb.
  • Bb remains bound to C3b forming an unstable C3 protease (C3bBb).
  • C3b binds to the membrane of an antigen presenting cell.
  • the binding of C3b to an antigen presenting cell facilitates opsonization of the antigen presenting cell.
  • the Factor B modulator is an antibody or a portion thereof, that binds to Factor B.
  • the Factor B modulator is TAl 06 (Taligen Therapeutics). Heparin and Thrombin Inhibitors
  • heparin or inhibitors of thrombin or a derivative thereof e.g. LU
  • heparin interacts with Cl, C2, C3, C4, C5, C6, Cl, C8, C9, ClINH, factor I, factor H, factor B and factor P.
  • heparin or inhibitors of thrombin partially or fully inhibits the formation of the alternative pathway C3 convertase (C3bBb),the classical pathway C3 convertase (C4bC2a) or all forms of C5 convertase (e.g., C3bBbC3b, C4bC2aC3b).
  • an MAC modulator is administered to an individual in need thereof.
  • the MAC modulator is clusterin, vitronectin.
  • clusterin partially or fully inhibits the formation of fluid-phase MAC.
  • vitronectin partially or fully inhibits the formation of fluid-phase MAC.
  • an anti-properdin antibody is administered to an individual in need thereof.
  • the anti-properdin antibody is a monoclonal antibody.
  • an anti-properdin inhibits the stabilization of the alternative pathway unstable C3 convertase (C3Bb).
  • an anti-properdin antibody inhibits the formation of the alternative pathway C5 convertase (C3BbC3).
  • an anti-properdin antibody inhibits the formation of MAC.
  • the complement modulator is glycyrrhizin, glycyrrhetinic acid, or combinations thereof.
  • glycyrrhetinic acid modulates C2 and thus modulates the formation of the classical pathway C3 convertase.
  • a hybridoma is an immortalized antibody producing cell.
  • a laboratory animal e.g., a mouse or a rabbit
  • B-cells from the laboratory animal's spleen are extracted.
  • a hybridoma is generated by fusing (1) an extracted B-cell with (2) a myeloma cell (i.e., hypoxanthine-guanine- phosphoribosyl transferase negative, immortalized myeloma cells).
  • the B- cell and the myeloma cells are cultured together and exposed to an agent that renders their cell membranes more permeable (e.g., PEG).
  • the culture comprises a plurality of hybridoma, a plurality of myeloma cells, and a plurality of B-cells.
  • the cells are individual to culturing conditions that select for hybridoma (e.g., culturing with HAT media).
  • an individual hybridoma i.e., the clone
  • the hybridoma are isolated and cultured.
  • the hybridoma are injected into a laboratory animal.
  • the hybridoma are cultured in a cell culture.
  • Humanized Antibodies [00179]
  • the methods described herein comprise a humanized monoclonal antibody.
  • a humanized monoclonal antibody comprises heavy and light chain constant regions from a human source and variable regions from a murine source.
  • humanized immunoglobulins are constructed by genetic engineering.
  • humanized immunoglobulins comprise a framework that is identical to the framework of a particular human immunoglobulin chain (i.e., an acceptor or recipient), and three CDRs from a non-human (donor) immunoglobulin chain.
  • a limited number of amino acids in the framework of a humanized immunoglobulin chain are identified and chosen to be the same as the amino acids at those positions in the donor rather than in the acceptor.
  • a framework is used from a particular human immunoglobulin that is homologous to the donor immunoglobulin to be humanized.
  • a data bank for example, the National Biomedical Research Foundation Protein Identification Resource or the protein sequence database of the National Center for Biotechnology Information - NCBI
  • the extent of homology to different human regions can vary greatly, for example from about 40% to about 60%, about 70%, about 80%, or higher.
  • acceptor immunoglobulin By choosing as the acceptor immunoglobulin one of the human heavy chain variable regions that is most homologous to the heavy chain variable region of the donor immunoglobulin, fewer amino acids will be changed in going from the donor immunoglobulin to the humanized immunoglobulin. By choosing as the acceptor immunoglobulin one of the human light chain variable regions that is most homologous to the light chain variable region of the donor immunoglobulin, fewer amino acids will be changed in going from the donor immunoglobulin to the humanized immunoglobulin.
  • a humanized immunoglobulin comprises light and heavy chains from the same human antibody as acceptor sequences.
  • a humanized immunoglobulin comprises light and heavy chains from different human antibody germline sequences as acceptor sequences; when such combinations are used, one can readily determine whether the VH and VL bind an epitope of interest using conventional assays (e.g., an ELISA).
  • the human antibody will be chosen in which the light and heavy chain variable regions sequences, taken together, are overall most homologous to the donor light and heavy chain variable region sequences.
  • higher affinity is achieved by selecting a small number of amino acids in the framework of the humanized immunoglobulin chain to be the same as the amino acids at those positions in the donor rather than in the acceptor.
  • the relevant framework amino acids to change are selected based on differences in amino acid framework residues between the donor and acceptor molecules.
  • the amino acid positions to change are residues known to be important or to contribute to CDR conformation (e.g., canonical framework residues are important for CDR conformation and/or structure).
  • the relevant framework amino acids to change are selected based on frequency of an amino acid residue at a particular framework position (e.g., comparison of the selected framework with other framework sequences within its subfamily can reveal residues that occur at minor frequencies at a particular position or positions).
  • the relevant framework amino acids to change are selected based on proximity to a CDR. In some embodiments, the relevant framework amino acids to change are selected based on known or predicted proximity to the antigen-CDR interface or predicted to modulate CDR activity. In some embodiments, the relevant framework amino acids to change are framework residues that are known to, or predicted to, form contacts between the heavy (VH) and light (VL) chain variable region interface. In some embodiments, the relevant framework amino acids to change are framework residues that are inaccessible to solvent.
  • amino acid changes at some or all of the selected positions are incorporated into encoding nucleic acids for the acceptor variable region framework and donor CDRs.
  • altered framework or CDR sequences are individually made and tested, or are sequentially or simultaneously combined and tested.
  • the variability at any or all of the altered positions is from a few to a plurality of different amino acid residues, including all twenty naturally occurring amino acids or functional equivalents and analogues thereof. In some embodiments, non-naturally occurring amino acids are considered.
  • the humanized antibody sequence is cloned into a vector.
  • any suitable vector is used.
  • the vector is a plasmid, viral e.g. 'phage, or phagemid, as appropriate.
  • plasmid a plasmid, viral e.g. 'phage, or phagemid, as appropriate.
  • Many known techniques and protocols for manipulation of nucleic acid for example in preparation of nucleic acid constructs, mutagenesis, sequencing, introduction of DNA into cells and gene expression, and analysis of proteins, are described in detail in Short Protocols in Molecular Biology, Second Edition, Ausubel et al. eds., John Wiley & Sons, 1992. The disclosures of Sambrook et al. and Ausubel et al. are incorporated herein by reference for such disclosure.
  • any suitable host cell is transformed with the vector expressing the humanized antibody sequence.
  • the host cell is bacteria, mammalian cells, yeast and baculovirus systems.
  • the expression of antibodies and antibody fragments in prokaryotic cells such as E. coli is well established in the art. For a review, see for example Pluckthun, A. Bio/Technology 9: 545-551 (1991). Expression in eukaryotic cells in culture is also available to those skilled in the art as an option for production of the antibodies and antigen-binding fragments described herein, see for recent reviews, for example Raff, M.E. (1993) Curr. Opinion Biotech. 4: 573-576; Trill JJ. et al. (1995) Curr. Opinion Biotech 6: 553- 560, each of which is which is incorporated herein by reference for such disclosure.
  • a mammalian expression system is used.
  • the mammalian expression system is dehydrofolate reductase deficient ("dhfr- ") Chinese hamster ovary cells.
  • dhfr- CHO cells are transfected with an expression vector containing a functional DHFR gene, together with a gene that encodes a desired humanized antibody.
  • DNA is transformed by any suitable method.
  • suitable techniques include, for example, calcium phosphate transfection, DEAE Dextran, electroporation, liposome -mediated transfection and transduction using retrovirus or other virus, e.g., vaccinia or, for insect cells, baculovirus.
  • suitable techniques include, for example, calcium chloride transformation, electroporation and transfection using bacteriophage.
  • a DNA sequence encoding an antibody or antigen-binding fragment thereof is prepared synthetically rather than cloned.
  • the DNA sequence is designed with the appropriate codons for the antibody or antigen-binding fragment amino acid sequence. In general, one will select preferred codons for the intended host if the sequence will be used for expression.
  • the complete sequence is assembled from overlapping oligonucleotides prepared by standard methods and assembled into a complete coding sequence. See, e.g., Edge, Nature, 292:756 (1981); Nambair et al., Science, 223: 1299 (1984); Jay et al., J. Biol. Chem., 259:6311 (1984), each of which is which is incorporated herein by reference for such disclosure.
  • compositions comprising (a) an antibody; (b) a complement modulating agent; and (c) one or more physiologically acceptable carriers.
  • Physiologically acceptable carriers include excipients and auxiliaries which facilitate processing of the active agents into preparations which are used pharmaceutically. In certain instances, proper formulation is dependent upon the route of administration chosen.
  • a summary of pharmaceutical compositions is found, for example, in Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H.A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins, 1999).
  • compositions comprising (a) an antibody; (b) a complement modulating agent; and (c) a pharmaceutically acceptable diluent(s), excipient(s), or carrier(s).
  • the pharmaceutical compositions includes other medicinal or pharmaceutical agents, carriers, adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure, and/or buffers.
  • the pharmaceutical compositions also contain other therapeutically valuable substances.
  • a pharmaceutical composition refers to a mixture of an (a) an antibody; (b) a complement modulating agent; with (c) other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients.
  • the pharmaceutical composition facilitates administration of an antibody and complement modulating agent to an organism.
  • therapeutically effective amounts of an antibody are administered in a pharmaceutical composition to a mammal having a condition, disease, or disorder to be treated.
  • the mammal is a human.
  • a therapeutically effective amount varies depending on the severity and stage of the condition, the age and relative health of the individual, the potency of the antibody used and other factors.
  • the pharmaceutical formulations described herein are optionally administered to an individual by multiple administration routes, including but not limited to, oral, parenteral (e.g., intravenous, subcutaneous, intramuscular), intranasal, buccal, topical, rectal, or transdermal administration routes.
  • the pharmaceutical formulations described herein include, but are not limited to, aqueous liquid dispersions, self-emulsifying dispersions, solid solutions, liposomal dispersions, aerosols, solid dosage forms, powders, immediate release formulations, controlled release formulations, fast melt formulations, tablets, capsules, pills, delayed release formulations, extended release formulations, pulsatile release formulations, multiparticulate formulations, and mixed immediate and controlled release formulations.
  • Formulations suitable for intramuscular, subcutaneous, or intravenous injection include physiologically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • suitable aqueous and non-aqueous carriers, diluents, solvents, or vehicles including water, ethanol, polyols (propyleneglycol, polyethylene-glycol, glycerol, cremophor and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • Formulations suitable for subcutaneous injection also contain optional additives such as preserving, wetting, emulsifying, and dispensing agents.
  • an active agent is optionally formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • appropriate formulations include aqueous or nonaqueous solutions, preferably with physiologically compatible buffers or excipients.
  • Parenteral injections optionally involve bolus injection or continuous infusion.
  • Formulations for injection are optionally presented in unit dosage form, e.g., in ampoules or in multi dose containers, with an added preservative.
  • the pharmaceutical composition described herein are in a form suitable for parenteral injection as a sterile suspensions, solutions or emulsions in oily or aqueous vehicles, and contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • Pharmaceutical formulations for parenteral administration include aqueous solutions of an active agent in water soluble form. Additionally, suspensions are optionally prepared as appropriate oily injection suspensions.
  • the composition described herein are formulated for controlled or sustained release.
  • sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing a compound (e.g., modified human C3 proteins), which matrices are in the form of shaped articles, e.g. films, or microcapsules.
  • sustained-release matrices include polyesters, hydrogels (for example, poly(2- hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • the sustained release drug-delivery system is ATRIGEL® made by Atrix Laboratories. See, for example U.S. Pat. No. 6,565,874.
  • the ATRIGEL® drug delivery system consists of biodegradable polymers, similar to those used in biodegradable sutures, dissolved in biocompatible carriers.
  • proteins are blended into this liquid delivery system at the time of manufacturing.
  • proteins are blended into this liquid delivery system by the physician at the time of use.
  • the liquid product is injected through a small gauge needle or placed into accessible tissue sites through a cannula, displacement of the carrier with water in the tissue fluids causes the polymer to precipitate to form a solid film or implant. Proteins encapsulated within the implant are then released in a controlled manner as the polymer matrix biodegrades with time.
  • the ATRIGEL® system releases proteins over multiple days.
  • the ATRIGEL® system releases proteins over multiple weeks. In some embodiments, the ATRIGEL® system releases proteins over multiple months. In some embodiments, injectable sustained release systems (e.g., such as ProLease®, Medisorb®, manufactured by Alkermes) are used.
  • injectable sustained release systems e.g., such as ProLease®, Medisorb®, manufactured by Alkermes
  • the methods and compositions described herein comprise a modified human C3 protein linked to another polypeptide or a carrier.
  • a modified human C3 protein is linked to an antibody or fragments thereof.
  • fusion proteins or conjugations are constructed by an suitable method. s
  • kits for use in the methods provided herein include one or more containers comprising any of the compounds (e.g., modified human C3 proteins) described herein and instructions for use in accordance with any of the methods of the invention described herein.
  • these instructions comprise a description of administration of the compound (e.g., modified human C3 proteins) to deplete complement according to any of the methods described herein.
  • the kit further comprises a description of selecting an individual suitable for treatment.
  • the instructions relating to the use of a compound include information as to dosage, dosing schedule, and route of administration for the intended treatment.
  • the containers are unit doses, bulk packages (e.g., multi-dose packages), sub-unit doses, or combinations thereof.
  • instructions supplied in the kits of the invention are typically written instructions on a label or package insert (e.g., a paper sheet included in the kit).
  • instructions supplied in the kits of the invention are machine-readable instructions (e.g., instructions carried on a magnetic or optical storage disk).
  • kits of this invention are in suitable packaging.
  • suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like.
  • the kits comprise packages for use in combination with a specific device, such as an inhaler, nasal administration device (e.g., an atomizer) or an infusion device such as a minipump.
  • a kit has a sterile access port (e.g., the container is an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the container has a sterile access port (e.g., the container is an intravenous solution bag having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is a compound disclosed herein (e.g., modified human C3 proteins).
  • the container further comprises a second pharmaceutically active agent.
  • the kits disclosed herein provide additional components such as buffers and interpretive information. Normally, the kit comprises a container and a label or package insert(s) on or associated with the container.
  • the kit contains instructions for administering the first and second therapies simultaneously and/or sequentially for the effective treatment of cancer.
  • the first and second therapies are present in separate containers or in a single container.
  • the kit comprises one distinct composition.
  • the kit comprises two or more compositions wherein one composition comprises a first therapy and one composition comprises a second therapy.
  • depletion of complement is effected by an ex vivo treatment, for example, by transfusing circulation of an individual through a matrix bearing a complement- depleting compound (e.g., a modified human C3 protein) to remove complement.
  • the method further comprises a step of removing (e.g. , by dialysis) anaphylactic peptides (e.g., C5a) and other low molecular weight inflammatory mediators (e.g., histamine and nitric oxide) prior to the decomplemented blood (or plasma) being returned to the individual.
  • the methods and compositions described herein comprises administering (a) a first therapy comprising an antibody and a complement modulating agent (e.g., a complement depleting agent and/or a C3b inhibiting agent) and (b) a second therapy useful for treating cancer.
  • a complement modulating agent e.g., a complement depleting agent and/or a C3b inhibiting agent
  • the second therapy includes surgery, radiation, hormone therapy, gene therapy, antibody therapy (including, but not limited to any of the antibodies disclosed herein), and/or chemotherapy.
  • compositions described herein are optionally used in the preparation of medicaments for the prophylactic and/or therapeutic treatment of conditions (e.g., an inflammatory disorder) that would benefit, at least in part, from amelioration.
  • conditions e.g., an inflammatory disorder
  • an individual in need of treatment with any of the methods and compositions described herein has been diagnosed with an inflammatory disorder.
  • an individual in need of treatment with any of the methods and compositions described herein has been diagnosed with Atopic dermatitis; Acute disseminated encephalomyelitis; Acute leukocyte -mediated lung injury; Addison's disease; AIDS dementia; Allergic rhinitis; Ankylosing spondylitis; Antiphospholipid antibody syndrome; Alzheimer's disorder; Asthma; Adult respiratory distress syndrome; Autoimmune hemolytic anemia; Autoimmune hepatitis; Autoimmune inner ear disease; Behcet's syndrome; Bronchitis; Bullous pemphigoid; Chagas disease; Chronic obstructive pulmonary disease; Coagulative Necrosis; Coeliac disease; Crohn's disorder; Collagenous colitis; Conjunctivitis; Cystic fibrosis; Dermatomyositis; Dermatitis; Diabetes mellitus type 1; Diabetes mellitus type 2; Distal proctitis; Diversion colitis; Ence
  • a method for treating any of the diseases or conditions described herein in an individual in need of such treatment involves administration of pharmaceutical compositions as described herein, or a pharmaceutically acceptable salt, pharmaceutically acceptable N-oxide, pharmaceutically active metabolite, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate thereof, in therapeutically effective amounts to said individual.
  • a complement modulating agent is co-administered with an antibody (e.g., during the administration of the antibody, or within one day of administration of the antibody) for the entire duration of antibody therapy.
  • a complement modulating agent is administered before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered one hour before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered two hours before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered three hours before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered four hours before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered five hours before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered six hours before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered eight hours before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered twelve hours before an antibody for the entire duration of antibody therapy.
  • a complement modulating agent is administered before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered one day before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered two days before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered three days before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered four days before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered five days before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered six days before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered seven days before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered two weeks before an antibody for the entire duration of antibody therapy.
  • the methods described herein comprise local administration of a synergistically-effective amount of (a) an antibody or fusion protein comprising an Fc domain; and (b) a complement depleting agent, an agent that inhibits the formation and/or activity of C3b, or a combination thereof.
  • the compositions disclosed herein are administered locally.
  • local treatment is effected by any suitable method, depending on the desired effect.
  • local treatment is effected by an autoinjector (e.g., a medical device that administers a device via a spring loaded syringe).
  • local depletion is effected when an effective amount of a complement-depleting compound (e.g., a modified human C3 protein) is administered locally to an organ, tissue, cavity, or intradermally. In some embodiments, this results in a temporary depletion of complement in the area.
  • a complement-depleting compound e.g., a modified human C3 protein
  • local depletion is effected using an insulin-type pump that produces an intermittent or constant flow of the compound to a selected site.
  • local treatment employs a specific antibody which, when attached (e.g., chemically) to the compound localizes it to a specific tissue, a disease, or an infected cell to cause continuous depletion of complement in that area.
  • the method includes chemically linking the compound to an antibody (such as a monoclonal antibody) with an affinity for a specific tissue prior to the delivering step.
  • an antibody such as a monoclonal antibody
  • the antibody is attached to the compound (e.g., a modified human C3 protein) via recombinant DNA technology.
  • the methods described herein comprise systemic administration of a synergistically-effective amount of (a) an antibody or fusion protein comprising an Fc domain; and (b) a complement depleting agent, an agent that inhibits the formation and/or activity of C3b, or a combination thereof.
  • the compositions disclosed herein are administered systemically.
  • Systemic depletion is effected when an effective amount of a complement-depleting compound (e.g., a modified human C3 protein) is administered systemically, for example, intravenously or intraperitoneally. In some embodiments, this results in a temporary depletion of complement systemically.
  • a complement-depleting compound e.g., a modified human C3 protein
  • a composition described herein is administered once a day, once a week, semi-weekly, bi-weekly, once a month, semi-monthly, bi-monthly, every six months, or once a year.
  • a composition disclosed herein is administered via pulse therapy (e.g., a short, intensive administration of a composition disclosed herein, given at weekly intervals; or a portion of a standard dose administered at regular intervals until the full dose is reached).
  • a standard dose is divided into four sub-dose and each sub-dose is administered at four hour intervals.
  • Empirical considerations such as the half-life, generally will contribute to determination of the dosage.
  • the treatment is sustained until a desired suppression of disease symptoms occurs or until sufficient therapeutic levels are achieved.
  • compositions described herein are optionally administered before, during or after the occurrence of a disease or condition, and the timing of administering the composition varies in some embodiments.
  • a composition described here is used as a prophylactic and is administered continuously to individuals with a propensity to develop conditions or diseases in order to prevent the occurrence of the disease or condition.
  • a composition described herein is optionally administered to an individual during or as soon as possible after the onset of the symptoms.
  • the administration of the agents are optionally initiated within the first 48 hours of the onset of the symptoms, preferably within the first 48 hours of the onset of the symptoms, more preferably within the first 6 hours of the onset of the symptoms, and most preferably within 3 hours of the onset of symptoms.
  • the initial administration is optionally via any route practical, such as, for example, an intravenous injection, a bolus injection, infusion over 5 minutes to about 5 hours, a pill, a capsule, transdermal patch, buccal delivery, and the like, or combination thereof.
  • a composition described herein is preferably administered as soon as is practicable after the onset of a disease or condition is detected or suspected, and for a length of time necessary for the treatment of the disease, such as, for example, from about 1 month to about 3 months.
  • the length of treatment optionally varies for each individual, and the length is then determined using the known criteria.
  • a composition described herein is administered for at least 2 weeks, preferably about 1 month to about 5 years, and more preferably from about 1 month to about 3 years.
  • the administration of an antibody is optionally administered chronically, that is, for an extended period of time, including throughout the duration of the patient's life in order to ameliorate or otherwise control or limit the symptoms of the patient's disease or condition.
  • the administration of an antibody is optionally given continuously; alternatively, the dose of drug being administered is temporarily reduced or temporarily suspended for a certain length of time (i.e., a "drug holiday").
  • the length of the drug holiday optionally varies between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days.
  • the dose reduction during a drug holiday includes from 10%- 100%, including, by way of example only, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%.
  • a maintenance dose is administered if necessary. Subsequently, the dosage or the frequency of administration, or both, is reduced, as a function of the symptoms, to a level at which the improved disease, disorder or condition is retained. In some embodiments, patients require intermittent treatment on a long- term basis upon any recurrence of symptoms.
  • the pharmaceutical composition described herein is in unit dosage forms suitable for single administration of precise dosages.
  • the formulation is divided into unit doses containing appropriate quantities of an antibody.
  • the unit dosage is in the form of a package containing discrete quantities of the formulation.
  • Non- limiting examples are packaged tablets or capsules, and powders in vials or ampoules.
  • aqueous suspension compositions are packaged in single-dose non-reclosable containers.
  • multiple-dose reclosable containers are used, in which case it is typical to include a preservative in the composition.
  • formulations for parenteral injection are presented in unit dosage form, which include, but are not limited to ampoules, or in multi dose containers, with an added preservative.
  • the dose of any of the compositions described herein is at least about 50 mg/kg body weight; at least about 20 mg/kg body weight; at least about 10 mg/kg body weight; at least about 5 mg/kg body weight; at least about 3 mg/kg body weight; at least about 2 mg/kg body weight; at least about 1 mg/kg body weight; at least about 750 ⁇ g/kg body weight; at least about 500 ⁇ g/kg body weight; at least about 250 ug/kg body weight; at least about 100 ⁇ g /kg body weight; at least about 50 ⁇ g /kg body weight; at least about 10 ug /kg body weight; at least about 1 ⁇ g/kg body weight, or more, is administered.
  • the dose of any of the compositions described herein is about 25 mg/m , 30 mg/m , 50 mg/m , 60 mg/m , 75 mg/m , 80 mg/m , 90 mg/m , 100 mg/m , 120 mg/m 2 , 160 mg/m 2 , 175 mg/m 2 , 180 mg/m 2 , 200 mg/m 2 , 210 mg/m 2 , 220 mg/m 2 , 250 mg/m 2 , 260 mg/m 2 , 300 mg/m 2 , 350 mg/m 2 , 400 mg/m 2 , 500 mg/m 2 , 540 mg/m 2 , 750 mg/m 2 , 1000 mg/m 2 , or 1080 mg/m 2 of a compound (e.g., modified human C3 proteins).
  • a compound e.g., modified human C3 proteins
  • the composition includes less than about any of 350 mg/m 2 , 300 mg/m 2 , 250 mg/m 2 , 200 mg/m 2 , 150 mg/m 2 , 120 mg/m 2 , 100 mg/m 2 , 90 mg/m 2 , 50 mg/m 2 , or 30 mg/m 2 of a compound (e.g., modified human C3 proteins).
  • a compound e.g., modified human C3 proteins
  • the effective amount of any of the compositions described herein is about 1 to about 5 mg/m 2 , about 5 to about 10 mg/m 2 , about 10 to about 25 mg/m 2 , about 25 to about 50 mg/m 2 , about 50 to about 75 mg/m 2 , about 75 to about 100 mg/m 2 , about 100 to about 125 mg/m , about 125 to about 150 mg/m , about 150 to about 175 mg/m , about 175 to about 200 mg/m 2 , about 200 to about 225 mg/m 2 , about 225 to about 250 mg/m 2 , about 250 to about 300 mg/m , about 300 to about 350 mg/m , or about 350 to about 400 mg/m .
  • Toxicity and therapeutic efficacy of such therapeutic regimens are optionally determined in cell cultures or experimental animals, including, but not limited to, the determination of the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between the toxic and therapeutic effects is the therapeutic index, which is expressed as the ratio between LD50 and ED50.
  • a composition described herein exhibiting high therapeutic indices is preferred.
  • the data obtained from cell culture assays and animal studies are optionally used in formulating a range of dosage for use in human.
  • the dosage of such a composition lies preferably within a range of circulating concentrations that include the ED50 with minimal toxicity.
  • the dosage optionally varies within this range depending upon the dosage form employed and the route of administration utilized.
  • rituximab is administered by slow perfusion at a dose of 375 mg/m body surface area. In some embodiments, rituximab is administered once a week for four weeks. In some embodiments, a complement modulating agent is co-administered with rituximab (e.g., during the administration of rituximab, or within one day of administration of rituximab) for the entire duration of rituximab therapy.
  • daclizumab is administered to an individual one hour before a transplant operation. In some embodiments, daclizumab is administered to an individual every two weeks for 10 weeks after a transplant. In some embodiments, a complement modulating agent is co-administered with daclizumab (e.g., during the administration of daclizumab, or within one day of administration of daclizumab) for the entire duration of daclizumab therapy.
  • omalizumab is administered by subcutaneous injection at a dose of 150 to 375 mg (based on patient's weight and serum IgE level), every 2 or 4 weeks.
  • a complement modulating agent is co-administered with omalizumab (e.g., during the administration of omalizumab, or within one day of administration of omalizumab) for the entire duration of omalizumab therapy.
  • efalizumab is administered by subcutaneous injection at a dose of 0.7 mg/kg. In some embodiments, efalizumab is administered at a dose 1 mg/kg weekly for 12 weeks. In some embodiments, a complement modulating agent is co-administered with efalizumab (e.g., during the administration of efalizumab, or within one day of administration of efalizumab) for the entire duration of efalizumab therapy.
  • infliximab is administered by infusion at a dose of 3 mg/kg (for RA) to 5mg/kg (e.g., for Crohn's Disease).
  • a maintenance dose of infliximab is administered 2 and 6 weeks after the first infusion then every 8 weeks thereafter.
  • infliximab is administered at a dose of up to 10 mg/kg as often as every 4 weeks.
  • a complement modulating agent is co-administered with infliximab (e.g., during the administration of infliximab, or within one day of administration of infliximab) for the entire duration of infliximab therapy.
  • compositions described herein are also optionally used in combination with other therapeutic reagents that are selected for their therapeutic value for the condition to be treated.
  • compositions described herein and, in embodiments where combinational therapy is employed other agents do not have to be administered in the same pharmaceutical composition, and, because of different physical and chemical characteristics, are optionally administered by different routes.
  • the initial administration is optionally made according to established protocols, and then, based upon the observed effects, the dosage, modes of administration and times of administration subsequently modified.
  • compositions as described herein in combination with another therapeutic agent.
  • another therapeutic agent i.e., by itself the adjuvant has minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced.
  • the benefit experienced by a patient is increased by administering a composition described herein with another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit.
  • the overall benefit experienced by the patient is either simply additive of the two therapeutic agents or the patient experiences a synergistic benefit.
  • Therapeutically-effective dosages vary when the drugs are used in treatment combinations. Methods for experimentally determining therapeutically-effective dosages of drugs and other agents for use in combination treatment regimens are documented methodologies. One example of such a method is the use of metronomic dosing, i.e., providing more frequent, lower doses in order to minimize toxic side effects. Combination treatment further includes periodic treatments that start and stop at various times to assist with the clinical management of the patient.
  • the multiple therapeutic agents are administered in any order, or even simultaneously. If simultaneously, the multiple therapeutic agents are optionally provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills).
  • the term "simultaneous administration,” as used herein, means that the composition comprising a complement-depleting compound and the second therapy (e.g., hormone therapy, gene therapy, antibody therapy, and/or chemotherapy) are administered with a time separation of no more than about 15 minute(s), such as no more than about any of 10, 5, or 1 minutes.
  • one of the therapeutic agents is given in multiple doses, or both are given as multiple doses.
  • the timing between the multiple doses optionally varies from more than zero weeks to less than four weeks.
  • the composition comprising a complement-depleting compound and the second therapy e.g., hormone therapy, gene therapy, antibody therapy, and/or chemotherapy
  • sequential administration means that the composition comprising a complement-depleting compound and the second therapy (e.g., hormone therapy, gene therapy, antibody therapy, and/or chemotherapy) are administered with a time separation of more than about 15 minutes, such as more than about any of 20, 30, 40, 50, or 60 minutes, more than about any of 1 day, 2 days, 3 days, 1 week, 2 weeks, or 1 month, or longer.
  • compositions and formulations are not to be limited to the use of only two agents; the use of multiple therapeutic combinations is also envisioned.
  • the dosage regimen to treat, prevent, or ameliorate the condition(s) for which relief is sought is optionally modified in accordance with a variety of factors. These factors include the disorder from which the individual suffers, as well as the age, weight, sex, diet, and medical condition of the individual. Thus, the dosage regimen actually employed varies widely, in some embodiments, and therefore deviates from the dosage regimens set forth herein.
  • the agents which make up the combination therapy disclosed herein are optionally a combined dosage form or in separate dosage forms intended for substantially simultaneous administration.
  • the pharmaceutical agents that make up the combination therapy are optionally also be administered sequentially, with either therapeutic agent being administered by a regimen calling for two-step administration.
  • the two-step administration regimen optionally calls for sequential administration of the active agents or spaced-apart administration of the separate active agents.
  • the time period between the multiple administration steps ranges from, a few minutes to several hours, depending upon the properties of each pharmaceutical agent, such as potency, solubility, bioavailability, plasma half- life and kinetic profile of the pharmaceutical agent. Circadian variation of the target molecule concentrations are optionally used to determine the optimal dose interval.
  • compositions disclosed herein are administered in combination with anti-TNF agent, an IL- 1 receptor antagonist, an IL-2 receptor antagonist, an immunomodulatory agent, an antibiotic, a T-cell co-stimulatory blocker, a disorder-modifying anti-rheumatic agent, a B cell depleting agent, an immunosuppresive agent, an anti-lymphocyte antibody, or combinations thereof.
  • compositions disclosed herein are administered in combination with an anti-inflammatory agent. In some embodiments, the compositions disclosed herein are administered in combination with a chemotherapeutic agent. In some embodiments, the compositions disclosed herein are administered in combination with radiation therapy.
  • compositions disclosed herein are administered in combinations with alefacept, efalizumab, methotrexate, acitretin, isotretinoin, hydroxyurea, mycophenolate mofetil, sulfasalazine, 6-Thioguanine, Dovonex, Taclonex, betamethasone, tazarotene, hydroxychloroquine, sulfasalazine, etanercept, adalimumab, infliximab, abatacept, rituximab, trastuzumab, Anti-CD45 monoclonal antibody AHN- 12 (NCI), Iodine- 131 Anti-Bl Antibody (Corixa Corp.), anti-CD66 monoclonal antibody BW 250/183 (NCI,shire General Hospital), anti-CD45 monoclonal antibody (NCI, Baylor College of Medicine), antibody anti- anb3 integrin (NCI),
  • the complement-depleting compounds (such as modified human C3 proteins) described herein are used to increase the efficiency and/or effectiveness of gene therapy to treat cancer by delivering the compound in an amount sufficient to deplete complement before applying gene therapy treatment to the individual.
  • the complement-depleting compound is administered to a subject before, during, or after a biopsy or a surgery associated with cancer so that the risk of metastasis is reduced.
  • mice Twenty- four female SCID mice weighing 20 to 24 g are used. The animals used were within an age range of 6 to 8 weeks. [00246] The mice are injected intradermally with 100 ag of collagen type-II suspended in complete
  • Example 2 Human Clinical Trial of Inflixumab in combination with HC3-1496 for for Treatment of
  • the primary outcome measurement is: a. To assess the efficacy of inflixumab alone versus inflixumab in combination with INC- 1496 in treatment of recent onset RA according to ACR50 criteria [00251]
  • the secondary outcome measurements are: a. Remission rate after completion of treatment b. Progression of articular damage c. Quality of life d. Toxicity after completion of treatment Treatment Regime
  • control group is administered inflixumab at a dose of 3mg/kg by slow infusion on days 0, 14, 28, 42, 56, 70, 84, 98, 112, 126, 140, 154, and 168.
  • the experimental group is administered inflixumab at a dose of 3mg/kg by slow infusion on days 0, 14, 28, 42, 56, 70, 84, 98, 112, 126, 140, 154, and 168.
  • the experimental group is coadministered HC3- 1496 (15 mg/m 2 ) on days 0, 14, 28, 42, 56, 70, 84, 98, 112, 126, 140, 154, and 168.
  • the primary objective is: a. To compare the ability of natalizumab alone versus natalizumab in combination with HC3- 1496 to induce a clinical response in subjects with moderately to severely active Crohn's disease (CD) and elevated serum C-reactive protein (CRP), as determined by the proportion (%) of subjects with a >70-point decrease from baseline (Week 0) in Crohn's Disease Activity Index (CDAI) score at both Weeks 8 and 12. b. To evaluate the safety and tolerability of natalizumab alone versus natalizumab in combination with HC3-1496 in subjects with moderately to severely active CD and elevated CRP as determined by the occurrence of and/or changes in: i. adverse events; ii.
  • the secondary objective is: a. To compare the ability of natalizumab alone versus natalizumab in combination with HC3- 1496 to induce a clinical remission in subjects with moderately to severely active CD and elevated CRP, as determined by the proportion (%) of subjects with a CDAI score ⁇ 150 at both Weeks 8 and 12. b. To compare the ability of natalizumab alone versus natalizumab in combination with HC3- 1496 to induce a clinical response or remission at Week 12, as determined by: i.
  • the control group receives monthly (defined as a 4-week period) intravenous (IV) infusions of natalizumab 300 mg at Weeks 0, 4, and 8 (IV infusion of 300 mg over approximately 60 minutes at a flow rate of 2 mL/min).
  • the experimental group receives monthly (defined as a 4-week period) intravenous (IV) infusions of natalizumab 300 mg in combination with HC3-1496 (15 mg/m 2 ) at Weeks 0, 4, and 8.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Endocrinology (AREA)
  • Biomedical Technology (AREA)
  • Pulmonology (AREA)
  • Dermatology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Disclosed herein are methods and compositions for modifying an immune system or treating an inflammatory disorder. In some embodiments, the methods and compositions disclosed herein comprise a complement modulating agent. In some embodiments, the methods and compositions further comprise a mAB.

Description

METHOD AND COMPOSITION FOR MODULATING THE IMMUNE SYSTEM AND VARIOUS INFLAMMATORY CONDITIONS COMPRISING COMPLEMENT DEPLETORS
CROSS-REFERENCE
[0001] This application claims the benefit of U.S. Provisional Application No. 61/115,481, filed November 17, 2008 which is herein incorporated by reference in its entirety.
BACKGROUND OF THE INVENTION
[0002] In certain instances, monoclonal antibodies bind to an antigen on a target cell. Following the binding of the mAB, in certain instances, an immune system effector cell (e.g. a NK cell) binds to the antibody and initiates apoptosis of the antibody-bound cell.
SUMMARY OF THE INVENTION
[0003] Disclosed herein are methods and compositions for modulating an immune system or an inflammatory disorder with a C3 complement depletor of various characteristics in its ability to deplete complement via the C3 complement protein, in terms of degree and time of complement depletion beyond variation in the amount given per administration, and without or with the coadministration of a monoclonal antibody, that would increase the efficacy of monoclonal antibody therapies without inducing inflammation.
[0004] Disclosed herein are methods and compositions for increasing the efficacy of monoclonal antibody therapies without inducing inflammation. In some embodiments, the methods and compositions disclosed herein comprise co-administering an antibody and a complement modulating agent. In some embodiments, the complement modulating agent is an agent that depletes the complement cascade without producing (or only minimally producing) C5a. In some embodiments, the complement modulating agent is an agent that inhibits the formation and/or activity of C3b without producing (or only minimally producing) C5a.
[0005] Disclosed herein, in certain embodiments, is a method of treating an inflammatory disorder, comprising co-administering to an individual in need thereof a synergistically-effective amount of (a) an antibody or fusion protein comprising an Fc domain; and (b) a second agent selected from: a complement depleting agent, an agent that inhibits the formation and/or activity of C3b, or a combination thereof; and wherein the production of C5a is not stimulated, or is only minimally stimulated. In some embodiments, the IgG is IgGl, IgG2, IgG3, IgG4, or combinations thereof. In some embodiments, the antibody is a monoclonal antibody, a polyclonal antibody, or a combination thereof. In some embodiments, the antibody is a monoclonal antibody. In some embodiments, the antibody is an anti-CD3 antibody, an anti- CD 1 la§ antibody, an anti-CD-20 antibody, an anti-CD22 antibody, an anti-CD-25 antibody, an anti-CD52 antibody, anti- TNF antibody, anti- IgE antibody, an anti- α4-integrin antibody, or combinations thereof. In some embodiments, the antibody is an anti-CD3 antibody. In some embodiments, the antibody is muromonab-CD3. In some embodiments, the antibody is an anti- CDl la§ antibody. In some embodiments, the antibody is efalizumab. In some embodiments, the antibody is an anti-CD20 antibody. In some embodiments, the antibody is GAlOl (R7159, Genentech), ocrelizumab (PRO70769), ofatumumab (HuMax-CD20 human IgGl monoclonal antibody or 2F2), PRO 131921 (Genentech), rituximab, veltuzumab (IMMU- 106 or hA20), or combinations thereof. In some embodiments, the antibody is an anti-CD22 antibody. In some embodiments, the antibody is epratuzumab. In some embodiments, the antibody is an anti-CD25 antibody. In some embodiments, the antibody is daclizumab, basiliximab, or a combination thereof. In some embodiments, the antibody is an anti-CD52 antibody. In some embodiments, the antibody is alemtuzumab. In some embodiments, the antibody is an anti- TNF antibody. In some embodiments, the antibody is infliximab, adalimumab, or a combination thereof. In some embodiments, the antibody is an anti- IgE antibody. In some embodiments, the antibody is omalizumab. In some embodiments, the antibody is an anti- α4-integrin antibody. In some embodiments, the antibody is natalizumab. In some embodiments, the complement depleting agent is GR-2II, AGIIa, AGIIb-I, AR-2IIa, AR-2IIb, AR-2IIc, AR-2IId, CVF, a hybrid human C3-CVF protein, rC3, HC3-1496, HC3-1496-2, HC3-1496-3, HC3-1496-4, HC3-1496/1617, HC3-1496-8, HC3-1496-9, HC3-1496-10, HC3-1496-11, HC3-1496-12, HC3-1496-13, HC3- 1496-14, HC3-1496-15, HC3-1496-16, HC3-1496-17, or combinations thereof. In some embodiments, the agent that inhibits the formation and/or activity of C3b is complement receptor 1, sCRl, APT070, TPlO, TP20, sCRl [desLHR-A]), sCRl-SLex, Crry, Crry-Ig, fucan, BS8, complestatin, Ecb, Efb, compstatin, rosmarinic acid, CRIT, CRIT-H17, glycyrrhetinic acid, CD55, sCD55, a CD55/MCP fusion protein, BCX- 1470, FUT- 175, Factor I, MCP, sMCP, heparin, an anti-properdin antibody, or combinations thereof. In some embodiments, the inflammatory disorder is Atopic dermatitis; Acute disseminated encephalomyelitis; Acute leukocyte -mediated lung injury; Addison's disease; AIDS dementia; Allergic rhinitis; Ankylosing spondylitis; Antiphospholipid antibody syndrome; Alzheimer's disorder; Asthma; Adult respiratory distress syndrome; Autoimmune hemolytic anemia; Autoimmune hepatitis; Autoimmune inner ear disease; Behcet's syndrome; Bronchitis; Bullous pemphigoid; Chagas disease; Chronic obstructive pulmonary disease; Coagulative Necrosis; Coeliac disease; Crohn's disorder; Collagenous colitis; Conjunctivitis; Cystic fibrosis; Dermatomyositis; Dermatitis; Diabetes mellitus type 1; Diabetes mellitus type 2; Distal proctitis; Diversion colitis; Encephalitis; Endometriosis; Endotoxin shock; Eczema; Epilepsy; Fibromyalgia; Fibrinoid Necrosis; Goodpasture's syndrome; Gouty arthritis; Graves' disease; Guillain-Barre syndrome; Hashimoto's disease; Idiopathic thrombocytopenic purpura; Indeterminate colitis; Infective colitis; Inflammatory liver disorder; Interstitial cystitis; Ischaemic colitis; Liquefactive Necrosis; Lymphocytic colitis; Meningitis; Multiple sclerosis; Myasthenia gravis; Myocarditis; Narcolepsy; Nephritis; Pancreatitis; Parkinson's disorder; Pemphigus Vulgaris; Periodontal gingivitis; Pernicious anaemia; Polymyositis; Polymyalgia rheumatica; Psoriasis; Primary biliary cirrhosis; Retinitis; Rheumatoid arthritis; Rheumatoid spondylitis; Schizophrenia; Scleroderma; Shingles; Sjogren's syndrome; Smooth muscle proliferation disorders; Systemic lupus erythematosus (SLE); Tuberculosis; Ulcerative colitis; Uveitis; Vasculitis; Vitiligo; Wegener's granulomatosis; or combinations thereof.
[0006] Disclosed herein, in certain embodiments, is a method of enhancing an immune response against a pathogen, comprising co-administering to an individual in need thereof a synergistically-effective amount of (a) an antibody or fusion protein comprising an Fc domain; and (b) a second agent selected from: a complement depleting agent, an agent that inhibits the formation and/or activity of C3b, or a combination thereof; and wherein the production of C5a is not stimulated, or is only minimally stimulated. In some embodiments, the pathogen is a bacterium, a virus, a fungus, a prion, a protozoan, or combinations thereof. In some embodiments, the pathogen is RSV, Vibrio cholerae, Plasmodium falciparum, Plasmodium vivax, Plasmodium ovale, Plasmodium malariae, Mycobacterium tuberculosis, a influenza, HIV, E. coli O157:H7, a hepatitis virus, Poliovirus, Mycobacterium leprae, Yersinia pestis, Rickettsia prowazekii, Borrelia burgdorferi, Borrelia afzelii, Borrelia garinii, or combinations thereof. In some embodiments, the IgG is IgGl, IgG2, IgG3, IgG4, or combinations thereof. In some embodiments, the antibody is a monoclonal antibody, a polyclonal antibody, or a combination thereof. In some embodiments, the antibody is a monoclonal antibody. In some embodiments, the antibody is an anti- RSV gpF antibody. In some embodiments, the antibody is palivizumab. In some embodiments, the antibody is palivizumab. In some embodiments, the antibody is an anti- Pfs25 (Plasmodium falciparum) antibody, an anti- Pfs28 (Plasmodium falciparum), or combinations thereof. In some embodiments, the antibody is an anti- phenolic glycolipid TB (PGL-Tb) antibody. In some embodiments, the antibody is an anti- influenza matrix 2 protein (M2e) antibody. In some embodiments, the antibody is an anti- Y. pestis capsular Fl (Cafl) antibody.
[0007] In some embodiments, the complement depleting agent is GR-2II, AGIIa, AGIIb- 1 , AR-2IIa, AR-2IIb, AR-2IIc, AR-2IH, CVF, a hybrid human C3-CVF protein, rC3, HC3-1496, HC3- 1496-2, HC3-1496-3, HC3-1496-4, HC3-1496/1617, HC3-1496-8, HC3-1496-9, HC3-1496-10, HC3-1496-11, HC3-1496-12, HC3-1496-13, HC3-1496-14, HC3-1496-15, HC3-1496-16, HC3- 1496-17, or combinations thereof. In some embodiments, the agent that inhibits the formation and/or activity of C3b is complement receptor 1, sCRl, APT070, TPlO, TP20, sCRl [desLHR- A]), sCRl-SLex, Crry, Crry-Ig, fucan, BS8, complestatin, Ecb, Efb, compstatin, rosmarinic acid, CRIT, CRIT-H17, glycyrrhetinic acid, CD55, sCD55, a CD55/MCP fusion protein, BCX- 1470, FUT- 175, Factor I, MCP, sMCP, heparin, an anti-properdin antibody, or combinations thereof.
[0008] Disclosed herein, in certain embodiments, is a modified human complement C3 protein comprising a human C3 protein, wherein one or more amino acid residues in the human C3 protein are substituted with a corresponding portion of a CVF protein; and wherein the protein does not comprise C3a. In some embodiments the amino acid residues in the human C3 protein corresponding to amino acid residues 1496 to 1663 of SEQ ID NO :1 are substituted with a corresponding portion of a CVF protein. In some embodiments, the protein is at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, or at least about 95% homologous with a human C3 sequence. In some embodiments, one or more amino acid residues in the CVF portion is modified. In some embodiments, one or more amino acid residues in the CVF portion is modified, and the modification is selected from the group consisting of: (a) T1499D and L1501K; (b) I1507R, G1508D, N1509E, and V1510L; (c) S1519F, S1520I, L1521Q, N1522K, H1523S, and Q1524D; (d) D1528T, V1529L, P1530E, L1531E, and Q1532R; (e) 1519-1550 replaced with corresponding amino acid residues of SEQ ID NO: 1; (f) Q 157 IS, T1573S, N1576V, P1577Q, and R1578V; (g) 1596-1617 replaced with corresponding amino acid residues of SEQ ID NO: 1 ; (h) 1596- 1611 replaced with corresponding amino acid residues of SEQ ID NO:l; (i) V1598L, N1599D, D1600N, S1607L, and R1608S; (j) V1598L, N1599D, and D 1600N; (k) S1607L, and R1608S, or combinations thereof. In some embodiments, an amino acid of human C3 is substituted or deleted in a domain selected from: MGl, MG2, MG6, alpha-NT', TED, MG7, or combinations thereof. In some embodiments, Glutamine (Q) 163 of human C3 is substituted. In some embodiments, Glutamine (Q) 163 of human C3 is deleted. In some embodiments, Glutamine (Q) 163 of human C3 is deleted, the amino acid residue immediately preceding Glutamine 163 is deleted, and the three amino acid residues immediately following Glutamine 163 are deleted. In some embodiments, Serine (S) 1075 of human C3 is substituted. In some embodiments, Serine (S) 1075 of human C3 is substituted with Histidine (H). In some embodiments, Serine (S) 1075 of human C3 is substituted with Histidine (H) and the two preceding amino acid residues are deleted. In some embodiments, Asparagine (N) 738 of human C3 is substituted. In some embodiments, Asparagine (N) 738 of human C3 is substituted with Aspartic Acid. In some embodiments, Asparagine (N) 738 of human C3 is substituted with Aspartic Acid and the two preceding Glutamic acid residues (EE) are substituted with aspartic acid and serine (DS). In some embodiments, Asparagine (N) 738 of human C3 is substituted with Aspartic acid and the immediately preceding Glutamic acid (E) is substituted with Serine (S). In some embodiments, the Glutamine (Q) at position 1139 of human C3 is substituted with a Lysine (K) or a Tyrosine (Y).
BRIEF DESCRIPTION OF THE DRAWINGS The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which: [0010] FIG. 1 depicts the chain structures of human C3 and CVF with shaded portions present in the mature proteins. [0011] FIGS. 2A-2D provide an alignment between prepro-CVF (SEQ ID NO:2) from Naja naja
Cobra and human prepro-C3 (SEQ ID NO: 1) amino acid sequences showing the corresponding regions of the two proteins. [0012] FIGS. 3A-3H show nucleic acid and amino acid sequence of human prepro-C3. The NH2- and C-termini of the α- and β-chains, functionally important regions, and ligand binding sites are indicated. Amino acid residue numbering starts at the NH2 -terminus of the prepro-C3 molecule. [0013] FIG. 4 shows survival curves of mice treated with CVF with or without monoclonal antibody
(mAb). [0014] FIG. 5 shows survival curves of mice treated with HC3- 1496 with or without mAb.
DETAILED DESCRIPTION OF THE INVENTION
[0015] Disclosed herein are methods and compositions for modulating an immune system or an inflammatory disorder with a C3 complement depletor of various characteristics in its ability to deplete complement via the C3 complement protein, in terms of degree and time of complement depletion beyond variation in the amount given per administration, and without or with the coadministration of a monoclonal antibody, that would increase the efficacy of monoclonal antibody therapies without inducing inflammation.
[0016] In certain instances, depletion of complement C3 protein modulates the innate immune system that includes the complement cascade as well as the immune system as a whole. Complement C3 is the center piece that enables further complement activation from the three known key pathways that initially activate the complement cascade: (1) the classical pathway, (2) the alternative pathway, and (3) mannose binding lectin pathway. In certain instances, the modulation of the innate immune system by depletion of complement C3 may alone alter the dynamics of the entire immune system including the adaptive immune system and also affect the interactions among the circulating blood cells and their secreted products as well as therapeutics such as monoclonal antibodies that is introduced into the circulatory system. When monoclonal antibodies are introduced into circulation, the antibodies bind to certain antigens on a target cell. In certain instances, an immune system effector cell (e.g. a NK cell) binds to the antibody and initiates apoptosis of the antibody-bound cell. In addition, complement depletion through C3 depletion alters the interaction among immune system and inflammatory proteins in vivo (e.g., within any organism) as well as ex vivo (e.g., within blood products that in certain instances contain activatable complement proteins). tain Definitions
[0017] The term "leukocyte" comprises, by way of non- limiting example, lymphocytes, monocytes, macrophages, eosinophils, neutrophils and basophils. Leukocytes include, but are not limited to, to hematopoietic stem cells and all myeloid and lymphoid lineages that arise from hematopoietic stem cells. Leukocytes further include, but are not limited to, all immature, mature, undifferentiated and differentiated white blood cell populations including tissue specific and specialized varieties.
[0018] The term "lymphocyte" encompasses, by way of non- limiting example, B-cells, T-cells, NKT cells, and NK cells. In some embodiments lymphocytes refers to all immature, mature, undifferentiated and differentiated white lymphocyte populations including tissue specific and specialized varieties. Lymphocytes include all B-cell lineages including pre-B-cells, Progenitor B cells, Early Pro-B cells, Late Pro-B cells, Large Pre-B cells, Small Pre-B cells, Immature B cells, Mature B cells, plasma B-cells, memory B-cells, B-I cells, B-2 cells and anergic AN1/T3 cell populations.
[0019] The term "B-cell", refers to, by way of non- limiting example, a pre-B-cell, Progenitor B cell, Early Pro-B cell, Late Pro-B cell, Large Pre-B cell, Small Pre-B cell, Immature B cell, Mature B cell, plasma B-cell, memory B-cell, B-I cell, B-2 cells and anergic AN1/T3 cell populations. The term B-cell includes a B-cell that expresses an immunoglobulin heavy chain and/or light chain on its cells surface. Further, the term B-cell includes a B-cell that expresses and secretes an immunoglobulin heavy chain and/or light chain. The term B-cell includes a cell that binds an antigen on its cell-surface. In some embodiments disclosed herein, B-cells or AN1/T3 cells are utilized in the processes described. In certain embodiments, such cells are optionally substituted with any animal cell suitable for expressing, capable of expressing (e.g., inducible expression), or capable of being differentiated into a cell suitable for expressing an antibody including, e.g., a hematopoietic stem cell, a B-cell, a pre-B-cell, a Progenitor B cell, a Early Pro-B cell, a Late Pro-B cell, a Large Pre-B cell, a Small Pre-B cell, an Immature B cell, a Mature B cell, a plasma B-cell, a memory B-cell, a B-I cell, a B-2 cell, an anergic B-cell, or an anergic AN1/T3 cell.
[0020] The term "immunized" refers to the introduction of an antigen into an animal by any route. Non- limiting examples of various routes are by way of intradermal injection, intravenous injection, intraocular administration, subcutaneous injection, intraperitoneal injection, oral administration, or topical administration.
[0021] The term "antigen" refers to a substance that is capable of inducing the production of an antibody. Further, antigen refers to a substance that binds to an antibody variable region.
[0022] The terms "antibody" and "antibodies" refer to monoclonal antibodies, polyclonal antibodies, bi-specific antibodies, multispecific antibodies, grafted antibodies, human antibodies, humanized antibodies, synthetic antibodies, chimeric antibodies, camelized antibodies, single- chain Fvs (scFv), single chain antibodies, Fab fragments, F(ab') fragments, disulfide-linked Fvs (sdFv), intrabodies, and anti-idiotypic (anti-Id) antibodies and antigen-binding fragments of any of the above. In particular, antibodies include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules, i.e., molecules that contain an antigen binding site. Immunoglobulin molecules are of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG i; IgG 2, IgG 3, IgG A, IgA 1 and IgA 2 ) or subclass. The terms "antibody" and "immunoglobulin" are used interchangeably in the broadest sense. In some embodiments, an antibody is part of a larger molecule, formed by covalent or non-covalent association of the antibody with one or more other proteins or peptides.
[0023] The term "derivative" in the context of a polypeptide or protein, e.g. an antibody, refers to a polypeptide or protein that comprises an amino acid sequence which has been altered by the introduction of amino acid residue substitutions, deletions or additions. The term "derivative" also refers to a polypeptide or protein which has been modified, i.e., by the covalent attachment of any type of molecule to the antibody. For example, in some embodiments a polypeptide or protein is modified, e.g., by glycosylation, acetylation, pegylation, phosphorylation, amidation, derivatization by protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc. In some embodiments, derivatives, polypeptides or proteins are produced by chemical modifications using suitable techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, metabolic synthesis of tunicamycin, etc. In some embodiments a derivative a polypeptide or protein possesses a similar or identical function as the polypeptide or protein from which it was derived.
[0024] The terms "full length antibody", "intact antibody" and "whole antibody" are used herein interchangeably, to refer to an antibody in its substantially intact form, and not antibody fragments as defined below. These terms particularly refer to an antibody with heavy chains contains Fc regions. In some embodiments, an antibody variant provided herein is a full length antibody. In some embodiments, the full length antibody is human, humanized, chimeric, and/or affinity matured.
[0025] An "affinity matured" antibody is one having one or more alteration in one or more CDRs thereof which result in an improvement in the affinity of the antibody for antigen, compared to a parent antibody which does not possess those alteration(s). Preferred affinity matured antibodies will have nanomolar or even picomolar affinities for the target antigen. Affinity matured antibodies are produced by suitable procedures. See, for example, Marks et al., (1992) Biotechnology 10:779-783 that describes affinity maturation by variable heavy chain (VH) and variable light chain (VL) domain shuffling. Random mutagenesis of CDR and/or framework residues is described in: Barbas, et al. (1994) Proc. Nat. Acad. Sci, USA 91 :3809-3813; Shier et al., (1995) Gene 169:147-155; Yelton et al., 1995, J. Immunol. 155:1994-2004; Jackson et al., 1995, J. Immunol. !54(7):3310-9; and Hawkins et al, (19920, J. MoI. Biol. 226:889-896, for example.
[0026] The terms "binding fragment", "antibody fragment" or "antigen binding fragment" are used herein, for purposes of the specification and claims, to mean a portion or fragment of an intact antibody molecule, preferably wherein the fragment retains antigen-binding function. Examples of antibody fragments include Fab, Fab', F(ab')2, Fd, Fd' and Fv fragments, diabodies, linear antibodies (Zapata et al. (1995) Protein Eng. 10: 1057), single-chain antibody molecules, single-chain binding polypeptides, scFv, bivalent scFv, tetravalent scFv, and bispecific or multispecific antibodies formed from antibody fragments.
[0027] "Fab" fragments are typically produced by papain digestion of antibodies resulting in the production of two identical antigen-binding fragments, each with a single antigen-binding site and a residual "Fc" fragment. Pepsin treatment yields a F(ab')2 fragment that has two antigen- combining sites capable of cross-linking antigen. An "Fv" is the minimum antibody fragment that contains a complete antigen recognition and binding site. In a two-chain Fv species, this region consists of a dimer of one heavy- and one light-chain variable domain in tight, non- covalent association. In a single-chain Fv (scFv) species, one heavy- and one light-chain variable domain are covalently linked by a flexible peptide linker such that the light and heavy chains associate in a "dimeric" structure analogous to that in a two-chain Fv species. It is in this configuration that the three CDRs of each variable domain interact to define an antigen-binding site on the surface of the VH — VL dimer. Collectively, the six CDRs confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although usually at a lower affinity than the entire binding site.
[0028] The Fab fragment also contains the constant domain of the light chain and the first constant domain (CH1) of the heavy chain. Fab fragments differ from Fab' fragments by the addition of a few residues at the carboxy terminus of the heavy-chain CH1 domain including one or more cysteines from the antibody hinge region. Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear a free thiol group. F(ab')2 antibody fragments originally were produced as pairs of Fab' fragments that have hinge cysteines between them. Other chemical couplings of antibody fragments are also suitable. Methods for producing the various fragments from monoclonal Abs include, e.g., Pluckthun, 1992, Immunol. Rev. 130: 152-188.
[0029] The terms "hypervariable region" and "CDR" when used herein, refer to the amino acid residues of an antibody which are responsible for antigen-binding. The CDRs comprise amino acid residues from three sequence regions which bind in a complementary manner to an antigen and are known as CDRl, CDR2, and CDR3 for each of the VH and VL chains. In the light chain variable domain, the CDRs typically correspond to approximately residues 24-34 (CDRLl), 50-56 (CDRL2) and 89-97 (CDRL3), and in the heavy chain variable domain the CDRs typically correspond to approximately residues 31-35 (CDRHl), 50-65 (CDRH2) and 95- 102 (CDRH3) according to Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)). It is understood that the CDRs of different antibodies may contain insertions, thus the amino acid numbering may differ. The Kabat numbering system accounts for such insertions with a numbering scheme that utilizes letters attached to specific residues (e.g., 27A, 27B, 27C, 27D, 27E, and 27F of CDRLl in the light chain) to reflect any insertions in the numberings between different antibodies. Alternatively, in the light chain variable domain, the CDRs typically correspond to approximately residues 26-32 (CDRLl), 50-52 (CDRL2) and 91-96 (CDRL3), and in the heavy chain variable domain, the CDRs typically correspond to approximately residues 26-32 (CDRHl), 53-55 (CDRH2) and 96-101 (CDRH3) according to Chothia and Lesk, J. MoI. Biol., 196: 901-917 (1987)).
[0030] The term "monoclonal antibody" refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that are present in minor amounts. In some embodiments monoclonal antibodies are made, for example, by the hybridoma method first described by Kδhler and Milstein (1975) Nature 256:495, or are made by recombinant methods, e.g., as described in U.S. Pat. No. 4,816,567. In some embodiments monoclonal antibodies are isolated from phage antibody libraries using the techniques described in Clackson et al, Nature 352:624-628 (1991), as well as in Marks et al, J. MoI. Biol. 222:581- 597 (1991).
[0031] The antibodies herein include monoclonal, polyclonal, recombinant, chimeric, humanized, bi- specific, grafted, human, and fragments thereof including antibodies altered by any means to be less immunogenic in humans. Thus, for example, the monoclonal antibodies and fragments, etc., herein include "chimeric" antibodies and "humanized" antibodies. In general, chimeric antibodies include a portion of the heavy and/or light chain that is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, so long as they exhibit the desired biological activity (U.S. Pat. No. 4,816,567); Morrison et al Proc. Natl Acad. ScL 81:6851-6855 (1984). For example in some embodiments a chimeric antibody contains variable regions derived from a mouse and constant regions derived from human in which the constant region contains sequences homologous to both human IgG2 and human IgG4. "Humanized" forms of non- human (e.g., murine) antibodies or fragments are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab')2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. Humanized antibodies include, grafted antibodies or CDR grafted antibodies wherein part or all of the amino acid sequence of one or more complementarity determining regions (CDRs) derived from a non-human animal antibody is grafted to an appropriate position of a human antibody while maintaining the desired binding specificity and/or affinity of the original non-human antibody. In some embodiments, corresponding non-human residues replace Fv framework residues of the human immunoglobulin. In some embodiments humanized antibodies comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications are made to further refine and optimize antibody performance. In some embodiments, the humanized antibody comprises substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. For further details, see, e.g.: Jones et al., Nature 321: 522-525 (1986); Reichmann et al., Nature 332: 323-329 (1988) and Presta, Curr. Op. Struct. Biol. 2: 593-596 (1992).
[0032] A humanized antibody also includes antibodies in which part, or all of the CDRs of the heavy and light chain are derived from a non-human monoclonal antibody, substantially all the remaining portions of the variable regions are derived from human variable region (both heavy and light chain), and the constant regions are derived from a human constant region. In one embodiment, the CDRl, CDR2 and CDR3 regions of the heavy and light chains are derived from a non-human antibody. In yet another embodiment, at least one CDR (e.g., a CDR3) of the heavy and light chains is derived from a non- human antibody. Various combinations of CDRl, CDR2, and CDR3 can be derived from a non-human antibody and are contemplated herein.
[0033] The phrase "specifically binds" when referring to the interaction between an antibody or other binding molecule and a protein or polypeptide or epitope, typically refers to an antibody or other binding molecule that recognizes and detectably binds with high affinity to the target of interest. Preferably, under designated or physiological conditions, the specified antibodies or binding molecules bind to a particular polypeptide, protein or epitope yet does not bind in a significant or undesirable amount to other molecules present in a sample. In other words, the specified antibody or binding molecule does not undesirably cross-react with non-target antigens and/or epitopes. A variety of immunoassay formats are used to select antibodies or other binding molecule that are immunoreactive with a particular polypeptide and have a desired specificity. For example, solid-phase ELISA immunoassays, BIAcore, flow cytometry and radioimmunoassays are used to select monoclonal antibodies having a desired immunoreactivity and specificity. See, Harlow, 1988, ANTIBODIES, A LABORATORY MANUAL, Cold Spring Harbor Publications, New York (hereinafter, "Harlow"), for a description of immunoassay formats and conditions that are used to determine or assess immunoreactivity and specificity.
[0034] "Selective binding", "selectivity", and the like refer the preference of an antibody to interact with one molecule as compared to another. Preferably, interactions between antibodies, particularly modulators, and proteins are both specific and selective. Note that in some embodiments an antibody is designed to "specifically bind" and "selectively bind" two distinct, yet similar targets without binding to other undesirable targets.
[0035] The terms "polypeptide", peptide" and "protein" are used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to naturally occurring amino acid polymers as well as amino acid polymers in which one or more amino acid residues is a non-naturally occurring amino acid, e.g., an amino acid analog. The terms encompass amino acid chains of any length, including full length proteins (i.e., antigens), wherein the amino acid residues are linked by covalent peptide bonds.
[0036] The terms "isolated" and "purified" refer to a material that is substantially or essentially removed from or concentrated in its natural environment. For example, an isolated nucleic acid is one that is separated from at least some of the nucleic acids that normally flank it or other nucleic acids or components (proteins, lipids, etc..) in a sample. In another example, a polypeptide is purified if it is substantially removed from or concentrated in its natural environment. Methods for purification and isolation of nucleic acids and proteins are documented methodologies. Embodiments of "substantially" include at least 20%, at least 40%, at least 50%, at least 75%, at least 85%, at least 90%, at least 95%, or at least 99%.
[0037] The terms "individual," "subject," or "patient" are used interchangeably. As used herein, they mean any mammal (i.e. species of any orders, families, and genus within the taxonomic classification animalia: chordata: vertebrata: mammalia). In some embodiments, the mammal is a human. In some embodiments, the mammal is a non-human. In some embodiments, the mammal is a member of the taxonomic orders: primates (e.g. lemurs, lorids, galagos, tarsiers, monkeys, apes, and humans); rodentia (e.g. mice, rats, squirrels, chipmunks, and gophers); lagomorpha (e.g. hares, rabbits, and pika); erinaceomorpha (e.g. hedgehogs and gymnures); soricomorpha (e.g. shrews, moles, and solenodons); chiroptera (e.g., bats); cetacea (e.g. whales, dolphins, and porpoises); carnivora (e.g. cats, lions, and other feliformia; dogs, bears, weasels, and seals); perissodactyla (e.g. horse, zebra, tapir, and rhinoceros); artiodactyla (e.g. pigs, camels, cattle, and deer); proboscidea (e.g. elephants); sirenia (e.g. manatees, dugong, and sea cows); cingulata (e.g. armadillos); pilosa (e.g. anteaters and sloths); didelphimorphia (e.g. american opossums); paucituberculata (e.g. shrew opossums); microbiotheria (e.g. Monito del Monte); notoryctemorphia (e.g. marsupial moles); dasyuromorphia (e.g. marsupial carnivores); peramelemorphia (e.g. bandicoots and bilbies); or diprotodontia (e.g. wombats, koalas, possums, gliders, kangaroos, wallaroos, and wallabies). In some embodiments, the animal is a reptile (i.e. species of any orders, families, and genus within the taxonomic classification animalia: chordata: vertebrata: reptilia). In some embodiments, the animal is a bird (i.e. animalia: chordata: vertebrata: aves). None of the terms require or are limited to situation characterized by the supervision (e.g. constant or intermittent) of a health care worker (e.g. a doctor, a registered nurse, a nurse practitioner, a physician's assistant, an orderly, or a hospice worker).
[0038] The terms "treat," "treating" or "treatment," and other grammatical equivalents as used herein, include alleviating, inhibiting or reducing symptoms, reducing or inhibiting severity of, reducing incidence of, prophylactic treatment of, reducing or inhibiting recurrence of, preventing, delaying onset of, delaying recurrence of, abating or ameliorating a disease or condition symptoms, ameliorating the underlying metabolic causes of symptoms, inhibiting the disease or condition, e.g., arresting the development of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a condition caused by the disease or condition, or stopping the symptoms of the disease or condition. The terms further include achieving a therapeutic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated, and/or the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the individual.
[0039] The terms "prevent," "preventing" or "prevention," and other grammatical equivalents as used herein, include preventing additional symptoms, preventing the underlying metabolic causes of symptoms, inhibiting the disease or condition, e.g., arresting the development of the disease or condition and are intended to include prophylaxis. The terms further include achieving a prophylactic benefit. For prophylactic benefit, the compositions are optionally administered to an individual at risk of developing a particular disease, to an individual reporting one or more of the physiological symptoms of a disease, or to an individual at risk of reoccurrence of the disease.
[0040] Where combination treatments or prevention methods are contemplated, it is not intended that the agents described herein be limited by the particular nature of the combination. For example, the agents described herein are optionally administered in combination as simple mixtures as well as chemical hybrids. An example of the latter is where the agent is covalently linked to a targeting carrier or to an active pharmaceutical. Covalent binding can be accomplished in many ways, such as, though not limited to, the use of a commercially available cross-linking agent. Furthermore, combination treatments are optionally administered separately or concomitantly.
[0041] As used herein, the terms "pharmaceutical combination", "administering an additional therapy", "administering an additional therapeutic agent" and the like refer to a pharmaceutical therapy resulting from the mixing or combining of more than one active ingredient and includes both fixed and non- fixed combinations of the active ingredients. The term "fixed combination" means that at least one of the agents described herein, and at least one co-agent, are both administered to an individual simultaneously in the form of a single entity or dosage. The term "non- fixed combination" means that at least one of the agents described herein, and at least one co-agent, are administered to an individual as separate entities either simultaneously, concurrently or sequentially with variable intervening time limits, wherein such administration provides effective levels of the two or more agents in the body of the individual. In some instances, the co-agent is administered once or for a period of time, after which the agent is administered once or over a period of time. In other instances, the co-agent is administered for a period of time, after which, a therapy involving the administration of both the co-agent and the agent are administered. In still other embodiments, the agent is administered once or over a period of time, after which, the co-agent is administered once or over a period of time. These also apply to cocktail therapies, e.g. the administration of three or more active ingredients.
[0042] As used herein, the terms "co-administration", "administered in combination with" and their grammatical equivalents are meant to encompass administration of the selected therapeutic agents to a single individual, and are intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different times. In some embodiments the agents described herein will be co-administered with other agents. These terms encompass administration of two or more agents to an animal so that both agents and/or their metabolites are present in the animal at the same time. They include simultaneous administration in separate compositions, administration at different times in separate compositions, and/or administration in a composition in which both agents are present. Thus, in some embodiments, the agents described herein and the other agent(s) are administered in a single composition. In some embodiments, the agents described herein and the other agent(s) are admixed in the composition.
[0043] The terms "effective amount" or "therapeutically effective amount" as used herein, refer to a sufficient amount of at least one agent being administered which achieve a desired result, e.g., to relieve to some extent one or more symptoms of a disease or condition being treated. In certain instances, the result is a reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. In specific instances, the result is a decrease in the growth of, the killing of, or the inducing of apoptosis in at least one abnormally proliferating cell, e.g., a cancer cell or a cancer stem cell. In certain instances, an "effective amount" for therapeutic uses is the amount of the composition comprising an agent as set forth herein required to provide a clinically significant decrease in a disease. An appropriate "effective" amount in any individual case is determined using any suitable technique, such as a dose escalation study. [0044] The terms "administer," "administering", "administration," and the like, as used herein, refer to the methods that are used to enable delivery of agents or compositions to the desired site of biological action. These methods include, but are not limited to oral routes, intraduodenal routes, parenteral injection (including intravenous, subcutaneous, intraperitoneal, intramuscular, intravascular or infusion), topical and rectal administration. Administration techniques that are optionally employed with the agents and methods described herein, include e.g., as discussed in Goodman and Gilman, The Pharmacological Basis of Therapeutics, current ed.; Pergamon; and Remington's, Pharmaceutical Sciences (current edition), Mack Publishing Co., Easton, Pa. In certain embodiments, the agents and compositions described herein are administered orally.
[0045] The term "pharmaceutically acceptable" as used herein, refers to a material that does not abrogate the biological activity or properties of the agents described herein, and is relatively nontoxic (i.e., the toxicity of the material significantly outweighs the benefit of the material). In some instances, a pharmaceutically acceptable material is administered to an individual without causing significant undesirable biological effects or significantly interacting in a deleterious manner with any of the components of the composition in which it is contained.
[0046] The term "carrier" as used herein, refers to relatively nontoxic chemical agents that, in certain instances, facilitate the incorporation of an agent into cells or tissues. noclonal Antibody Therapy and ADCC
[0047] Disclosed herein are methods and compositions for increasing the efficacy of monoclonal antibody therapies without inducing inflammation. Further disclosed herein, in certain instances, is a method of treating an immune disorder. In some embodiments, the methods described herein comprise co-administering an antibody and a complement modulating agent.
[0048] In some embodiments, the antibody is a monoclonal antibody, a polyclonal antibody, or a combination thereof. In some embodiments, the antibody is a monoclonal antibody.
[0049] In some embodiments, the antibody is a murine antibody. In some embodiments, the antibody is a rabbit antibody. In some embodiments, the antibody is a chimeric antibody. In some embodiments, the antibody is a humanized antibody. In some embodiments, the antibody comprises a murine variable region and a human Fc region.
[0050] In some embodiments, the antibody is conjugated to a cytotoxic agent. In some embodiments, the antibody is conjugated to a chemotherapeutic agent. In some embodiments, the antibody is conjugated to a drug-activating enzyme. In certain instances, the drug-activating enzyme converts a non-toxic agent into a toxic agent. In some embodiments, the antibody is conjugated to a liposome. In some embodiments, the antibody is covalently bound to a radioactive isotope
(e.g., I131)- [0051] In certain instances, the antibody binds to a specific antigen (e.g., CD20; CD25, IgE, TNF, flagella, fimbrae, LPS, toxins) differentially expressed and/or overexpressed on a target cell. In certain instances, the binding of the antibody to the antigen results (either partially or fully) in an immune response against the cell.
[0052] In some embodiments, the antibody comprises at least a portion of an Immunoglobulin G
(IgG) antibody. In some embodiments, the IgG antibody is IgGl, IgG2, IgG3, or IgG4. In some embodiments, the antibody is an IgGl antibody. In some embodiments, the antibody is an IgG2 antibody. In some embodiments, the antibody comprises at least a portion of an IgGl or IgG2Fc region. In certain instances, the Fc portion of an IgGl or IgG2 antibody interacts with (e.g., binds to) cell surface receptors called Fc receptors. In certain instances, an Fc receptor is expressed on NK cells, macrophages, neutrophils, and mast cells.
[0053] In some embodiments, the methods described herein induce ADCC by natural killer (NK) cells or related cellular killing mechanisms. In certain instances, the binding of the Fc region of an antibody to an Fc receptor on a NK cell (i.e., CD 16/FcγRIII) results in (either partially or fully) the NK cell releasing cytotoxic molecules (IFN-γ) and cytotoxic granules containing perforin and proteases. In certain instances, perforin forms pores in the membrane of an antigen presenting cell. In certain instances, the cytotoxic molecules and proteases enter the antigen presenting cell and induce apoptosis. This process is called antibody dependent cell-mediated cytotoxicity (ADCC).
[0054] In some embodiments, the methods described herein are used to treat an immune-mediated inflammatory disorder (e.g., by administering an antibody to an antigen expressed on a leukocyte). In certain instances, an immune -mediated inflammatory disorder results from (either partially or fully) a derangement of an immune system whereby the immune system (e.g., via leukocytes) attacks self antigens. In some embodiments, the methods and compositions disclosed herein treat an immune-mediated inflammatory disorder by initiating an immune response (e.g., ADCC or related cellular killing mechanisms) against leukocytes.
[0055] In some embodiments, the methods described herein are used to treat an infectious disorder (e.g., by administering an antibody to an antigen expressed on a pathogen). In some embodiments, the methods and compositions disclosed herein treat an infectious disorder by initiating an immune response (e.g., ADCC or related cellular killing mechanisms) against an infectious agent.
III. The Complement System
[0056] Disclosed herein are methods and compositions for increasing the efficacy of monoclonal antibody therapies without inducing inflammation. Further disclosed herein, in certain instances, is a method of activating an immune system, a method of inducing apoptosis in a cell, and a method of treating a neoplasia. In some embodiments, the methods described herein comprise co-administering an antibody and a complement modulating agent. [0057] In some embodiments the complement modulating agent is a complement depleting agent and/or an agent that inhibits the formation and/or activity of C3b.
[0058] The complement system is part of the innate immune system. It attacks pathogens in a nonspecific manner (i.e., in a non-adaptive manner). In certain instances, the complement system functions by recruiting immune system cells (e.g. macrophages and neutrophils) to the site of an infection by chemotaxis. In certain instances, the complement system also utilizes the complement cascade to attack pathogens and recruit immune system cells. In certain instances, the complement system also removes foreign substances by action of white blood cells (e.g. neutrophils and macrophages).
[0059] In certain instances, the inactive complement system comprises over 20 proteins and enzymes, most of which are present in an inactive form. In certain instances, activation of complement (e.g., by the presence of antibodies, the presence of antigens, or the spontaneous hydrolysis of C3) activates the inactive proteases in the system. In certain instances, the proteases cleave targets (e.g., C3). In certain instances, the initial cleavage of the targets results (either partially or fully) in a cascade of cleavages. In certain instances, an active complement system comprises anaphylatoxins (e.g. C3a and C5a), the membrane attack complex (MAC), and proteins that facilitate opsonization (e.g. C3b).
Complement cascade pathways
[0060] In certain instances, the complement system is activated by three pathways; the classical pathway, the alternative pathway, and the mannose-binding lectin pathway.
[0061] In certain instances, the classical pathway begins with the activation of the enzyme Cl
(Clq2Clr2Cls). In certain instances, the CIq subunit of this enzyme either directly binds to an antigen or it binds to an antibody bound to an antigen. In certain instances, the binding of CIq leads to a conformational change in CIq. In certain instances, the conformational change in CIq leads to the activation of the two CIr subunits (CIr*) and CIs subunits (CIs*). In certain instances, the activation of the CIr and CIs subunits results in an active Cl enzyme (Clq2Clr*2Cls*).
[0062] In certain instances, activated C 1 cleaves the protein complement component 4 (C4) into C4a and C4b. In certain instances, C4b binds to the plasma membrane of a pathogen or a host cell. In certain instances, activated C 1 also cleaves the protein complement component 2 (C2) into C2a and C2b. In certain instances, C2a binds to C4b forming a C3 convertase (C4bC2a). In certain instances, C4bC2a cleaves the protein C3 into C3a and C3b. In certain instances, C3b binds to the membrane of a pathogen or host cell facilitating opsonization (e.g. the promotion of leukocyte chemoattraction, antigen binding, and phagocytosis) of the pathogen or host cell. In certain instances, C3b binds to C4bC2a forming a C5 convertase (C4bC2aC3a) which cleaves complement component 5 (C5) into C5a and C5b. [0063] In certain instances, the alternative pathway begins with the spontaneous hydrolysis of protein C3 (complement component 3), forming C3(H2O). In certain instances, the hydrolysis of C3 causes a conformational change that allows Factor B to bind to C3(H2O). In certain instances, Factor D cleaves Factor B into Ba and Bb. In certain instances, Bb remains bound to C3(H2O) forming the complex C3 (H2O)Bb (the fluid phase C3 convertase). In certain instances, the fluid phase C3 convertase cleaves C3 into C3a and C3b. In certain instances, C3b binds to the plasma membrane of a pathogen or a host cell where it facilitates opsonization of the host cell or pathogen. In certain instances, the C3b is bound by Factor B. In certain instances, when bound to C3b, Factor B is cleaved by Factor D into Ba and Bb. In certain instances, Bb remains bound to C3b forming an unstable C3 protease (C3Bb). In certain instances, the unstable C3bBb protease is stabilized by the binding of the protein properdin (P) forming a more stable C3 convertase (C3bBbP). In certain instances, upon the binding of a second C3b component, the C3bBbP becomes a C5 convertase (C3bBbC3bP).
[0064] In certain instances, C3b binds to the membrane of an antigen presenting cell. In certain instances, the binding of C3b to an antigen presenting cell facilitates opsonization of the antigen presenting cell. In certain instances, the binding of C3b to an antigen presenting cell interferes with ADCC. In some embodiments, the methods described herein comprise depleting and/or inhibiting the activity of a C3 convertase (e.g., C4bC2a, C3(H2O)Bb, C3bBb, C3bBbP). In some embodiments, the methods described herein comprise inhibiting the formation of a C3 convertase. In some embodiments, depleting and/or inhibiting the activity of a C3 convertase, or inhibiting the formation of a C3 convertase comprises inhibiting the expression of C3, inhibiting the expression of any of the subunits of Cl, inhibiting the activity of Cl, inhibiting the expression of C4, inhibiting the expression of C2, inhibiting the expression of Factor B, increasing the expression of Factor I, administering exogenous Factor I, administering exogenous CRl. MAC
[0065] In certain instances, the Membrane Attack Complex (MAC) comprises five protein sub-units: C5b, C6, C7, C8, and C9. In certain instances, C5b is produced by the cleavage of C5. In certain instances, following the cleavage of C5, C5b binds C6. C5bC6 is then bound by C7. In certain instances, the binding of C7 induces a conformational change in C7, exposing a hydrophobic domain. In certain instances, the hydrophobic domain enables C7 to insert itself into the plasma membrane of a pathogen or host cell. In certain instances, C8 binds to the C5bC6C7 complex. In certain instances, the binding of C8 also induces a conformational change in C8, exposing a hydrophobic domain that enables C8 to insert itself into the plasma membrane. In certain instances, the C5bC6C7C8 complex induces the polymerization of multiple C9 proteins. In certain instances, the C9 proteins form a pore in a plasma membrane. In certain instances, the pore allows the free diffusion of fluids, ions, and proteins into and out of the cell; a process that ultimately leads to the death of a cell. Anaphylatoxins
[0066] C3a, C4a, and C5a are anaphylatoxins. In certain instances, anaphylatoxins are fluid phase proteins that bind to receptors on mast cells. In certain instances, anaphylatoxins regulate smooth muscle spasms (e.g. bronchospasms), increase in the permeability of capillaries, and are chemotactic targets of leukocytes (e.g. the follow the increasing concentration gradient of an anaphylatoxin). In certain instances, C3a and C5a are the most potent anaphylatoxins.
[0067] In certain instances, C3a regulates degranulation of Mast-cells and serves as a chemotactic target for eosinophile granulocytes.
[0068] In certain instances, C5a serves as a chemotactic target for granulocytes and macrophages, and regulates vascular permeability, smooth muscle spasms and mast cell degranulation. In certain instance, C5a accelerates the growth of a neoplasm. In certain instances, C5a recruits myeloid-derived suppressor cells (MDSC). In certain instances, MDSC inhibit (either partially or fully) the activity of CD8+ T-cells. In certain instances, antagonizing C5a and/or C5aR inhibits (either partially or fully) the growth of a neoplasm. In some embodiments, the methods described herein deplete and/or inhibit the complement cascade without or only minimally cleaving C5 (i.e., producing C5a).
IV. Inflammatory Disorders
[0069] In some embodiments, the methods and compositions described herein treat inflammation (e.g., acute or chronic). In certain instances, inflammation results from (either partially or fully) an infection. In certain instances, inflammation results from (either partially or fully) damage to a tissue (e.g., by a burn, by frostbite, by exposure to a cytotoxic agent, or by trauma). In certain instances, inflammation results from (either partially or fully) an autoimmune disorder. In certain instances, inflammation results from (either partially or fully) the presence of a foreign body (e.g., a splinter). In certain instances, inflammation results from exposure to a toxin and/or chemical irritant.
[0070] As used herein, "acute inflammation" refers to inflammation characterized in that it develops over the course of a few minutes to a few hours, and ceases once the stimulus has been removed (e.g., an infectious agent has been killed by an immune response or administration of a therapeutic agent, a foreign body has been removed by an immune response or extraction, or damaged tissue has healed). The short duration of acute inflammation results from the short half- lives of most inflammatory mediators.
[0071] In certain instances, acute inflammation begins with the activation of leukocytes (e.g., dendritic cells, endothelial cells and mastocytes). In certain instances, the leukocytes release inflammatory mediators (e.g., histamines, proteoglycans, serine proteases, eicosanoids, and cytokines). In certain instances, inflammatory mediators result in (either partially or fully) the symptoms associated with inflammation. For example, in certain instances an inflammatory mediator dilates post capillary venules, and increases blood vessel permeability. In certain instances, the increased blood flow that follows vasodilation results in (either partially or fully) rubor and calor. In certain instances, increased permeability of the blood vessels results in an exudation of plasma into the tissue leading to edema. In certain instances, the latter allows leukocytes to migrate along a chemotactic gradient to the site of the inflammatory stimulant. Further, in certain instances, structural changes to blood vessels (e.g., capillaries and venules) occur. In certain instances, the structural changes are induced (either partially or fully) by monocytes and/or macrophages. In certain instances, the structural changes include, but are not limited to, remodeling of vessels, and angiogenesis. In certain instances, angiogenesis contributes to the maintenance of chronic inflammation by allowing for increased transport of leukocytes. Additionally, in certain instances, histamines and bradykinin irritate nerve endings leading to itching and/or pain.
[0072] In certain instances, chronic inflammation results from the presence of a persistent stimulant (e.g., persistent acute inflammation, bacterial infection (e.g., by Mycobacterium tuberculosis), prolonged exposure to chemical agents (e.g., silica, or tobacco smoke) and autoimmune reactions (e.g., rheumatoid arthritis)). In certain instances, the persistent stimulant results in continuous inflammation (e.g., due to the continuous recruitment of monocytes, and the proliferation of macrophages). In certain instances, the continuous inflammation further damages tissues which results in the additional recruitment of mononuclear cells thus maintaining and exacerbating the inflammation. In certain instances, physiological responses to inflammation further include angiogenesis and fibrosis.
[0073] Multiple disorders are associated with inflammation (i.e., inflammatory disorders).
Inflammatory disorders include, but are not limited to, Atopic dermatitis; Acute disseminated encephalomyelitis; Acute leukocyte-mediated lung injury; Addison's disease; AIDS dementia; Allergic rhinitis; Ankylosing spondylitis; Antiphospholipid antibody syndrome; Alzheimer's disorder; Asthma; Adult respiratory distress syndrome; Autoimmune hemolytic anemia; Autoimmune hepatitis; Autoimmune inner ear disease; Behcet's syndrome; Bronchitis; Bullous pemphigoid; Chagas disease; Chronic obstructive pulmonary disease; Coagulative Necrosis; Coeliac disease; Crohn's disorder; Collagenous colitis; Conjunctivitis; Cystic fibrosis; Dermatomyositis; Dermatitis; Diabetes mellitus type 1; Diabetes mellitus type 2; Distal proctitis; Diversion colitis; Encephalitis; Endometriosis; Endotoxin shock; Eczema; Epilepsy; Fibromyalgia; Fibrinoid Necrosis; Goodpasture's syndrome; Gouty arthritis; Graves' disease; Guillain-Barre syndrome; Hashimoto's disease; Idiopathic thrombocytopenic purpura; Indeterminate colitis; Infective colitis; Inflammatory liver disorder; Interstitial cystitis; Ischaemic colitis; Liquefactive Necrosis; Lymphocytic colitis; Meningitis; Multiple sclerosis; Myasthenia gravis; Myocarditis; Narcolepsy; Nephritis; Pancreatitis; Parkinson's disorder; Pemphigus Vulgaris; Periodontal gingivitis; Pernicious anaemia; Polymyositis; Polymyalgia rheumatica; Psoriasis; Primary biliary cirrhosis; Retinitis; Rheumatoid arthritis; Rheumatoid spondylitis; Schizophrenia; Scleroderma; Shingles; Sjogren's syndrome; Smooth muscle proliferation disorders; Systemic lupus erythematosus (SLE); Tuberculosis; Ulcerative colitis; Uveitis; Vasculitis; Vitiligo; Wegener's granulomatosis; Transplant complication; Hyperacute transplant rejection; Acute transplant rejection; Chronic transplant rejection; Acute graft-versus- host disease; Chronic graft-versus-host disease, or combinations thereof.
[0074] In some embodiments, the methods and compositions described herein treat a T-cell mediated autoimmune disorder. In certain instances, a T-cell mediated autoimmune disorder is characterized by a T-cell mediated immune response against self (e.g., native cells and tissues).
[0075] Examples of T-cell mediated autoimmune disorders include, but are not limited to colitis, multiple sclerosis, arthritis, rheumatoid arthritis, osteoarthritis, juvenile arthritis, psoriatic arthritis, acute pancreatitis, chronic pancreatitis, diabetes, insulin-dependent diabetes mellitus (IDDM or type I diabetes), insulitis, inflammatory bowel disease, Crohn's disease, ulcerative colitis, autoimmune hemolytic syndromes, autoimmune hepatitis, autoimmune neuropathy, autoimmune ovarian failure, autoimmune orchitis, autoimmune thrombocytopenia, reactive arthritis, ankylosing spondylitis, silicone implant associated autoimmune disease, Sjogren's syndrome, systemic lupus erythematosus (SLE), vasculitis syndromes (e.g., giant cell arteritis, Behcet's disease & Wegener's granulomatosis), vitiligo, secondary hematologic manifestation of autoimmune diseases (e.g., anemias), drug- induced autoimmunity, Hashimoto's thyroiditis, hypophysitis, idiopathic thrombocytic pupura, metal- induced autoimmunity, myasthenia gravis, pemphigus, autoimmune deafness (e.g., Meniere's disease), Goodpasture's syndrome, Graves' disease, HIV-related autoimmune syndromes and Gullain- Barre disease.
[0076] In some embodiments, the methods and compositions described herein treat pain. Pain includes, but is not limited to acute pain, acute inflammatory pain, chronic inflammatory pain and neuropathic pain.
[0077] In some embodiments, the methods and compositions described herein treat hypersensitivity. As used herein, "hypersensitivity" refers to an undesirable immune system response. Hypersensitivity is divided into four categories. Type I hypersensitivity includes allergies (e.g., Atopy, Anaphylaxis, or Asthma). Type II hypersensitivity is cytotoxic/antibody mediated (e.g., Autoimmune hemolytic anemia, Thrombocytopenia, Erythroblastosis fetalis, or Goodpasture's syndrome). Type III is immune complex diseases (e.g., Serum sickness, Arthus reaction, or SLE). Type IV is delayed-type hypersensitivity (DTH), Cell-mediated immune memory response, and antibody-independent (e.g., Contact dermatitis, Tuberculin skin test, or Chronic transplant rejection). [0078] As used herein, "allergy" means a disorder characterized by excessive activation of mast cells and basophils by IgE. In certain instances, the excessive activation of mast cells and basophils by IgE results (either partially or fully) in an inflammatory response. In certain instances, the inflammatory response is local. In certain instances, the inflammatory response results in the narrowing of airways (i.e., bronchoconstriction). In certain instances, the inflammatory response results in inflammation of the nose (i.e., rhinitis). In certain instances, the inflammatory response is systemic (i.e., anaphylaxis).
[0079] In some embodiments, the methods and compositions described herein treat non- cancer-based angiogenesis. As used herein, "non-cancer-based angiogenesis" refers to the formations of new blood vessels that arises from non-cancer conditions. In certain instances, angiogenesis occurs with chronic inflammation. In certain instances, angiogenesis is induced by monocytes and/or macrophages.
[0080] In some embodiments, the methods and compositions described herein treat obesity. As used herein, "obesity" means an accumulation of adipose tissue with a BMI of greater than or equal to 30kg/m . In certain instances, obesity is characterized a proinflammatory state, increasing the risk of thrombosis. In certain instances, obesity is associated with a low-grade inflammation of white adipose tissue (WAT). In certain instances, WAT associated with obesity is characterized by an increased production and secretion of a wide range of inflammatory molecules including TNF-alpha and interleukin-6 (IL-6). In certain instances, WAT is infiltrated by macrophages, which produce pro-inflammatory cytokines. In certain instances, TNF-alpha is overproduced in adipose tissue. In certain instances, IL-6 production increases during obesity.
[0081] In some embodiments, the methods and compositions described herein treat metabolic syndrome. In certain instances, metabolic syndrome is associated with fasting hyperglycemia; high blood pressure; central obesity; decreased HDL levels; elevated triglyceride levels; systemic inflammation; or combinations thereof. In certain instances, metabolic syndrome is characterized by an increase in the levels of C-reactive protein, fibrinogen, (IL-6), and TNFα.
[0082] As used herein, "transplant complications" means an immune-mediated complication resulting from an organ, a plurality of cells, and/or a tissue transplant. For example, a "transplant complication" can result from the recipient's immune system mounting an immune response against the transplanted organ, tissue, and/or plurality of cells. A "transplant complication" also includes an immune response mounted against the recipient by the immune cells from the transplanted organ, plurality of cells, and/or tissue. "Transplant complications" includes hyperacute transplant rejection, acute transplant rejection, chronic transplant rejection, acute graft-versus-host disease, chronic graft-versus-host disease, or combinations thereof. ccines [0083] Disclosed herein, in certain embodiments, is a method of manufacturing a vaccine. In certain instances, a pathogen (e.g., a virus) activates complement in organisms as they infect the organism. Thus, in certain instances, complement is needed to mount a successful immune reaction. In certain instances, inhibiting or depleting the complement system in an organism with active infection facilitates replication of the pathogen.
[0084] In some embodiments, the method comprises administering a complement depletor (e.g., CVF, hybrid human C3-CVF protein, or a modified human complement C3 protein) to a fertilized egg or a live chick embryos that has been inoculated (or challenged) with a pathogen. In some embodiments, the complement depletor depletes C3 (i.e., removes the process of amplification of complement activation as well as activation of the terminal complement pathway). In some embodiments, the complement depletor removes the process of amplification of complement activation as well as activation of the terminal complement pathway. In some embodiments, depleting or inhibiting complement increases pathogen replication. In some embodiments, increasing pathogen replication increases production of a vaccine. In some embodiment, the method applies to any pathogen used in vaccine production. In some embodiment, the method applies to any in- vivo system, including certain cellular systems, used in vaccine production. ood Storage
[0085] Disclosed herein, in certain embodiments, is a method of collecting and storing whole blood or blood components. In certain instances, collecting blood (and the necessary exposure to collection tubes and bags), separating blood into components (e.g., plasma) and storing whole blood or blood components activate the complement cascade. In certain instances, activation of the complement cascade is amplified due to the activation of C3. In certain instances, the activation of C3 leads to the production of and then the release of C3a and C3b. In certain instances, the production of and then the release of C3a and C3b lead further to the activation of the terminal complement cascade and production of C5a and C5b-9. In certain instances, the activation of C3 and any or all of the downstream complement proteins induces and/or exacerbates the degradation of whole blood (i.e., induces the degradation of components of whole blood), or lyses blood cells and/or blood components. In certain instances, transfusing an organism with whole blood comprising activated C3 with or without activated C5 and related downstream proteins further results (partially or fully) in organ dysfunction or systemic inflammatory response. Thus, in certain instances, depleting the complement system in whole blood (a) during collection, (b) before storage, or (c) before separation of the blood into components increases: (a) the shelf-life of blood and/or blood components and (b) the safety of infusion of whole blood, packed red blood cells and/or human blood components. [0086] In some embodiments, the method comprises contacting a complement depletor (e.g., CVF, recombinant CVF, hybrid human C3-CVF protein, a modified human complement C3 protein which depletes complement at C3 without activating C5) with whole blood or blood components. In some embodiments, the method comprises contacting a complement depletor (e.g., CVF, hybrid human C3-CVF protein, a modified human complement C3 protein) with whole blood (a) during collection, (b) before storage of the blood, or (c) before separation of the blood into components. In some embodiments, the method comprises contacting an HC3-1496 protein, an analogue thereof, or combinations thereof with whole blood. In some embodiments, the method comprises contacting an HC3-1496 protein, an analogue thereof, or combinations thereof with whole blood before storage of the blood or separation of the blood into components. In some embodiments, contacting a complement depletor (e.g., HC3-1496), an analogue thereof, or combinations thereof with whole blood removes the process of amplification of complement activation as well as activation of the terminal complement pathway. In some embodiments, depleting complement decreases the concentration of C3a and/or C3b and/or C5a and/or C5b, including C5b-9 complex. In some embodiments, decreasing the concentration of C3a and/ or C3b and/or C5a and/or C5b, including C5b-9 complex increases the amount of whole blood (or blood components) that can be transfused, increases the storage time for whole blood (or blood components), protects organs, reduces systemic inflammatory response, allows for reinfusion of shed blood which has been decomplemented by a complement depletor, or combinations thereof.
VII. Therapeutic Agents
Antibodies
[0087] In some embodiments, the antibody is an anti-CD3 antibody, an anti-CD 11 a§ antibody, an anti-CD-20 antibody, an anti-CD22 antibody, an anti-CD-25 antibody, an anti-CD52 antibody, anti- TNF antibody, anti- IgE antibody, an anti- α4-integrin antibody, anti- RSV gpF antibody, or combinations thereof.
[0088] In some embodiments, the antibody is an anti-CD3 antibody. In certain instances, CD3 is expressed on T-cells. In certain instances, the binding of an antibody to a T-cell induces an immune response against the T-cell. Further, in certain instances, antagonizing CD3 prevents T cells from replicating, and from activating B cells. In certain instances, B-cells bind to antigens (e.g., those expressed by transplanted organs and stimulate an immune response against the antigen presenting cell. In some embodiments, an anti-CD3 antibody is administered to treat an immune disorder. In some embodiments, an anti-CD3 antibody is administered to treat transplant rejection. In some embodiments, an anti-CD3 antibody is administered to treat kidney transplant rejection. In some embodiments, the antibody is muromonab-CD3. [0089] In some embodiments, the antibody is an anti-CD 11a antibody. In certain instances, CD 11 a is the α subunit of leukocyte function antigen- 1 (LFA-I). In certain instances, LFA-I is expressed by T-cells. Further, in certain instances, the binding of an antibody to a T-cell induces an immune response against the T-cell. In certain instances, LFA-I binds to intercellular adhesion molecule (ICAM)- 1. In certain instances, ICAM- 1 modulates the migration of T cells to sites of inflammation. In some embodiments, an anti- CD 11a antibody is administered to treat an immune disorder. In some embodiments, an anti- CD 11a antibody is administered to treat plaque psoriasis. In some embodiments, the antibody is efalizumab.
[0090] In some embodiments, the monoclonal antibody is an anti-CD20 antibody. In certain instances, CD20 is expressed on about 90% of mature B-cells. In certain instances, CD20 is absent (either partially or fully) on progenitor B-cells. In some embodiments, an anti-CD20 antibody is administered to treat a B-cell mediated disorder. In some embodiments, an anti- CD20 antibody is administered to treat an autoimmune disorder. In some embodiments, an anti- CD20 antibody is administered to treat rheumatoid arthritis, a transplant complication, a transplant rejection (e.g., hyperacute, actue, and chronic), graft-versus-host disease (e.g., actue, and chronic), idiopathic autoimmune hemolytic anemia, pure red cell aplasia, idiopathic thrombocytopenic purpura, Evans syndrome, vasculitis, multiple sclerosis, bullous skin disorders (e.g., pemphigus, pemphigoid), type 1 diabetes mellitus, Sjogren's syndrome, Devic's Syndrome, systemic lupus erythematosus, or a combination thereof. In some embodiments, the anti-CD20 antibody is epratuzumab, GAlOl (R7159, Genentech), ocrelizumab (PRO70769), ofatumumab (HuMax-CD20 human IgGl monoclonal antibody or 2F2), PRO 131921 (Genentech), rituximab, veltuzumab (IMMU- 106 or hA20), or combinations thereof.
[0091] In some embodiments, the monoclonal antibody is an anti-CD22 antibody. In certain instances, CD22 present or all or most mature B-lineage cells. In certain instances, CD22 is not expressed (or only minimally expressed) on hematopoietic stem cells. In some embodiments, an anti-CD22 antibody is administered to treat an inflammatory disorder. In some embodiments, an anti-CD22 antibody is administered to treat lupus (SLE). In some embodiments, the anti-CD22 antibody is epratuzumab.
[0092] In some embodiments, the monoclonal antibody is an anti-CD25 antibody. In certain instances, CD25 is expressed on T-cells. In certain instances, the binding of an antibody to a T- cell induces an immune response against the T-cell. Further, in certain instances, antagonizing CD25 prevents T cells from replicating, and from activating B cells. In certain instances, B-cells bind to antigens (e.g., those expressed by transplanted organs and stimulate an immune response against the antigen presenting cell. In some embodiments, an anti-CD25 antibody is administered to treat an immune disorder. In some embodiments, an anti-CD3 antibody is administered to treat transplant rejection. In some embodiments, an anti-CD3 antibody is administered to treat kidney transplant rejection. In some embodiments, an anti-CD3 antibody is administered to treat graft-versus-host disease (e.g., acute, and chronic). In some embodiments, an anti-CD3 antibody is administered to treat multiple sclerosis. In some embodiments, the monoclonal antibody is daclizumab, basiliximab, or a combination thereof.
[0093] In some embodiments, the monoclonal antibody is an anti-CD52 antibody. In certain instances, CD52 is expressed on the surface of mature lymphocytes (e.g., expressed by lymphocytes, especially T cells, monocytes, macrophages, and monocyte-derived dendritic cells). In certain instances, CD52 is absent (either partially or fully) from hematopoietic progenitor cells. In some embodiments, an anti-CD52 antibody is administered to condition an individual for a transplant. In some embodiments, an anti-CD52 antibody is administered to condition an individual for a bone marrow transplant and/or kidney transplant. In some embodiments, an anti-CD52 antibody is administered to treat an immune disorder. In some embodiments, an anti-CD52 antibody is administered to treat multiple sclerosis and/or graft- versus-host disease (e.g., acute, and chronic). In some embodiments, the anti-CD52 antibody is alemtuzumab.
[0094] In some embodiments, the monoclonal antibody is an anti- TNF antibody. In certain instances, TNF- α mediates inflammation. In certain instances, TNF-α levels are elevated in individuals with Crohn's Disease. In certain instances, TNF-α levels are elevated in sera and synovial fluid of individuals with rheumatoid arthritis. In certain instances, the binding of an antibody to a TNF- α molecule initiates an immune response against the TNF- α molecule. In certain instances, an immune response against a TNF- α molecule inhibits (either partially or fully) the ability of the TNF- α molecule to bind to TNF receptors. In certain instances, inhibiting the ability of the TNF- α molecule to bind to TNF receptors inhibits (either partially or fully) the production of proinflammatory cytokines (e.g., IL-6, IL-I), the production of proteases (e.g., the proteases that render blood vessels permeable to leukocytes), and the release of adhesion molecules. In some embodiments, an anti- TNF antibody is administered to treat an immune disorder. In some embodiments, an anti- TNF antibody is administered to treat rheumatoid arthritis, psoriatic arthritis, ulcerative colitis, Crohn's Disease, ankylosing spondylitis, or a combination thereof. In some embodiments, the monoclonal antibody is infliximab, adalimumab, or a combination thereof.
[0095] In some embodiments, the monoclonal antibody is an anti- IgE antibody. In certain instances, IgE mediates immune response to allergens. In certain instances, the binding of an antibody to a IgE cell induces an immune response against the IgE cell. Further, in certain instances, IgE bound by an antibody cannot bind to a basophil. In certain instances, IgE bound by an antibody attenuates degranulation. In some embodiments, an anti- IgE antibody is administered to treat an immune disorder. In some embodiments, an anti- IgE antibody is administered to treat an allergic reaction and/or hypersensitivity. In some embodiments, an anti- IgE antibody is administered to treat asthma. In some embodiments, the monoclonal antibody is omalizumab. [0096] In some embodiments, the monoclonal antibody is an anti- RSV gpF antibody. In some embodiments, an anti- RSV antibody is administered to treat an RSV infection. In some embodiments, the monoclonal antibody is palivizumab.
[0097] In some embodiments, the monoclonal antibody is an anti-α4-integrin (also known as VLA-4) antibody. In certain instances, VLA-4 is a receptor found on leukocytes that mediates adhesion and migration of immune cells through interaction with its ligand, (VCAM)-I. In certain instances, VLA-4 is expressed by leukocytes infiltrating the central nervous system and the gut. In certain instances, the binding of an antibody to a leukocyte induces an immune response against the leukocyte. In certain instances, VLA-4 mediates Th- 1 cell migration in animal models of MS, and experimental autoimmune encephalomyelitis. In certain instances, the binding of an antibody to a Th- 1 cell induces an immune response against the Th- 1 cell. In some embodiments, an anti- α4-integrin antibody is administered to treat an immune disorder. In some embodiments, an anti- α4-integrin antibody is administered to multiple sclerosis, Crohn's disease, rheumatoid arthritis, allergic encephalomyelitis, adjuvant induced arthritis, diabetes mellitus, graft-versus-host disease (e.g., acute, and chronic), allograft rejection and/or immediate hypersensitivity reactions. In some embodiments, the monoclonal antibody is natalizumab.
Complement Modulating Agents
[0098] In some embodiments, the methods described herein comprising co-administering to an individual in need thereof a synergistically-effective amount of (a) an antibody or fusion protein comprising an Fc domain; and (b) a complement modulating agent. In some embodiments, the complement modulating agent is a complement depleting agent, an agent that inhibits the formation and/or activity of C3b, or a combination thereof. In some embodiments, the complement modulating agent depletes and/or inhibits the activity of a C3 convertase; or inhibits the formation of a C3 convertase (e.g., by inhibiting the expression of C3, inhibiting the expression of any of the subunits of Cl, inhibiting the activity of Cl, inhibiting the expression of C4, inhibiting the expression of C2, inhibiting the expression of Factor B, or increasing the expression of Factor I).
[0099] In some embodiments, the methods described herein comprising co-administering to an individual in need thereof a synergistically-effective amount of (a) an antibody or fusion protein comprising an Fc domain; and (b) a complement modulating agent. In some embodiments, the complement modulating agent is a complement enhancing (i.e., agonizing) agent. In some embodiments, a complement modulating agent increases the expression of a complement protein. In some embodiments, a complement modulating agent catalyzes the cleavage of a complement component (e.g., C3 or C5) by a complement convertase (e.g., a C3 convertase, or C5 convertase). [00100] For methods of assaying complement depletion (e.g., detecting lysis of antibody-sensitized sheep erythrocytes induced by a serum (such as a human serum) that has reacted with the compound in vitro and in vivo) see, e.g., WO 2005/107785; US 2005/0079585; Ballow et al., J. Immunol. 103:944-952, 1969; Cochrane et al., J. Immunol. 105:55-69, 1970 which are hereby incorpiorated by reference for such disclosures. Anaphylatoxin Modulators
[00101] In some embodiments, an anaphylatoxin modulator is administered to an individual in need thereof. In some embodiments, the anaphylatoxin modulator is an antagonist of C5a. In some embodiments, the C5a antagonist is chemotaxis inhibitory protein of S. aureus (CHIPS), PMX53 (AcF[OP-DCha-WR]), PMX205 (HC-[OPdChaWR]), PMX273 (AcF[OP-DPhe-WR]), PMX201 (AcF[OP-DCha-WCit]), PMX218 (HC-[OPdPheWR]), C089 (NMePhe-Lys-Pro- dCha-X-dArg), L- 156,602 (D-Alanine,(RS,2R,5R,6R)-tetrahydro-R,2- dihydroxy-R,6- dimethyl-5-[(2S)-2-methylbutyl]- 2H-pyran-2-acetyl-(3S)-3-hydroxy- L-leucyl-(3R)- hexahydro-3- pyridazinecarbonyl-N-hydroxy-L-alanylglycyl-( 3S)-hexahydro-3- pyridazinecarbonyl- N-hydroxy-,(7f2)-lactone, CAS #: 125228-51-5), C5aRAM, C5aRAD, or combinations thereof. In certain instances, a C5a antagonist binds to a C5aR and thereby antagonizes the binding of C5a. In certain instances, CHIPS binds to the C5a receptor (C5aR) on a macrophage thereby inhibiting the C5a induced chemotaxis of a macrophage. In certain instances, C5aRAM and C5aRAD are derived from the modification of the C terminus of C5a.
[00102] In some embodiments, the antagonist of C5aR activation is an antisense peptide. In some embodiments, the antisense peptide of C5a is PR226-MAP (LRTWSRRATRSTKTLKVV), PL37-MAP (RAARISLGPRCIKAFTE), or combinations thereof. In certain instances, a C5a antagonist binds to a C5aR and thereby antagonizes the binding of C5a.
[00103] In some embodiments, an anaphylatoxin modulator is administered to an individual in need thereof. In some embodiments, the anaphylatoxin modulator is an antagonist of C3a. In some embodiments, the C3a antagonist is SB-290157 (N(2)-[(2,2-diphenylethoxy)acetyl]-L-arginine). In certain instances, SB-290157 binds to the C3a receptor (C3aR) thereby blocking the binding of C3a. Compliment Activating Agents/Complement Depleting Agents
[00104] In some embodiments, a complement activator is administered to an individual in need thereof. In some embodiments, the complement activator is GR-2II, a pectic arabinogalactan (e.g. AGIIa, and AGIIb-I), a pectin (e.g. AR-2IIa, AR-2IIb, AR-2IIc, and AR-2IId), CVF, a hybrid human C3-CVF protein, or combinations thereof. In certain instances, AR-2IIa, AR- 21Ib, and AR-2IIc activate the complement system via the classical pathway and not the alternative pathway.
[00105] Cobra Venom Factor (CVF) is a three-chain (α-chain, β-chain, and γ-chain) glycoprotein extracted from the reptile Naja sp. CVF is a human complement system activating protein. It is structurally homologous to C3b. In certain instances, CVF binds to Factor B which is then cleaved by Factor D. The resulting complex, CVFBb, functions as a C3 convertase and a C5 convertase. At 7.5 hours CVFBb exhibits a longer half-life than C3bBb (1.5 minutes). Further, CVFBb is resistant to disassembly by Factor H and CVF is resistant to inactivation by Factor I. As a result, CVFBb will continuously hydro lyze C3 and C5. The continuous hydro lyzation of C3 and C5 results in the depletion (or exhaustion) of the complement system within several hours. However, resynthesis of the components of the complement system begins quickly and the entire system is reconstituted with 5-10 days.
[00106] In certain instances, CVF is highly antigenic in vivo. As a result, several humanized analogs and/or derivatives of CVF (also known as, "humanized CVF" and "hybrid human C3-CVF protein") have been engineered. In certain instances, these derivatives exhibit similar activity to natural CVF (e.g. 50-97% of the activity of natural CVF); however, they do not or have a reduced capability of activating an immune response in vivo. In certain analogs and/or derivatives, several amino acids from the β-chain of the CVF polypeptide are removed. In further analogs and/or derivatives, the CVF polypeptide is conjugated to a human antibody (e.g. monoclonal antibodies against antigen on human leukemia cells, human neuroblastoma cells, and human melanoma cells). In some analogs and/or derivatives, a human C3 derivative and/or analog (e.g. recombinant C3, rC3, humanized CVF) is engineered such that the human C3 derivative and/or analog comprises a portion of a CVF polypeptide sequence. In other analogs and/or derivatives, portions of a human C3 polypeptide (e.g. the α-chain; or portions of the carboxy terminal) are replaced with the corresponding portion of the CVF polypeptide. In certain derivatives and/or analogs, the α-chain of the human C3 is replaced by the corresponding carboxy terminal amino acids of the CVF polypeptide. In some embodiments, the CVF analog and/or derivative is HC3-1496, HC3-1496-2, HC3-1496-3, HC3-1496-4, HC3- 1496/1617, HC3-1496-8, HC3-1496-9, HC3-1496-10, HC3-1496-11, HC3-1496-12, HC3- 1496-13, HC3-1496-14, HC3-1496-15, HC3-1496-16, HC3-1496-17, or combinations thereof. For disclosures regarding the aforementioned CVF analogs and/or derivatives see PCT Pub. No. WO 2005/003159; and PCT Pub. No. WO 2008/060634, which are herein incorporated by reference for such disclosures. For additional disclosures regarding CVF derivatives and/or analogs see U.S. Patent No. 5,714,344, which is hereby incorporated by reference for such disclosures. Corresponding amino acid residues between prepro-CVF from Naja naja cobra and prepro-human C3 are shown in Figs. 2A-2D.
[00107] Disclosed herein, in certain embodiments, is a hybrid human C3-CVF protein that has one or more of the following characteristics: (a) ability to deplete a complement component; (b) ability to mediate the cleavage of factor B; (c) ability to form C3 convertase; (d) ability to cleave C3 and/or C5 upon activation; (e) increased resistance to the regulatory actions of factors H and/or I as compared to native human C3 protein; (f) binding affinity to factor D; (g) increased intrinsic half-life than native human C3 protein; and (h) less immunogenicity than CVF. In some embodiments, the hybrid human C3-CVF protein has sequence changes that have C3 complement depleting characteristics similar or superior to that of recombinant or natural cobra venom factor, but without activation of C5.
[00108] In some embodiments, a hybrid human C3-CVF protein has at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, or at least about 95% identity with the human C3 sequence.
[00109] Two polynucleotide or polypeptide sequences are said to be "identical" if the sequence of nucleotides or amino acids in the two sequences is the same when aligned for maximum correspondence as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity. A "comparison window" as used herein, refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence is compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
[00110] In some embodiments, optimal alignment of sequences for comparison is conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, WI), using default parameters. This program embodies several alignment schemes described in the following references: Dayhoff, M.O. (1978) A model of evolutionary change in proteins - Matrices for detecting distant relationships. In Dayhoff, M.O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington DC Vol. 5, Suppl. 3, pp. 345-358; Hein J., 1990, Unified Approach to Alignment and Phylogenes pp. 626- 645 Methods in Enzymology vol. 183, Academic Press, Inc., San Diego, CA; Higgins, D. G. and Sharp, P.M., 1989, CABIOS 5:151-153; Myers, E. W. and Muller W., 1988, CABIOS 4: 11-17; Robinson, E.D., 1971, Comb. Theor. 11: 105; Santou, N., Nes, M., 1987, MoI. Biol. Evol. 4:406-425; Sneath, P.H.A. and Sokal, R.R., 1973, Numerical Taxonomy the Principles and Practice of Numerical Taxonomy, Freeman Press, San Francisco, CA; Wilbur, W.J. and Lipman, D. J., 1983, Proc. Natl. Acad. Sci. USA 80:726-730.
[00111] Preferably, the "percentage of sequence identity" is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window comprises additions or deletions (i.e. gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid bases or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e. the window size) and multiplying the results by 100 to yield the percentage of sequence identity.
[00112] In some embodiments, the substituted portion of the CVF is within the alpha chain of C3. In some embodiments, the substituted portion of the CVF is a C-terminal portion of the alpha chain of C3. In some embodiments, only the C-terminal portion of the alpha chain of C3 is substituted with the corresponding region of CVF protein. In some embodiments, the substituted portion of CVF is within the beta chain of C3. In some embodiments, the substituted C-terminal portion includes amino acid 1663 of SEQ ID NO: 1 (amino acid sequence of prepro- human C3). In some embodiments, the substituted C-terminal portion is an internal portion that does not extend through the entire C-terminus of the human C3 protein.
[00113] In some embodiments, the modified human C3 protein is a single chain protein. In some embodiments, the modified human C3 protein is cleaved into at least two chains in a form that resembles native human C3. In some embodiments, the modified human C3 protein is proteolytic cleaved to release a portion therefrom (e.g., a portion like a C3a). In some embodiments, the modified human C3 protein is a mature protein. In some embodiments, the modified human C3 protein has 1 to about 19 amino acids at the N-terminus that are not part of human C3 or CVF. In some embodiments, the modified human C3 protein contains the signal sequence. In some embodiments, the signal sequence is a non-human C3 signal peptide, such as a Drosophila signal sequence. In some embodiments, the modified human C3 protein has an affinity for factor B and supports formation of a convertase. In some embodiments, the resulting convertase cleaves C3 and not C5. In some embodiments, the modified C3 protein has modified affinity for factor B and/or factor D. In some embodiments, the modified human C3 protein shows partial or complete resistance to Factor H and/or Factor I. In some embodiments, the modified human C3 protein is substantially non-immunogenic. In some embodiments, the convertase has an intrinsic half-life between about 1.5 minutes and about 7 hours at 37°C. In some embodiments, the resulting convertase has an intrinsic half- life of at least about 7 hours at 37°C.
[00114] In some embodiments, the methods described herein comprise administering to an individual in need thereof a modified human C3 protein, comprising a human C3 protein, wherein some or all amino acid residues in the human C3 protein corresponding to amino acid residues 700- 1663 of SEQ ID NO: 1 are substituted with a corresponding portion of a CVF protein, including but not limited to regions of from 20 to about 1000 amino acids, including but not limited to: 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 150, 175, 180, 190, 200, 250, 275, 300, 350, 375, 400, 450, 475, 500, 550, 575, 600, 650, 675, 700, 750, 775, 800, 850, 875, 900, 950 975, 1000. In some embodiments, the beta chain of C3 is intact, that is, is the same as in natural C3. In some embodiments, the invention provides a modified human C3 protein, comprising human C3 protein, wherein amino acid residues in the human C3 protein corresponding to amino acid residues 1550-1663, 1504-1663, 1348-1663, 1550-1617, 1504-1617, 1470-1663, 1348-1617, 1470-1617, 1264-1324, 1348-1386, 749-784, 874-921, 994-1663, 994-1550, 1496-1663, 1496- 1617, or 936-970 of SEQ ID NO:1 are substituted with a corresponding portion of CVF protein. In some embodiments, still smaller substitutions in these areas corresponds to smaller regions that result in a C3 with the desired CVF functions or qualities.
[00115] In some embodiments, the methods described herein comprise administering to an individual in need thereof a modified human C3 protein, comprising a human C3 protein, wherein the amino acid residues in the human C3 protein corresponding to amino acid residues 1550-1663, 1504-1663, 1496-1663 or 1348-1663 of SEQ ID NO: 1 are substituted with a corresponding portion of CVF protein. The modified human C3 proteins are referred to as HC3-1550, HC3- 1504, HC3-1496 and HC3-1348, respectively. In some embodiments, these proteins comprise a modified human C3 protein, comprising a human C3 protein, wherein the amino acid residues in the human C3 protein corresponding to amino acid residues 1550-1617 of SEQ ID NO: 1 are substituted with a corresponding protein of CVF protein. In some embodiments, these proteins comprise a modified human C3 protein, comprising a human C3 protein, wherein the amino acid residues in the human C3 protein corresponding to amino acid residues 1496-1663 of SEQ ID NO: 1 are substituted with a corresponding protein of CVF protein.
[00116] In some embodiments, the methods described herein comprise administering to an individual in need thereof a modified human C3 protein, comprising a human C3 protein, wherein at least 68 amino acid residues in the human C3 protein corresponding to amino acid residues 1596 to 1638 of SEQ ID NO: 1 (amino acid sequence of prepro-human C3) are substituted with the corresponding portion of CVF. In some embodiments, the amino acid residues in the human C3 protein corresponding to amino acid residues 1596 to 1638 of SEQ ID NO: 1 (amino acid sequence of prepro-human C3) are substituted with amino acid residues 1575 to 1617 of SEQ ID NO:2 (amino acid sequence of prepro-CVF). n some embodiments, the modified human C3 protein is any as described in US 2005/0079585, filed 07/02/2004 and published 04/14/2005.
[00117] In some embodiments, the methods described herein comprise administering to an individual in need thereof a proteins comprising a human C3 protein, wherein the amino acid residues in the human C3 protein corresponding to amino acid residues 1496 to 1663 of SEQ ID NO:1 (amino acid sequence of pro-human C3) are substituted with a corresponding portion of a CVF protein, and wherein one or more amino acid residues in the CVF portion of the modified human complement C3 protein are further modified. In some embodiments, the one or more amino acids in one or more of the following regions is modified: 1499-1501, 1507-1510, 1519- 1550, 1519-1524, 1528-1532, 1571-1578, 1596-1617, 1596-1611, and 1598-1608. In some embodiments, at least one, at least two, at least three, at least four, at least five, at least six, or more amino acids in one or more of these regions are modified. In some embodiments, any of one, two, three, four, five, six or more amino acids in one or more of these regions are modified. In some embodiments, the amino acids in one or more of these regions are substituted into corresponding amino acids in a human C3 protein, a cobra C3 protein, or other mammalian C3 proteins. Examples of the modifications of amino acid residues in the CVF portion of the modified human complement C3 protein include: 1) T1499D and L1501K; 2) I1507R, G1508D, N1509E, and V1510L; 3) S1519F, S 15201, L1521Q, N1522K, H1523S, and Q1524D; 4) D1528T, V1529L, P 1530E, L 153 IE, and Q1532R; 5) 1519-1550 replaced with corresponding amino acid residues of SEQ ID NO: 1; 6) Q1571S, T1573S, N1576V, P1577Q, and R1578V; 7) 1596-1617 replaced with corresponding amino acid residues of SEQ ID NO: 1; 8) 1596-1611 replaced with corresponding amino acid residues of SEQ ID NO: l; 9) V1598L, N1599D, D1600N, S1607L, and R1608S; 10) V1598L, N1599D, and D1600N; or 11) S1607L, and R1608S. In some embodiments, the amino acid positions indicated herein correspond to the amino acid positions in SEQ ID NO: 1, are in the actual position(s) in the modified human C3 protein. In some embodiments, the amino acid positions indicated herein correspond to the amino acid positions in SEQ ID NO: 1, are not in the actual position(s) in the modified human C3 protein.
[00118] In some embodiments, the methods described herein comprise administering to an individual in need thereof a modified human complement C3 proteins comprising a human C3 protein, wherein the amino acid residues in the human C3 protein corresponding to amino acid residues 1496 to 1617 of SEQ ID NO: 1 are substituted with a corresponding portion of a CVF protein, and wherein amino acid residues in the CVF portion of the modified human complement C3 protein are modified with E1654Y and V1658E.
[00119] In some embodiments, the modified human C3 protein is any as described in WO 05/107785, filed 04/29/2005 and published 11/17/2005, and US 60/859,330, filed on 11/15/2006.
[00120] In some embodiments, the modified human C3 protein is selected from Table 1. The modified human C3 proteins in Table 1 contain specific CVF sequences so as to create modified human C3 proteins with CVF function. The portions of human C3 that are replaced with corresponding CVF sequences are indicated in the table. This table should in no way be construed as limiting. Other possible modifications are possible and are intended to be included within the rubric of "hybrid human C3-CVF protein" or "modified CVF". Table 1 : Exemplary Modified Human C3 Proteins
Figure imgf000034_0001
Figure imgf000035_0001
[00121] In some embodiments, the modified human C3 protein is substantially the same number of amino acid residues as an unmodified human C3 protein. In some embodiments, the modified human C3 protein has additional non-C3 and non-CVF amino acids attached to the carboxy terminus of the protein.
[00122] The modified human C3 proteins of the present invention are in any form. In some embodiments, the modified human C3 protein has additional non-C3 and non-CVF amino acids attached to the carboxy terminus of the protein. In some embodiments, the modified C3 protein is in a single chain form comprising the signal sequence or in a single chain form with the signal sequence being cleaved. The modified C3 protein is in a two-chain form that resembles the structure of human C3 as shown in Figure 1. In some embodiments, the modified human C3 protein is in a form that resembles human C3b, wherein the C3a portion is cleaved. In some embodiments, the cleavage sites are not the cleavage sites for human C3 proteins (e.g., depending on the host cells). For example, one, two, or more amino acids on either end of the cleavage site of human C3 are cleaved for the modified C3 protein.
[00123] In some embodiments, the catalytic activity of a convertase containing the modified human C3 protein is at least 50% that of the convertase containing CVF. In some embodiments, the catalytic activity of a convertase containing the modified human C3 protein is greater than that of the convertase containing native human C3. In some embodiments, the catalytic activity is about any of 60%, 70%, 80% 90% or 100% that of the CVF convertase. In some embodiments, the invention provides convertases containing the modified human C3 protein having a catalytic activity that falls between the two, or that exceeds the activity of the convertase containing native human C3. Thus, the invention additionally provides convertases containing the modified human C3 protein having catalytic activity from about 10% to about 1000%, or more, that of the convertase containing CVF, including but not limited to about any of 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 110%, 135%, 150%, 200%, 300%, 400%, 500%, 750%, 1000% and more.
[00124] Based on the kcat/Km, the catalytic efficiency is approximately eight-fold greater for C3bBb compared to CVFBb when cleaving C3. In some embodiments, the catalytic efficiency of a convertase containing the modified human C3 protein is at least 50% that of the convertase containing CVF, and is greater than that of the convertase containing native human C3b. In some embodiments, the invention also provides convertases containing the modified human C3 protein having a catalytic efficiency that falls between the two, or that exceeds the efficiency of the convertase containing native human C3b. Thus, the invention additionally provides convertases containing the modified human C3 protein having catalytic efficiency from about 10% to about 1000%, or more, that of the convertase containing CVF, including but not limited to about any of 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 110%, 135%, 150%, 200%, 300%, 400%, 500%, 750%, 1000% and more.
[00125] In some embodiments, the C5 cleaving activity of the modified human C3 proteins is decreased as compared to native human C3. In some embodiments, the C5 cleaving activity is from non-detectable to about 50% of the activity native human C3, including but not limited to about any of 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, and 50%.
[00126] In some embodiments, the binding affinity of the modified human C3 proteins to Factor B and/or its subsequent cleavage by factor D is reduced. However, as long as at least a functional amount of the catalytic activity and the complement-depleting activity remain, the modified human C3 protein is useful.
[00127] In some embodiments, the methods described herein comprise administering to an individual in need thereof a convertases having the modified human C3 proteins exhibiting substantially the same complement-activating activity of those containing natural CVF. The term "exhibit substantially the same complement-activating activity of natural CVF" means that the modified human C3 protein of the present invention have from about 5% to about 100%, from about 50 to about 97%, from about 80 to about 97% of the level of the complement activating activity of natural CVF as measured by the method of Cochran et al., (1970) J. Immunol. 105(1), 55-69.
[00128] In some embodiments, the modified human C3 proteins disclosed herein have immunogenicity less than that of CVF. In some embodiments, the modified human C3 protein is substantially non-immunogenic. In some embodiments, the modified human C3 protein are as non-immunogenic as C3, or is at least about any of 50%, 60%, 70%, 80%, 90% less immunogenic than CVF. In some embodiments, immunogenicity is measured by any suitable method.
[00129] In some embodiments, the intrinsic half- life of the convertase formed with the modified human C3 protein is greater than about 1.5 minutes or greater than about 10 minutes. In some embodiments, the intrinsic half-life is between that of the CVF-containing convertase (7 hours or longer) and that of native human C3 (1.5 minutes), including but not limited to about any of: 2 minutes, 10 minutes, 20 minutes, 30 minutes, 40 minutes, 50 minutes, 60 minutes, 90 minutes, 2 hours, 2.5 hours, 3 hours, 3.5 hours, 4 hours, 4.5 hours, 5 hours, 5.5 hours, 6 hours, 6.5 hours, 7 hours, 8 hours, 9 hours, 10 hours, 10.5 hours, or more.
[00130] In some embodiments, the resistance of the modified human C3 proteins to factors H and/or I is greater than that for native human C3. In some embodiments, the modified human C3 proteins have about the same level of resistance to factors H and/or I as that of CVF. Methods of Producing Modified Human C3 Proteins
[00131] In some embodiments, a polynucleotide containing the nucleic acid sequence encoding of any of the modified human C3 proteins described herein is cloned into a vector. In some embodiments, suitable cloning vectors are constructed by any suitable manner, or are selected from a large number of cloning vectors commercially available. In some embodiments, cloning vectors have the ability to self-replicate, possess a single target for a particular restriction endonuclease, and/or carry genes for a marker for use in selecting clones containing the vector. Suitable examples include plasmids and bacterial viruses, e.g., pUC18, pUC19, Bluescript (e.g., pBS SK+) and its derivatives, mpl8, mpl9, pBR322, pMB9, CoIEl, pCRl, RP4, phage DNAs, and shuttle vectors such as pSA3 and pAT28. Examples of vectors for expression in Drosophila cells are pMT/BiP-V5-HisA, pMT/BiP-V5-HisB, and pMT/BiP-V5-HisC. Example of vectors for expression in Pichia pastoris are pPICZA, pPICZB, pPICZC, pPICZf -A, pPICZf -B, and pPICZf -C. These and many other cloning vectors are available from commercial vendors such as BioRad, Strategene, and Invitrogen. Suitable vectors also include mammalian expression vectors (e.g., pCDNA2, as pcDNA3.1, or pSecTag2 cloning vectors).
[00132] Expression vectors generally are replicable polynucleotide constructs that contain a polynucleotide according to the invention. It is implied that an expression vector must be replicable in the host cells either as episomes or as an integral part of the chromosomal DNA. Suitable expression vectors include but are not limited to plasmids, viral vectors, including adenoviruses, adeno-associated viruses, retroviruses, cosmids, and expression vector(s) disclosed in PCT Publication No. WO 87/04462. In certain instances, vector components include, but are not limited to, one or more of the following: a signal sequence; an origin of replication; one or more marker genes; suitable transcriptional controlling elements (such as promoters, enhancers and terminator). For expression (i.e., translation), one or more translational controlling elements are also usually required, such as ribosome binding sites, translation initiation sites, and stop codons.
[00133] In some embodiments, the vectors containing the polynucleotides of interest are introduced into the host cell by any by any suitable method, including electroporation, transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; and infection (e.g., where the vector is an infectious agent such as vaccinia virus). The choice of introducing vectors or polynucleotides will often depend on features of the host cell.
[00134] In some embodiments, the host cell is any suitable host cells (i.e., a cell capable of over- expressing heterologous DNAs). Non-limiting examples of mammalian host cells include but not limited to COS, HeLa, and CHO cells. See also PCT Publication No. WO 87/04462. Suitable non-mammalian host cells include prokaryotes (such as E. coli or B. subtillis) and yeast (such as S. cerevisae, S. pombe, P. pastoris, or K. lactis). Other examples of host cells include, but are not limited to, Drosophila S2 cell, a Sf9 cell, a HiFive cell, a BHK cell, and an HEK293 cell.
[00135] In some embodiments, a modified human C3 protein is produced by culturing the host cell comprising the polynucleotides under a condition that the modified human C3 protein is expressed; and purifying the modified human C3 protein. In some embodiments, the modified human C3 protein is purified from the culture medium of the host cell. The invention also provides modified human C3 proteins produced by the method. In some embodiments, the modified C3 protein produced is in more than one form (e.g., the protein produced is a mixture containing both single and double forms).
• Methods of removing excess N-terminal residues
[00136] In certain instances, an HC3-1496 protein contains 19 additional amino acid residues on the N-terminus of the β-chain that are an artifact of the cloning procedure. In certain instances, the coding sequence for the protein was engineered so that the signal sequence was removed, and a unique Afel site was engineered in to facilitate cloning of the insert into blunt-ended cloning sites. In certain instances, in the S2 expression vector (pMT/BiP-V5-HisA), an EcoRV site in the multiple cloning site region of the vector is used. In certain instances, the cleavage site for the signal protease is coded for by the vector, resulting in an HC3-1496 protein as expressed in S2 cells having an additional 19 amino acids on the N-terminus of the protein, coded for by the vector between the signal cleavage site and the cloning site. When the same expression cassette is cloned into pSecTag2/Hygro, in certain instances, there are 27 vector-coded residues on the N-terminus of the protein.
[00137] In some embodiments, an HC3- 1496 protein with a mature, C3-like N-terminus is obtained by utilizing a version of an HC3-1496 protein that is expressed with the native, human C3 signal sequence. In certain instances, the mammalian C3 is produced and exported efficiently in mammalian cells in vivo. In some embodiments, a construct containing the native signal sequence is exported in mammalian expression systems (e.g., CHO cells). In some embodiments, a plasmid used in a method disclosed herein encodes (a) HC3-1496 and (b) the native signal sequence. In some embodiments, the coding sequence further comprises a restriction site on the 5' end (e.g., to facilitate easy transfer to an expression vector). In some embodiments, the coding sequence is engineered to comprise a restriction site on the 5' end by any suitable method (e.g., PCR).
• Methods of selecting for one HC3-1496 form
[00138] In some embodiments, an HC3-1496 protein is produced as two forms: (a) a two-chain form resembling mature C3 or "C3-like" form; and (a) a form resembling an activated C3 or "C3b- like" form, in which 77 amino acid residues (the C3a domain) are removed from the N-terminus of the larger (α) chain of the protein. In some embodiments, the C3b-likeform accounts for about 40% to about 70% of the protein. In some embodiments, a small amount of a single chain form is present. In some embodiments, the growth conditions of the CHO cells are optimized for the production of one form of the protein. In some embodiments, the C3a domain is removed and the construct comprises the mature N-terminus of the α' -chain directly after the four arginines that constitute a furin-cleavage site (e.g., the furin site that is efficiently cleaved both in S2 cells and in vivo). In some embodiments, the site where S2 cells cleave the protein is modified by any suitable technique. Increasing the stability of human C3/CVF hybrid proteins
[00139] In certain instances, injection of CVF into animals results in relatively long-term serum complement depletion (three to four days, depending on species). In some embodiments, the complement depletion caused by several hybrid human C3-CVF proteins is much shorter in duration (e.g., 24 hours or less).
[00140] Disclosed herein, in some embodiments, is a more inherently stable hybrid human C3-CVF protein. In some embodiments, the hybrid human C3-CVF protein is any protein disclosed herein (e.g., HC3-1496 or analogues thereof). In some embodiments, the hybrid human C3-CVF protein is more resistant to dissociation of the C3b-like moiety and Bb. In some embodiments, the increased stability affects a longer biological life in depleting complement.
[00141] Disclosed herein, in some embodiments, is a hybrid human C3-CVF protein-containing convertase that is nearly as stable as the CVF-containing enzyme. In some embodiments, the hybrid human C3-CVF protein has reduced affinity for factor H. In some embodiments, reducing the affinity of hybrid human C3-CVF protein for factor H increases the stability of the convertase and of the protein itself.
[00142] In some embodiments, a hybrid human C3-CVF protein with increased stability is prepared by any suitable method (e.g., modification of one or more amino acids (e.g., by PCR)). In some embodiments, an amino acid is modified in a domain selected from: MGl, MG2, MG6, alpha- NT', TED, MG7, or combinations thereof. In some embodiments, the amino acid that is modified is Glutamine (Q) 163 of human C3. In some embodiments, Glutamine (Q) 163 of human C3 is deleted. In some embodiments, Glutamine (Q) 163 of human C3 is deleted, the amino acid residue immediately preceding Glutamine 163 is deleted, and the three amino acid residues immediately following Glutamine 163 are deleted. In some embodiments, the amino acid that is modified is Serine (S) 1075 of human C3. In some embodiments, Serine (S) 1075 of human C3 is replaced with Histidine (H). In some embodiments, Serine (S) 1075 of human C3 is replaced with Histidine (H) and the two preceding amino acid residues are removed. In some embodiments, the amino acid that is modified is Asparagine (N) 738 of human C3. In some embodiments, Asparagine (N) 738 of human C3 is replaced with Aspartic Acid. In some embodiments, Asparagine (N) 738 of human C3 is replaced with Aspartic Acid and the two preceding Glutamic acid residues (EE) are replaced with aspartic acid and serine (DS). In some embodiments, Asparagine (N) 738 of human C3 is replaced with Aspartic acid and the immediately preceding Glutamic acid (E) is replaced with Serine (S). In some embodiments, a Glutamine (Q) at position 1139 of human C3 is modified into a Lysine or to a Tyrosine. The aforementioned amino acids are identified according to mature C3b numbering. In some embodiments, a hybrid human C3-CVF protein is modified such that some of the residues of C3b that interact with factor H are replaced with sequences from CVF that interact with factor H by any suitable method (e.g., by analyzing a crystal structure). In some embodiments, the hybrid protein is more stable to cleavage in the presence of factors H and I. In some embodiments, the hybrid protein has an increased life in vivo and in vitro. Complement Component 1 Modulators
[00143] In some embodiments, a complement component 1 (Cl) modulator is administered to an individual in need thereof. In some embodiments, the complement Cl modulator is a Cl inhibitor. In certain instances, the C 1 inhibitor prevents fluid-phase C 1 activation. In certain instances, administration of C 1 inhibitor prevents reperfusion injury. In some embodiments, dextran sulfate is administered to an individual in need thereof. In some embodiments, Cl inhibitor is administered before, after, or simultaneous with dextran sulfate. In certain instances, dextran sulfate potentiates C 1 inhibitor.
[00144] In some embodiments, a complement component Iq receptor (CIqR) is administered to an individual in need thereof. In certain instances, CIq regulates the presentation of adhesion molecules on endothelial cells. In certain instances, a CIq receptor (e.g. cClqR, ClqRp, and gClqR) prevents complement-mediated lysis of CIq sensitized erythrocytes. In certain instances, administration of Cl inhibitor prevents reperfusion injury.
[00145] In some embodiments, an antagonist of CIq binding is administered to an individual in need thereof. In some embodiments, the antagonist of CIq binding is CIq inhibitor, decorin, CSPG (chondroitin sulfate proteoglycan), CBP2 (complement binding peptide T), or combinations thereof. In certain instances, CSPG partially or fully inhibits the binding of CIq to CIs and CIr, thereby interfering with the formation of the enzyme Cl. In certain instances, CBP2 interferes with the binding of CIq to an antigen or antigen-bound antibody. Complement Receptor 1
[00146] In some embodiments, a complement receptor 1 (CRl) is administered to an individual in need thereof. By binding to C3b and C4b CRl promotes phagocytosis and clearance of antigen- antibody complexes. Further, it inhibits both the classic and alternative pathways. In certain instances, CRl acts as a decay-accelerator for both C3 and C5. Additionally, in certain instances, CRl acts as a Factor I cofactor.
[00147] In some embodiments, a soluble CRl (sCRl) is administered to an individual in need thereof. Soluble CRl lacks the transmembrane and cytoplasmic domains of CRl. In certain instances, sCRl decrease the amount of MAC produced by the complement system. In certain instances, sCRI ameliorates ischemic/reperfusion injuries. In certain instances, sCRl reduces cellular and tissue injuries in animal models with acute or chronic inflammatory disorders. In some embodiments, the sCRl is APT070 (Mirococept), TPlO (Avant Immunotherapeutics), TP20 (Avant Immunotherapeutics), or combinations thereof.
[00148] In some embodiments, a soluble CRl (sCRl) lacking the long homologous repeat A (LHR-A) domain (sCRl [desLHR-A]) is administered to an individual in need thereof. sCRl [desLHR-A] lacks the transmembrane and cytoplasmic domains of CRl and the C4b binding domain. In certain instances, sCRl [desLHR-A] inhibits the alternative pathway but exhibits a diminished ability to inhibit the classical pathway as compared to sCRl.
[00149] In some embodiments, a soluble CRl (sCRl) bound by SLe x moieties is administered to an individual in need thereof. SLex is a carbohydrate ligand for selectins that, in certain instances, inhibits E-selectin and P-selectin mediated neutrophil adhesion. In certain instances, sCRl-SLex inhibits complement activation and inhibits the recruitment of neutrophils to the site of inflammation. Complement Receptor 1 -Related Gene/Protein y
[00150] In some embodiments, a complement receptor 1 -related gene/protein y (Crry) is administered to an individual in need thereof. In some embodiments, a recombinant Crry (Crry-Ig) is administered to an individual in need thereof. Crry inhibits both the classic and alternative pathways. In certain instances, Crry acts as a decay-accelerator for both C3 and C5. Additionally, in certain instances, Crry acts as a Factor I cofactor. Complement Component 3 Convertase Modulators
[00151] In some embodiments, a modulator of C3 convertase is administered to an individual in need thereof. In some embodiments, the C3 convertase modulator is a fucan. In some embodiments, a fucan is extracted from brown seaweed (e.g. Phaeophyceae, Ascophyllum nodosum, and Ecklonia kurome). In certain instances, a fucan partially or fully suppresses the classical pathway. In certain instances, a fucan partially or fully suppresses the alternative pathway. In some embodiments, the fucan is BS8. In certain instances, BS8 partially or fully inhibited formation of C4bC2a by interfering with Cl activation. In certain instances, BS8 partially or fully inhibited formation of C4bC2a by interfering C4 cleavage. In certain instances, BS8, partially or fully inhibits C3Bb by interfering with the binding of Factor B to C3b and by interfering with the binding of properdin.
[00152] In some embodiments, a modulator of C3 convertase is administered to an individual in need thereof. In some embodiments, the C3 convertase modulator is complestatin. In certain instances, complestatin interferes with the binding of C4b and C2b, and thus antagonizes the formation of the classical C3 convertase (C4bC2b).
[00153] In some embodiments, a modulator of C3 convertase is administered to an individual in need thereof. In some embodiments, the C3 convertase modulator is extracellular complement- binding protein (Ecb). In certain instances, Ecb is isolated from S. aureus. In certain instances, it modulates C3b containing molecules (e.g. the alternative C3 convertase C3bB3, and the C5convertases C4bC2aC3b and C3bBbC3b) by blocking the ability of C3b containing molecules to cleave their substrates (e.g. C3 and C5).
[00154] In some embodiments, a modulator of C3 convertase is administered to an individual in need thereof. In some embodiments, the C3 convertase modulator is extracellular fibrinogen-binding protein (Efb). In certain instances, Efb is isolated from S. aureus. In certain instances, Efb modulates C3b containing molecules (e.g. the alternative C3 convertase C3bB3) by blocking the ability of C3bBb to cleave C3.
[00155] In some embodiments, a modulator of C3 convertase is administered to an individual in need thereof. In some embodiments, the C3 convertase modulator is compstatin. In certain instances, compstatin antagonizes C3 convertases by binding to C3 and partially or fully inhibiting the ability for a C3 convertase to bind to and cleave C3.
[00156] In some embodiments, a modulator of C3 convertase is administered to an individual in need thereof. In some embodiments, the C3 convertase modulator is rosmarinic acid. In certain instances, rosmarinic acid reacts with the activated thioester of metastable C3b. In certain instances, the reaction of rosmarinic acid and the activated thioester of metastable C3b results in covalent attachment of rosmarinic acid to a C3 convertase. In certain instances, the covalent attachment of rosmarinic acid to a C3 convertase prevents the binding of a C3 convertase to a host cell or pathogen.
[00157] In some embodiments, a modulator of C3 convertase is administered to an individual in need thereof. In some embodiments, the C3 convertase modulator is CRIT. In some embodiments, the C3 convertase modulator is a peptide sequence comprising the C-terminal 11 -amino-acid of the first CRIT-extracellular domain (CRIT-H 17). In certain instances, CRIT inhibits the formation of C3 convertase. In certain instances, CRIT binds to C2, thus inhibiting C4b from binding to C2 and forming C3 convertase.
[00158] In some embodiments, a modulator of C3 convertase is administered to an individual in need thereof. In some embodiments, the C3 convertase modulator is glycyrrhetinic acid. In certain instances, glycyrrhetinic acid modulates C2 and thus modulates the formation of the classical pathway C3 convertase. Complement Component 5 Convertase Modulators
[00159] In some embodiments, a modulator of C5 convertase is administered to an individual in need thereof. In some embodiments, the C5 convertase modulator is an anti-complement component 5 (C5) murine monoclonal. In certain instances, an anti-C5 mAb partially or fully inhibits the cleavage of C5 by C5 convertase. In certain instances, an anti-C5 mAB inhibits the formation of C5a. In certain instances, an anti-C5 mAb partially or fully inhibits the formation of C5b and thus the formation of a MAC. In certain instances, an anti-C5 mAB does not inhibit the cleavage of a C3. In some embodiments, an anti-C5 mAB is derived from the variable region of the N 19/8 mAb. In certain instances, administration of an anti-C5 mAB ameliorates an autoimmune disease. In certain instances, an anti-C5 mAB partially or fully inhibits CDl Ib up- regulation. In certain instances, decreases the number of P-selectin presenting platelets. In certain instances, an anti-C5 mAB reduces the formation of leukocyte -platelet aggregates. In some embodiments, the C5 antibody is pexelizumab.
[00160] In some embodiments, an anti-C5 murine single-chain antibody is administered to an individual in need thereof. In some embodiments, an anti-C5 murine single-chain antibody is derived from the variable region of the N 19/8 mAb. In certain instances, an anti-C5 murine single-chain antibody is inhibits the cleavage of C5 and the production of C5a. In certain instances, an anti-C5 murine single-chain antibody is partially or fully inhibits C5b-9-mediated hemolysis of erythrocytes. In some embodiments, an anti-C5 humanized single-chain antibody (e.g. 5Gl.1-SC) is administered to an individual in need thereof.
[00161] In some embodiments, a modulator of C5 convertase is administered to an individual in need thereof. In some embodiments, the C5 convertase modulator is K76 (6,7-diformyl- 3',4',4a',5',6',7',8',8a'-octahydro-4,6',7'-trihydroxy-2',5',5',8a'-tetrame thyl spiro[l'(2'H)- naphthalene-2(3M)-benzofuran]), or a derivative thereof (e.g. TKIXc, and K76 COOH). In certain instances, K76 antagonizes C5 convertase by interfering with the ability of C5 convertase to bind to and/or cleave C5.
[00162] In some embodiments, a modulator of C5 convertase is administered to an individual in need thereof. In some embodiments, the C5 convertase modulator is a staphylococcal complement inhibitor (e.g. SCIN, SCIN-B, and SCIN-C). In certain instances, a staphylococcal complement inhibitor is isolated from S. aureus. In certain instances, an SCIN binds to and stabilizes a C3 convertase (e.g. C4bC2a and C3bBb). In certain instances, the binding of an SCIN prevents the binding of a C3b subunit to the complex; thus, preventing the formation of a C5 convertase from a C3 convertase. CD55
[00163] In some embodiments, a CD55 is administered to an individual in need thereof. CD55, also known as Decay Accelerating Factor (DAF), binds both C4b and C3b. In certain instances, the binding of CD55 to C4b disassociates the C3 convertase of the classical pathway and thus also inhibits the formation of the classical C5 convertase. In certain instances, the binding of CD55 to C3b disassociates the C3 and C5 convertases of the alternative pathway. In some embodiments, a CD55 protein is a soluble protein (sCD55). In some embodiments, sCD55 is administered to an individual in need thereof. CD59
[00164] In some embodiments, a CD59 protein is administered to an individual in need thereof. In certain instances, CD59 inhibits the formation of a MAC by binding to C8 and C9 and thereby preventing their binding to the C5bC6C7 complex. In some embodiments, a soluble CD59 (sCD59) protein is administered to an individual in need thereof.
CD55/CD59 Fusion Proteins
[00165] In some embodiments, a CD59/CD55 fusion protein is administered to an individual in need thereof. In certain instances, the CD59 subunit inhibits the formation of a MAC by binding to C8 and C9 and thereby preventing their binding to the C5bC6C7 complex. In certain instances, a CD59/CD55 fusion protein prevents the formation of a MAC, and prevents the formation of or inhibits the activity of a C5 convertase. In certain instances, the CD55 subunit binds to C4b thereby disassociating the C3 convertase of the classical pathway and inhibiting the formation of the classical C5 convertase. In certain instances, the CD55 subunit binds to C3b thereby disassociating the C5 convertase of the alternative pathway.
CD55/MCP Fusion Proteins
[00166] In some embodiments, a CD55/MCP fusion protein is administered to an individual in need thereof. In certain instances, the CD55 subunit binds to C4b thereby disassociating the C3 convertase of the classical pathway and inhibiting the formation of the classical C5 convertase. In certain instances, the CD55 subunit binds to C3b thereby disassociating the C5 convertase of the alternative pathway. In certain instances, the MCP (Membrane Cofactor Protein, or CD46) subunit is a co-factor of Factor I. In certain instances, the MCP subunit activates Factor I leading to the inactivation of a C3 convertase of the classical pathway and/or a C3 convertase of the alternative pathway. In some embodiments, the CD55/MCP fusion protein is a soluble protein sCD55/MCP (Complement Activation Blocker-2, CAB-2). In certain instances, CAB-2 exhibits greater antagonism of convertases (e.g. C3 and C5) as compared to either CD55 administered alone, MCP administer alone, or CD55 and MCP administered in combination. In certain instances, CAB-2 inhibits complement activation in vivo.
Factor D Modulators
[00167] In some embodiments, a Factor D modulator is administered to an individual in need thereof. In some embodiments, the Factor D modulator is a Factor D antagonist. In some embodiments, the Factor D antagonist is BCX- 1470 (2-amidino-6-(2-thiophene carboxy)benzothiophene methanesulfonate); FUT- 175 (6-amidino-2-naphthyl p-guanidinobenzoate dimethane- sulphonate); or combinations thereof. In certain instances, Factor D antagonists inhibit the formation of the alternative pathway fluid phase C3 convertase by antagonizing Factor D's ability to bind to and cleave Factor B.
Factor I and Factor I Co-Factors
[00168] In some embodiments, a Factor I protease and a co-factor thereof are administered to an individual in need thereof. In certain instances, Factor I when bound to a co-factor, cleaves C3b and/or C4b; thus, inactivating them. The inactivation of C4b (iC4b) inhibits the activity of a C3 convertase of the classical pathway and thus also inhibits the formation of the classical C5 convertase. Further, the inactivation of C3b (iC3b) inhibits the activity of a C3 and C5 convertases of the alternative pathway.
[00169] In some embodiments, a Membrane Cofactor Protein (MCP OR CD46) is administered to an individual in need thereof. MCP is a co-factor of Factor I. In some embodiments, MCP is administered to an individual in need thereof in soluble form (sMCP). In some embodiments, sMCP and/or MCP is administered before, after, or simultaneously with Factor I. In some embodiments, sMCP and/or MCP is administered with CD55. In certain instances, administration of sMCP inhibits complement-mediated inflammation. In certain instances, administration of MCP activates Factor I leading to the inactivation of a C3 convertase of the classical pathway and/or a C3 convertase of the alternative pathway. In certain instances, administration of MCP activates Factor I leading to a decrease in the production of C5 convertase the classical pathway and/or inactivation of a C5 convertase of the alternative pathway. Factor H Modulators
[00170] In some embodiments, a Factor H modulator is administered to an individual in need thereof. In certain instances, Factor H serves as a co-factor for Factor I which cleaves C3b and/or C4b; thus, inactivating them. The inactivation of C4b (iC4b) inhibits the activity of a C3 convertase of the classical pathway and thus also inhibits the formation of the classical C5 convertase. Further, the inactivation of C3b (iC3b) inhibits the activity of a C3 and C5 convertases of the alternative pathway. In some embodiments, the Factor H modulator is a Factor H mimic, an agent that that increases the concentration of Factor H, or a combination thereof. In some embodiments, the Factor H modulator is TT30 (Taligen Therapeutics). Factor B Modulators
[00171] In some embodiments, a Factor B modulator is administered to an individual in need thereof. In certain instances, the C3b is bound by Factor B. In certain instances, when bound to C3b, Factor B is cleaved by Factor D into Ba and Bb. In certain instances, Bb remains bound to C3b forming an unstable C3 protease (C3bBb). In certain instances, C3b binds to the membrane of an antigen presenting cell. In certain instances, the binding of C3b to an antigen presenting cell facilitates opsonization of the antigen presenting cell. In some embodiments, the Factor B modulator is an antibody or a portion thereof, that binds to Factor B. In some embodiments, the Factor B modulator is TAl 06 (Taligen Therapeutics). Heparin and Thrombin Inhibitors
[00172] In some embodiments, heparin or inhibitors of thrombin or a derivative thereof (e.g. LU
51198) is administered to an individual in need thereof. In certain instances, heparin interacts with Cl, C2, C3, C4, C5, C6, Cl, C8, C9, ClINH, factor I, factor H, factor B and factor P. In certain instances, heparin or inhibitors of thrombin partially or fully inhibits the formation of the alternative pathway C3 convertase (C3bBb),the classical pathway C3 convertase (C4bC2a) or all forms of C5 convertase (e.g., C3bBbC3b, C4bC2aC3b).
MAC Modulators
[00173] In some embodiments, an MAC modulator is administered to an individual in need thereof. In some embodiments, the MAC modulator is clusterin, vitronectin. In certain instances, clusterin partially or fully inhibits the formation of fluid-phase MAC. In certain instances, vitronectin partially or fully inhibits the formation of fluid-phase MAC.
Properdin Antibodies
[00174] In some embodiments, an anti-properdin antibody is administered to an individual in need thereof. In some embodiments, the anti-properdin antibody is a monoclonal antibody. In certain instances, an anti-properdin inhibits the stabilization of the alternative pathway unstable C3 convertase (C3Bb). In certain instances, an anti-properdin antibody inhibits the formation of the alternative pathway C5 convertase (C3BbC3). In certain instances, an anti-properdin antibody inhibits the formation of MAC.
Miscellaneous Complement Modulators
[00175] In some embodiments, the complement modulator is glycyrrhizin, glycyrrhetinic acid, or combinations thereof. In certain instances, glycyrrhetinic acid modulates C2 and thus modulates the formation of the classical pathway C3 convertase.
VIII. Production of Monoclonal Antibodies
Murine Antibodies
[00176] In certain instances, monoclonal antibodies (mAb) produced via the use of a hybridoma. In certain instances, a hybridoma is an immortalized antibody producing cell. In certain instances, a laboratory animal (e.g., a mouse or a rabbit) is inoculated with an antigen. In certain instances, B-cells from the laboratory animal's spleen are extracted. In certain instances, a hybridoma is generated by fusing (1) an extracted B-cell with (2) a myeloma cell (i.e., hypoxanthine-guanine- phosphoribosyl transferase negative, immortalized myeloma cells). In certain instances, the B- cell and the myeloma cells are cultured together and exposed to an agent that renders their cell membranes more permeable (e.g., PEG).
[00177] In certain instances, the culture comprises a plurality of hybridoma, a plurality of myeloma cells, and a plurality of B-cells. In certain instances, the cells are individual to culturing conditions that select for hybridoma (e.g., culturing with HAT media).
[00178] In certain instances, an individual hybridoma (i.e., the clone) is isolated and cultured. In some embodiments, the hybridoma are injected into a laboratory animal. In some embodiments, the hybridoma are cultured in a cell culture. Humanized Antibodies [00179] In some embodiments, the methods described herein comprise a humanized monoclonal antibody. In some embodiments, a humanized monoclonal antibody comprises heavy and light chain constant regions from a human source and variable regions from a murine source.
[00180] In some embodiments, humanized immunoglobulins, including humanized antibodies, are constructed by genetic engineering. In some embodiments, humanized immunoglobulins comprise a framework that is identical to the framework of a particular human immunoglobulin chain (i.e., an acceptor or recipient), and three CDRs from a non-human (donor) immunoglobulin chain. In some embodiments, a limited number of amino acids in the framework of a humanized immunoglobulin chain are identified and chosen to be the same as the amino acids at those positions in the donor rather than in the acceptor.
[00181] In some embodiments, a framework is used from a particular human immunoglobulin that is homologous to the donor immunoglobulin to be humanized. For example, comparison of the sequence of a mouse heavy (or light) chain variable region against human heavy (or light) variable regions in a data bank (for example, the National Biomedical Research Foundation Protein Identification Resource or the protein sequence database of the National Center for Biotechnology Information - NCBI) shows that the extent of homology to different human regions can vary greatly, for example from about 40% to about 60%, about 70%, about 80%, or higher. By choosing as the acceptor immunoglobulin one of the human heavy chain variable regions that is most homologous to the heavy chain variable region of the donor immunoglobulin, fewer amino acids will be changed in going from the donor immunoglobulin to the humanized immunoglobulin. By choosing as the acceptor immunoglobulin one of the human light chain variable regions that is most homologous to the light chain variable region of the donor immunoglobulin, fewer amino acids will be changed in going from the donor immunoglobulin to the humanized immunoglobulin.
[00182] In some embodiments, a humanized immunoglobulin comprises light and heavy chains from the same human antibody as acceptor sequences. In some embodiments, a humanized immunoglobulin comprises light and heavy chains from different human antibody germline sequences as acceptor sequences; when such combinations are used, one can readily determine whether the VH and VL bind an epitope of interest using conventional assays (e.g., an ELISA). In some embodiments, the human antibody will be chosen in which the light and heavy chain variable regions sequences, taken together, are overall most homologous to the donor light and heavy chain variable region sequences. In some embodiments, higher affinity is achieved by selecting a small number of amino acids in the framework of the humanized immunoglobulin chain to be the same as the amino acids at those positions in the donor rather than in the acceptor.
[00183] Any suitable method of modifying a framework region is contemplated herein. In some embodiments, the relevant framework amino acids to change are selected based on differences in amino acid framework residues between the donor and acceptor molecules. In some embodiments, the amino acid positions to change are residues known to be important or to contribute to CDR conformation (e.g., canonical framework residues are important for CDR conformation and/or structure). In some embodiments, the relevant framework amino acids to change are selected based on frequency of an amino acid residue at a particular framework position (e.g., comparison of the selected framework with other framework sequences within its subfamily can reveal residues that occur at minor frequencies at a particular position or positions). In some embodiments, the relevant framework amino acids to change are selected based on proximity to a CDR. In some embodiments, the relevant framework amino acids to change are selected based on known or predicted proximity to the antigen-CDR interface or predicted to modulate CDR activity. In some embodiments, the relevant framework amino acids to change are framework residues that are known to, or predicted to, form contacts between the heavy (VH) and light (VL) chain variable region interface. In some embodiments, the relevant framework amino acids to change are framework residues that are inaccessible to solvent.
[00184] In some embodiments, amino acid changes at some or all of the selected positions are incorporated into encoding nucleic acids for the acceptor variable region framework and donor CDRs. In some embodiments, altered framework or CDR sequences are individually made and tested, or are sequentially or simultaneously combined and tested.
[00185] In some embodiments, the variability at any or all of the altered positions is from a few to a plurality of different amino acid residues, including all twenty naturally occurring amino acids or functional equivalents and analogues thereof. In some embodiments, non-naturally occurring amino acids are considered.
[00186] In some embodiments, the humanized antibody sequence is cloned into a vector. In some embodiments, any suitable vector is used. In some embodiments, the vector is a plasmid, viral e.g. 'phage, or phagemid, as appropriate. For further details see, for example, Molecular Cloning: a Laboratory Manual: 2nd edition, Sambrook et al., 1989, Cold Spring Harbor Laboratory Press. Many known techniques and protocols for manipulation of nucleic acid, for example in preparation of nucleic acid constructs, mutagenesis, sequencing, introduction of DNA into cells and gene expression, and analysis of proteins, are described in detail in Short Protocols in Molecular Biology, Second Edition, Ausubel et al. eds., John Wiley & Sons, 1992. The disclosures of Sambrook et al. and Ausubel et al. are incorporated herein by reference for such disclosure.
[00187] In some embodiments, any suitable host cell is transformed with the vector expressing the humanized antibody sequence. In some embodiments, the host cell is bacteria, mammalian cells, yeast and baculovirus systems. The expression of antibodies and antibody fragments in prokaryotic cells such as E. coli is well established in the art. For a review, see for example Pluckthun, A. Bio/Technology 9: 545-551 (1991). Expression in eukaryotic cells in culture is also available to those skilled in the art as an option for production of the antibodies and antigen-binding fragments described herein, see for recent reviews, for example Raff, M.E. (1993) Curr. Opinion Biotech. 4: 573-576; Trill JJ. et al. (1995) Curr. Opinion Biotech 6: 553- 560, each of which is which is incorporated herein by reference for such disclosure.
[00188] In some embodiments, a mammalian expression system is used. In some embodiments, the mammalian expression system is dehydrofolate reductase deficient ("dhfr- ") Chinese hamster ovary cells. In some embodiments, dhfr- CHO cells are transfected with an expression vector containing a functional DHFR gene, together with a gene that encodes a desired humanized antibody.
[00189] In some embodiments, DNA is transformed by any suitable method. For eukaryotic cells, suitable techniques include, for example, calcium phosphate transfection, DEAE Dextran, electroporation, liposome -mediated transfection and transduction using retrovirus or other virus, e.g., vaccinia or, for insect cells, baculovirus. For bacterial cells, suitable techniques include, for example, calcium chloride transformation, electroporation and transfection using bacteriophage.
[00190] In some embodiments, a DNA sequence encoding an antibody or antigen-binding fragment thereof is prepared synthetically rather than cloned. In some embodiments, the DNA sequence is designed with the appropriate codons for the antibody or antigen-binding fragment amino acid sequence. In general, one will select preferred codons for the intended host if the sequence will be used for expression. In some embodiments, the complete sequence is assembled from overlapping oligonucleotides prepared by standard methods and assembled into a complete coding sequence. See, e.g., Edge, Nature, 292:756 (1981); Nambair et al., Science, 223: 1299 (1984); Jay et al., J. Biol. Chem., 259:6311 (1984), each of which is which is incorporated herein by reference for such disclosure.
IX. Pharmaceutical Compositions and Methods of Administration
[00191] In certain embodiments, provided herein are pharmaceutical compositions comprising (a) an antibody; (b) a complement modulating agent; and (c) one or more physiologically acceptable carriers.
[00192] Physiologically acceptable carriers include excipients and auxiliaries which facilitate processing of the active agents into preparations which are used pharmaceutically. In certain instances, proper formulation is dependent upon the route of administration chosen. A summary of pharmaceutical compositions is found, for example, in Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H.A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins, 1999). [00193] Provided herein are pharmaceutical compositions comprising (a) an antibody; (b) a complement modulating agent; and (c) a pharmaceutically acceptable diluent(s), excipient(s), or carrier(s). In some embodiments, the pharmaceutical compositions includes other medicinal or pharmaceutical agents, carriers, adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure, and/or buffers. In addition, the pharmaceutical compositions also contain other therapeutically valuable substances.
[00194] A pharmaceutical composition refers to a mixture of an (a) an antibody; (b) a complement modulating agent; with (c) other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients. The pharmaceutical composition facilitates administration of an antibody and complement modulating agent to an organism. In practicing the methods of treatment or use provided herein, therapeutically effective amounts of an antibody are administered in a pharmaceutical composition to a mammal having a condition, disease, or disorder to be treated. Preferably, the mammal is a human. A therapeutically effective amount varies depending on the severity and stage of the condition, the age and relative health of the individual, the potency of the antibody used and other factors.
[00195] The pharmaceutical formulations described herein are optionally administered to an individual by multiple administration routes, including but not limited to, oral, parenteral (e.g., intravenous, subcutaneous, intramuscular), intranasal, buccal, topical, rectal, or transdermal administration routes. The pharmaceutical formulations described herein include, but are not limited to, aqueous liquid dispersions, self-emulsifying dispersions, solid solutions, liposomal dispersions, aerosols, solid dosage forms, powders, immediate release formulations, controlled release formulations, fast melt formulations, tablets, capsules, pills, delayed release formulations, extended release formulations, pulsatile release formulations, multiparticulate formulations, and mixed immediate and controlled release formulations.
[00196] Formulations suitable for intramuscular, subcutaneous, or intravenous injection include physiologically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Examples of suitable aqueous and non-aqueous carriers, diluents, solvents, or vehicles including water, ethanol, polyols (propyleneglycol, polyethylene-glycol, glycerol, cremophor and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate. Proper fluidity is maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. Formulations suitable for subcutaneous injection also contain optional additives such as preserving, wetting, emulsifying, and dispensing agents. [00197] For intravenous injections, an active agent is optionally formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. For other parenteral injections, appropriate formulations include aqueous or nonaqueous solutions, preferably with physiologically compatible buffers or excipients.
[00198] Parenteral injections optionally involve bolus injection or continuous infusion. Formulations for injection are optionally presented in unit dosage form, e.g., in ampoules or in multi dose containers, with an added preservative. In some embodiments, the pharmaceutical composition described herein are in a form suitable for parenteral injection as a sterile suspensions, solutions or emulsions in oily or aqueous vehicles, and contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Pharmaceutical formulations for parenteral administration include aqueous solutions of an active agent in water soluble form. Additionally, suspensions are optionally prepared as appropriate oily injection suspensions.
[00199] In some embodiments, the composition described herein are formulated for controlled or sustained release. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing a compound (e.g., modified human C3 proteins), which matrices are in the form of shaped articles, e.g. films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2- hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and 7 ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT TM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), sucrose acetate isobutyrate, and poly-D-(-)-3-hydroxybutyric acid. In some embodiments, the sustained release drug-delivery system is ATRIGEL® made by Atrix Laboratories. See, for example U.S. Pat. No. 6,565,874. The ATRIGEL® drug delivery system consists of biodegradable polymers, similar to those used in biodegradable sutures, dissolved in biocompatible carriers. In some embodiments, proteins are blended into this liquid delivery system at the time of manufacturing. In some embodiments, proteins are blended into this liquid delivery system by the physician at the time of use. When the liquid product is injected through a small gauge needle or placed into accessible tissue sites through a cannula, displacement of the carrier with water in the tissue fluids causes the polymer to precipitate to form a solid film or implant. Proteins encapsulated within the implant are then released in a controlled manner as the polymer matrix biodegrades with time. In some embodiments, the ATRIGEL® system releases proteins over multiple days. In some embodiments, the ATRIGEL® system releases proteins over multiple weeks. In some embodiments, the ATRIGEL® system releases proteins over multiple months. In some embodiments, injectable sustained release systems (e.g., such as ProLease®, Medisorb®, manufactured by Alkermes) are used.
[00200] In some embodiments, the methods and compositions described herein comprise a modified human C3 protein linked to another polypeptide or a carrier. For example, a modified human C3 protein is linked to an antibody or fragments thereof. In some embodiments, fusion proteins or conjugations are constructed by an suitable method. s
[00201] Disclosed herein, in some embodiments, are kits for use in the methods provided herein. In some embodiments, kits of the invention include one or more containers comprising any of the compounds (e.g., modified human C3 proteins) described herein and instructions for use in accordance with any of the methods of the invention described herein. In some embodiments, these instructions comprise a description of administration of the compound (e.g., modified human C3 proteins) to deplete complement according to any of the methods described herein. In some embodiments, the kit further comprises a description of selecting an individual suitable for treatment.
[00202] In some embodiments, the instructions relating to the use of a compound (e.g., modified human C3 proteins) include information as to dosage, dosing schedule, and route of administration for the intended treatment. In some embodiments, the containers are unit doses, bulk packages (e.g., multi-dose packages), sub-unit doses, or combinations thereof. In some embodiments, instructions supplied in the kits of the invention are typically written instructions on a label or package insert (e.g., a paper sheet included in the kit). In some embodiments, instructions supplied in the kits of the invention are machine-readable instructions (e.g., instructions carried on a magnetic or optical storage disk).
[00203] The kits of this invention are in suitable packaging. Suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like. In some embodiments, the kits comprise packages for use in combination with a specific device, such as an inhaler, nasal administration device (e.g., an atomizer) or an infusion device such as a minipump. In some embodiments, a kit has a sterile access port (e.g., the container is an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). In some embodiments, the container has a sterile access port (e.g., the container is an intravenous solution bag having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is a compound disclosed herein (e.g., modified human C3 proteins). In some embodiments, the container further comprises a second pharmaceutically active agent. [00204] In some embodiments, the kits disclosed herein provide additional components such as buffers and interpretive information. Normally, the kit comprises a container and a label or package insert(s) on or associated with the container.
[00205] For combination therapies of the invention, the kit contains instructions for administering the first and second therapies simultaneously and/or sequentially for the effective treatment of cancer. In some embodiments, the first and second therapies are present in separate containers or in a single container. In some embodiments, the kit comprises one distinct composition. In some embodiments, the kit comprises two or more compositions wherein one composition comprises a first therapy and one composition comprises a second therapy.
[00206] In some embodiments, depletion of complement is effected by an ex vivo treatment, for example, by transfusing circulation of an individual through a matrix bearing a complement- depleting compound (e.g., a modified human C3 protein) to remove complement. In some embodiments, the method further comprises a step of removing (e.g. , by dialysis) anaphylactic peptides (e.g., C5a) and other low molecular weight inflammatory mediators (e.g., histamine and nitric oxide) prior to the decomplemented blood (or plasma) being returned to the individual.
[00207] In some embodiments, the methods and compositions described herein comprises administering (a) a first therapy comprising an antibody and a complement modulating agent (e.g., a complement depleting agent and/or a C3b inhibiting agent) and (b) a second therapy useful for treating cancer. In some embodiments, the second therapy includes surgery, radiation, hormone therapy, gene therapy, antibody therapy (including, but not limited to any of the antibodies disclosed herein), and/or chemotherapy. ethods of Dosing and Treatment Regimens
[00208] Any of the compositions described herein are optionally used in the preparation of medicaments for the prophylactic and/or therapeutic treatment of conditions (e.g., an inflammatory disorder) that would benefit, at least in part, from amelioration. In some embodiments, an individual in need of treatment with any of the methods and compositions described herein has been diagnosed with an inflammatory disorder. In some embodiments, an individual in need of treatment with any of the methods and compositions described herein has been diagnosed with Atopic dermatitis; Acute disseminated encephalomyelitis; Acute leukocyte -mediated lung injury; Addison's disease; AIDS dementia; Allergic rhinitis; Ankylosing spondylitis; Antiphospholipid antibody syndrome; Alzheimer's disorder; Asthma; Adult respiratory distress syndrome; Autoimmune hemolytic anemia; Autoimmune hepatitis; Autoimmune inner ear disease; Behcet's syndrome; Bronchitis; Bullous pemphigoid; Chagas disease; Chronic obstructive pulmonary disease; Coagulative Necrosis; Coeliac disease; Crohn's disorder; Collagenous colitis; Conjunctivitis; Cystic fibrosis; Dermatomyositis; Dermatitis; Diabetes mellitus type 1; Diabetes mellitus type 2; Distal proctitis; Diversion colitis; Encephalitis; Endometriosis; Endotoxin shock; Eczema; Epilepsy; Fibromyalgia; Fibrinoid Necrosis; Goodpasture's syndrome; Gouty arthritis; Graves' disease; Guillain-Barre syndrome; Hashimoto's disease; Idiopathic thrombocytopenic purpura; Indeterminate colitis; Infective colitis; Inflammatory liver disorder; Interstitial cystitis; Ischaemic colitis; Liquefactive Necrosis; Lymphocytic colitis; Meningitis; Multiple sclerosis; Myasthenia gravis; Myocarditis; Narcolepsy; Nephritis; Pancreatitis; Parkinson's disorder; Pemphigus Vulgaris; Periodontal gingivitis; Pernicious anaemia; Polymyositis; Polymyalgia rheumatica; Psoriasis; Primary biliary cirrhosis; Retinitis; Rheumatoid arthritis; Rheumatoid spondylitis; Schizophrenia; Scleroderma; Shingles; Sjogren's syndrome; Smooth muscle proliferation disorders; Systemic lupus erythematosus (SLE); Tuberculosis; Ulcerative colitis; Uveitis; Vasculitis; Vitiligo; Wegener's granulomatosis; or combinations thereof.
[00209] In addition, a method for treating any of the diseases or conditions described herein in an individual in need of such treatment, involves administration of pharmaceutical compositions as described herein, or a pharmaceutically acceptable salt, pharmaceutically acceptable N-oxide, pharmaceutically active metabolite, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate thereof, in therapeutically effective amounts to said individual.
[00210] In some embodiments, a complement modulating agent is co-administered with an antibody (e.g., during the administration of the antibody, or within one day of administration of the antibody) for the entire duration of antibody therapy.
[00211] In some embodiments, a complement modulating agent is administered before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered one hour before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered two hours before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered three hours before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered four hours before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered five hours before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered six hours before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered eight hours before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered twelve hours before an antibody for the entire duration of antibody therapy.
[00212] In some embodiments, a complement modulating agent is administered before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered one day before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered two days before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered three days before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered four days before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered five days before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered six days before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered seven days before an antibody for the entire duration of antibody therapy. In some embodiments, a complement modulating agent is administered two weeks before an antibody for the entire duration of antibody therapy.
[00213] In some embodiments, the methods described herein comprise local administration of a synergistically-effective amount of (a) an antibody or fusion protein comprising an Fc domain; and (b) a complement depleting agent, an agent that inhibits the formation and/or activity of C3b, or a combination thereof. In some embodiments, the compositions disclosed herein are administered locally. In some embodiments, local treatment is effected by any suitable method, depending on the desired effect. In some embodiments, local treatment is effected by an autoinjector (e.g., a medical device that administers a device via a spring loaded syringe). In one embodiment, local depletion is effected when an effective amount of a complement-depleting compound (e.g., a modified human C3 protein) is administered locally to an organ, tissue, cavity, or intradermally. In some embodiments, this results in a temporary depletion of complement in the area. In some embodiments, local depletion is effected using an insulin-type pump that produces an intermittent or constant flow of the compound to a selected site. Alternatively, in some embodiments, local treatment employs a specific antibody which, when attached (e.g., chemically) to the compound localizes it to a specific tissue, a disease, or an infected cell to cause continuous depletion of complement in that area. In some embodiments, the method includes chemically linking the compound to an antibody (such as a monoclonal antibody) with an affinity for a specific tissue prior to the delivering step. In other embodiments, the antibody is attached to the compound (e.g., a modified human C3 protein) via recombinant DNA technology.
[00214] In some embodiments, the methods described herein comprise systemic administration of a synergistically-effective amount of (a) an antibody or fusion protein comprising an Fc domain; and (b) a complement depleting agent, an agent that inhibits the formation and/or activity of C3b, or a combination thereof. In some embodiments, the compositions disclosed herein are administered systemically. Systemic depletion is effected when an effective amount of a complement-depleting compound (e.g., a modified human C3 protein) is administered systemically, for example, intravenously or intraperitoneally. In some embodiments, this results in a temporary depletion of complement systemically.
[00215] In some embodiments, a composition described herein is administered once a day, once a week, semi-weekly, bi-weekly, once a month, semi-monthly, bi-monthly, every six months, or once a year. In some embodiments, a composition disclosed herein is administered via pulse therapy (e.g., a short, intensive administration of a composition disclosed herein, given at weekly intervals; or a portion of a standard dose administered at regular intervals until the full dose is reached). In some embodiments, a standard dose is divided into four sub-dose and each sub-dose is administered at four hour intervals.
[00216] Empirical considerations, such as the half-life, generally will contribute to determination of the dosage. For repeated administrations over several days or longer, depending on the condition, the treatment is sustained until a desired suppression of disease symptoms occurs or until sufficient therapeutic levels are achieved.
[00217] The compositions described herein are optionally administered before, during or after the occurrence of a disease or condition, and the timing of administering the composition varies in some embodiments. Thus, for example, a composition described here is used as a prophylactic and is administered continuously to individuals with a propensity to develop conditions or diseases in order to prevent the occurrence of the disease or condition. A composition described herein is optionally administered to an individual during or as soon as possible after the onset of the symptoms. The administration of the agents are optionally initiated within the first 48 hours of the onset of the symptoms, preferably within the first 48 hours of the onset of the symptoms, more preferably within the first 6 hours of the onset of the symptoms, and most preferably within 3 hours of the onset of symptoms. The initial administration is optionally via any route practical, such as, for example, an intravenous injection, a bolus injection, infusion over 5 minutes to about 5 hours, a pill, a capsule, transdermal patch, buccal delivery, and the like, or combination thereof. A composition described herein is preferably administered as soon as is practicable after the onset of a disease or condition is detected or suspected, and for a length of time necessary for the treatment of the disease, such as, for example, from about 1 month to about 3 months. The length of treatment optionally varies for each individual, and the length is then determined using the known criteria. For example, a composition described herein is administered for at least 2 weeks, preferably about 1 month to about 5 years, and more preferably from about 1 month to about 3 years.
[00218] In certain instances wherein the patient's condition does not improve, upon the doctor's discretion the administration of an antibody is optionally administered chronically, that is, for an extended period of time, including throughout the duration of the patient's life in order to ameliorate or otherwise control or limit the symptoms of the patient's disease or condition. [00219] In the case wherein the patient's status does improve, upon the doctor's discretion the administration of an antibody is optionally given continuously; alternatively, the dose of drug being administered is temporarily reduced or temporarily suspended for a certain length of time (i.e., a "drug holiday"). The length of the drug holiday optionally varies between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days. The dose reduction during a drug holiday includes from 10%- 100%, including, by way of example only, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%.
[00220] Once improvement of the patient's conditions has occurred, a maintenance dose is administered if necessary. Subsequently, the dosage or the frequency of administration, or both, is reduced, as a function of the symptoms, to a level at which the improved disease, disorder or condition is retained. In some embodiments, patients require intermittent treatment on a long- term basis upon any recurrence of symptoms.
[00221] In some embodiments, the pharmaceutical composition described herein is in unit dosage forms suitable for single administration of precise dosages. In unit dosage form, the formulation is divided into unit doses containing appropriate quantities of an antibody. In some embodiments, the unit dosage is in the form of a package containing discrete quantities of the formulation. Non- limiting examples are packaged tablets or capsules, and powders in vials or ampoules. In some embodiments, aqueous suspension compositions are packaged in single-dose non-reclosable containers. Alternatively, multiple-dose reclosable containers are used, in which case it is typical to include a preservative in the composition. By way of example only, formulations for parenteral injection are presented in unit dosage form, which include, but are not limited to ampoules, or in multi dose containers, with an added preservative.
[00222] In some embodiments, the dose of any of the compositions described herein is at least about 50 mg/kg body weight; at least about 20 mg/kg body weight; at least about 10 mg/kg body weight; at least about 5 mg/kg body weight; at least about 3 mg/kg body weight; at least about 2 mg/kg body weight; at least about 1 mg/kg body weight; at least about 750 μg/kg body weight; at least about 500 μg/kg body weight; at least about 250 ug/kg body weight; at least about 100 μg /kg body weight; at least about 50 μg /kg body weight; at least about 10 ug /kg body weight; at least about 1 μg/kg body weight, or more, is administered.
[00223] In some embodiments, the dose of any of the compositions described herein is about 25 mg/m , 30 mg/m , 50 mg/m , 60 mg/m , 75 mg/m , 80 mg/m , 90 mg/m , 100 mg/m , 120 mg/m2, 160 mg/m2, 175 mg/m2, 180 mg/m2, 200 mg/m2, 210 mg/m2, 220 mg/m2, 250 mg/m2, 260 mg/m2, 300 mg/m2, 350 mg/m2, 400 mg/m2, 500 mg/m2, 540 mg/m2, 750 mg/m2, 1000 mg/m2, or 1080 mg/m2 of a compound (e.g., modified human C3 proteins). In various embodiments, the composition includes less than about any of 350 mg/m2, 300 mg/m2, 250 mg/m2, 200 mg/m2, 150 mg/m2, 120 mg/m2, 100 mg/m2, 90 mg/m2, 50 mg/m2, or 30 mg/m2 of a compound (e.g., modified human C3 proteins).
[00224] In some embodiments, the effective amount of any of the compositions described herein is about 1 to about 5 mg/m2, about 5 to about 10 mg/m2, about 10 to about 25 mg/m2, about 25 to about 50 mg/m2, about 50 to about 75 mg/m2, about 75 to about 100 mg/m2, about 100 to about 125 mg/m , about 125 to about 150 mg/m , about 150 to about 175 mg/m , about 175 to about 200 mg/m2, about 200 to about 225 mg/m2, about 225 to about 250 mg/m2, about 250 to about 300 mg/m , about 300 to about 350 mg/m , or about 350 to about 400 mg/m .
[00225] The foregoing ranges are merely suggestive, as the number of variables in regard to an individual treatment regime is large, and considerable excursions from these recommended values are not uncommon. Such dosages are optionally altered depending on a number of variables, not limited to the activity of the antibody used, the disease or condition to be treated, the mode of administration, the requirements of the individual, the severity of the disease or condition being treated, and the judgment of the practitioner.
[00226] Toxicity and therapeutic efficacy of such therapeutic regimens are optionally determined in cell cultures or experimental animals, including, but not limited to, the determination of the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between the toxic and therapeutic effects is the therapeutic index, which is expressed as the ratio between LD50 and ED50. A composition described herein exhibiting high therapeutic indices is preferred. The data obtained from cell culture assays and animal studies are optionally used in formulating a range of dosage for use in human. The dosage of such a composition lies preferably within a range of circulating concentrations that include the ED50 with minimal toxicity. The dosage optionally varies within this range depending upon the dosage form employed and the route of administration utilized.
[00227] In some embodiments, rituximab is administered by slow perfusion at a dose of 375 mg/m body surface area. In some embodiments, rituximab is administered once a week for four weeks. In some embodiments, a complement modulating agent is co-administered with rituximab (e.g., during the administration of rituximab, or within one day of administration of rituximab) for the entire duration of rituximab therapy.
[00228] In some embodiments, daclizumab is administered to an individual one hour before a transplant operation. In some embodiments, daclizumab is administered to an individual every two weeks for 10 weeks after a transplant. In some embodiments, a complement modulating agent is co-administered with daclizumab (e.g., during the administration of daclizumab, or within one day of administration of daclizumab) for the entire duration of daclizumab therapy.
[00229] In some embodiments, omalizumab is administered by subcutaneous injection at a dose of 150 to 375 mg (based on patient's weight and serum IgE level), every 2 or 4 weeks. In some embodiments, a complement modulating agent is co-administered with omalizumab (e.g., during the administration of omalizumab, or within one day of administration of omalizumab) for the entire duration of omalizumab therapy.
[00230] In some embodiments, efalizumab is administered by subcutaneous injection at a dose of 0.7 mg/kg. In some embodiments, efalizumab is administered at a dose 1 mg/kg weekly for 12 weeks. In some embodiments, a complement modulating agent is co-administered with efalizumab (e.g., during the administration of efalizumab, or within one day of administration of efalizumab) for the entire duration of efalizumab therapy.
[00231] In some embodiments, infliximab is administered by infusion at a dose of 3 mg/kg (for RA) to 5mg/kg (e.g., for Crohn's Disease). In some embodiments, a maintenance dose of infliximab is administered 2 and 6 weeks after the first infusion then every 8 weeks thereafter. For individuals that do not respond adequately to the standard dose, infliximab is administered at a dose of up to 10 mg/kg as often as every 4 weeks. In some embodiments, a complement modulating agent is co-administered with infliximab (e.g., during the administration of infliximab, or within one day of administration of infliximab) for the entire duration of infliximab therapy.
XII. Combination Treatments
[00232] Any of the compositions described herein are also optionally used in combination with other therapeutic reagents that are selected for their therapeutic value for the condition to be treated. In general, the compositions described herein and, in embodiments where combinational therapy is employed, other agents do not have to be administered in the same pharmaceutical composition, and, because of different physical and chemical characteristics, are optionally administered by different routes. The initial administration is optionally made according to established protocols, and then, based upon the observed effects, the dosage, modes of administration and times of administration subsequently modified.
[00233] In certain instances, it is appropriate to administer a composition as described herein in combination with another therapeutic agent. By way of example only, if one of the side effects experienced by a patient upon receiving a composition as described herein is nausea, then it is appropriate to administer an anti-emetic agent in combination with the initial therapeutic agent. Or, by way of example only, the therapeutic effectiveness of a composition described herein is enhanced by administration of an adjuvant (i.e., by itself the adjuvant has minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced). Or, by way of example only, the benefit experienced by a patient is increased by administering a composition described herein with another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit. In any case, regardless of the disease, disorder or condition being treated, the overall benefit experienced by the patient is either simply additive of the two therapeutic agents or the patient experiences a synergistic benefit.
[00234] Therapeutically-effective dosages vary when the drugs are used in treatment combinations. Methods for experimentally determining therapeutically-effective dosages of drugs and other agents for use in combination treatment regimens are documented methodologies. One example of such a method is the use of metronomic dosing, i.e., providing more frequent, lower doses in order to minimize toxic side effects. Combination treatment further includes periodic treatments that start and stop at various times to assist with the clinical management of the patient.
[00235] In any case, the multiple therapeutic agents are administered in any order, or even simultaneously. If simultaneously, the multiple therapeutic agents are optionally provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills). The term "simultaneous administration," as used herein, means that the composition comprising a complement-depleting compound and the second therapy (e.g., hormone therapy, gene therapy, antibody therapy, and/or chemotherapy) are administered with a time separation of no more than about 15 minute(s), such as no more than about any of 10, 5, or 1 minutes. In some embodiments, one of the therapeutic agents is given in multiple doses, or both are given as multiple doses.
[00236] If not simultaneous, the timing between the multiple doses optionally varies from more than zero weeks to less than four weeks. In some embodiments, the composition comprising a complement-depleting compound and the second therapy (e.g., hormone therapy, gene therapy, antibody therapy, and/or chemotherapy) are administered sequentially. The term "sequential administration" as used herein means that the composition comprising a complement-depleting compound and the second therapy (e.g., hormone therapy, gene therapy, antibody therapy, and/or chemotherapy) are administered with a time separation of more than about 15 minutes, such as more than about any of 20, 30, 40, 50, or 60 minutes, more than about any of 1 day, 2 days, 3 days, 1 week, 2 weeks, or 1 month, or longer.
[00237] In addition, the combination methods, compositions and formulations are not to be limited to the use of only two agents; the use of multiple therapeutic combinations is also envisioned.
[00238] It is understood that the dosage regimen to treat, prevent, or ameliorate the condition(s) for which relief is sought, is optionally modified in accordance with a variety of factors. These factors include the disorder from which the individual suffers, as well as the age, weight, sex, diet, and medical condition of the individual. Thus, the dosage regimen actually employed varies widely, in some embodiments, and therefore deviates from the dosage regimens set forth herein.
[00239] The agents which make up the combination therapy disclosed herein are optionally a combined dosage form or in separate dosage forms intended for substantially simultaneous administration. The pharmaceutical agents that make up the combination therapy are optionally also be administered sequentially, with either therapeutic agent being administered by a regimen calling for two-step administration. The two-step administration regimen optionally calls for sequential administration of the active agents or spaced-apart administration of the separate active agents. The time period between the multiple administration steps ranges from, a few minutes to several hours, depending upon the properties of each pharmaceutical agent, such as potency, solubility, bioavailability, plasma half- life and kinetic profile of the pharmaceutical agent. Circadian variation of the target molecule concentrations are optionally used to determine the optimal dose interval.
[00240] In some embodiments, the compositions disclosed herein are administered in combination with anti-TNF agent, an IL- 1 receptor antagonist, an IL-2 receptor antagonist, an immunomodulatory agent, an antibiotic, a T-cell co-stimulatory blocker, a disorder-modifying anti-rheumatic agent, a B cell depleting agent, an immunosuppresive agent, an anti-lymphocyte antibody, or combinations thereof.
[00241] In some embodiments, the compositions disclosed herein are administered in combination with an anti-inflammatory agent. In some embodiments, the compositions disclosed herein are administered in combination with a chemotherapeutic agent. In some embodiments, the compositions disclosed herein are administered in combination with radiation therapy. In some embodiments, the compositions disclosed herein are administered in combinations with alefacept, efalizumab, methotrexate, acitretin, isotretinoin, hydroxyurea, mycophenolate mofetil, sulfasalazine, 6-Thioguanine, Dovonex, Taclonex, betamethasone, tazarotene, hydroxychloroquine, sulfasalazine, etanercept, adalimumab, infliximab, abatacept, rituximab, trastuzumab, Anti-CD45 monoclonal antibody AHN- 12 (NCI), Iodine- 131 Anti-Bl Antibody (Corixa Corp.), anti-CD66 monoclonal antibody BW 250/183 (NCI, Southampton General Hospital), anti-CD45 monoclonal antibody (NCI, Baylor College of Medicine), antibody anti- anb3 integrin (NCI), BIW-8962 (Bio Wa Inc.), Antibody BC8 (NCI), antibody muJ591 (NCI), indium In 111 monoclonal antibody MN- 14 (NCI), yttrium Y 90 monoclonal antibody MN- 14 (NCI), F 105 Monoclonal Antibody (NIAID), Monoclonal Antibody RAV 12 (Raven Biotechnologies), CAT- 192 (Human Anti-TGF-Betal Monoclonal Antibody, Genzyme), antibody 3F8 (NCI), 177Lu-J591 (Weill Medical College of Cornell University), TB-403 (Biolnvent International AB), anakinra, azathioprine, cyclophosphamide, cyclosporine A, leflunomide, d-penicillamine, amitriptyline, or nortriptyline, chlorambucil, nitrogen mustard, prasterone, LJP 394 (abetimus sodium), LJP 1082 (La Jolla Pharmaceutical), eculizumab, belibumab, rhuCD40L (NIAID), epratuzumab, sirolimus, tacrolimus, pimecrolimus, thalidomide, antithymocyte globulin- equine (Atgam, Pharmacia Upjohn), antithymocyte globulin- rabbit (Thymoglobulin, Genzyme), Muromonab-CD3 (FDA Office of Orphan Products Development), basiliximab, daclizumab, riluzole, cladribine, natalizumab, interferon beta- Ib, interferon beta- 1 a, tizanidine, baclofen, mesalazine, asacol, pentasa, mesalamine, balsalazide, olsalazine, 6-mercaptopurine, AIN457 (Anti IL- 17 Monoclonal Antibody, Novartis), theophylline, D2E7 (a human anti-TNF mAb from Knoll Pharmaceuticals), Mepolizumab (Anti-IL-5 antibody, SB 240563), Canakinumab (Anti-IL-1 Beta Antibody, NIAMS), Anti-IL-2 Receptor Antibody (Daclizumab, NHLBI), CNTO 328 (Anti IL-6 Monoclonal Antibody, Centocor), ACZ885 (fully human anti-interleukin-lbeta monoclonal antibody, Novartis), CNTO 1275 (Fully Human Anti-IL-12 Monoclonal Antibody, Centocor), (3S)-N-hydroxy-4-({4-[(4-hydroxy-2-butynyl)oxy]phenyl}sulfonyl)-2,2-dimet- hyl-3- thiomorpholine carboxamide (apratastat), golimumab (CNTO 148), Onercept, BG9924 (Biogen Idee), Certolizumab Pegol (CDP870, UCB Pharma), AZD9056 (AstraZeneca), AZD5069 (AstraZeneca), AZD9668 (AstraZeneca), AZD7928 (AstraZeneca), AZD2914 (AstraZeneca), AZD6067 (AstraZeneca), AZD3342 (AstraZeneca), AZD8309 (AstraZeneca), ), [(lR)-3- methyl-l-({(2S)-3-phenyl-2-[(pyrazin-2-ylcarbonyl)amino]propanoyl}amino)butyl]boronic acid (Bortezomib), AMG-714, (Anti-IL 15 Human Monoclonal Antibody, Amgen), ABT-874 (Anti IL- 12 monoclonal antibody, Abbott Labs), MRA(Tocilizumab, an Anti IL-6 Receptor Monoclonal Antibody, Chugai Pharmaceutical), CAT-354 (a human anti-interleukin-13 monoclonal antibody, Cambridge Antibody Technology, Medlmmune), aspirin, salicylic acid, gentisic acid, choline magnesium salicylate, choline salicylate, choline magnesium salicylate, choline salicylate, magnesium salicylate, sodium salicylate, diflunisal, carprofen, fenoprofen, fenoprofen calcium, flurobiprofen, ibuprofen, ketoprofen, nabutone, ketolorac, ketorolac tromethamine, naproxen, oxaprozin, diclofenac, etodolac, indomethacin, sulindac, tolmetin, meclofenamate, meclofenamate sodium, mefenamic acid, piroxicam, meloxicam, celecoxib, rofecoxib, valdecoxib, parecoxib, etoricoxib, lumiracoxib, CS-502 (Sankyo), JTE-522 (Japan Tobacco Inc.), L-745,337 (Almirall), NS398 (Sigma), betamethasone (Celestone), prednisone (Deltasone), alclometasone, aldosterone, amcinonide, beclometasone, betamethasone, budesonide, ciclesonide, clobetasol, clobetasone, clocortolone, cloprednol, cortisone, cortivazol, deflazacort, deoxycorticosterone, desonide, desoximetasone, desoxycortone, dexamethasone, diflorasone, diflucortolone, difluprednate, fluclorolone, fludrocortisone, fludroxycortide, flumetasone, flunisolide, fluocinolone acetonide, fluocinonide, fluocortin, fluocortolone, fluorometholone, fluperolone, fluprednidene, fluticasone, formocortal, formoterol, halcinonide, halometasone, hydrocortisone, hydrocortisone aceponate, hydrocortisone buteprate, hydrocortisone butyrate, loteprednol, medrysone, meprednisone, methylprednisolone, methylprednisolone aceponate, mometasone furoate, paramethasone, prednicarbate, prednisone, rimexolone, tixocortol, triamcinolone, and ulobetasol. In some embodiments, the complement-depleting compounds (such as modified human C3 proteins) described herein are used to increase the efficiency and/or effectiveness of gene therapy to treat cancer by delivering the compound in an amount sufficient to deplete complement before applying gene therapy treatment to the individual. [00243] In some embodiments, the complement-depleting compound is administered to a subject before, during, or after a biopsy or a surgery associated with cancer so that the risk of metastasis is reduced.
[00244] While certain embodiments of the present invention have been shown and described herein, it will be apparent to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that in some embodiments of the invention various alternatives to the embodiments described herein are employed in practicing the invention.
Examples
Example 1 : Rituximab/HC3-1496 Combination for Treatment of Mouse Model of Arthritis Mouse Model
[00245] Twenty- four female SCID mice weighing 20 to 24 g are used. The animals used were within an age range of 6 to 8 weeks. [00246] The mice are injected intradermally with 100 ag of collagen type-II suspended in complete
Freund's adjuvant (CFA) at the base of the tail (t=0). Study Design
[00247] The mice are divided into two groups: the experimental group (n=12) and the control group
(n=12). The experimental group receives rituximab (300 μg) and HC3-1496 (3mg/kg) weekly by intraperitoneal injection starting on day 7. The control group receives rituximab (300 μg) weekly by intraperitoneal injection starting on day 7. [00248] At day 49 mice are sacrificed. Mice are evaluated for arthritis using an established macroscopic scoring system ranging from 0 to 4 (0 = no detectable arthritis, 1 = swelling and/or redness of paw or one digit, 2 = two joints involved, 3 = three or four joints involved and 4 = severe arthritis of the entire paw and digits).
Example 2: Human Clinical Trial of Inflixumab in combination with HC3-1496 for for Treatment of
Rheumatoid Arthritis
Design
[00249] Multicenter, randomized, double-blind, parallel group study Outcome Measures
[00250] The primary outcome measurement is: a. To assess the efficacy of inflixumab alone versus inflixumab in combination with INC- 1496 in treatment of recent onset RA according to ACR50 criteria [00251] The secondary outcome measurements are: a. Remission rate after completion of treatment b. Progression of articular damage c. Quality of life d. Toxicity after completion of treatment Treatment Regime
[00252] Individuals are randomized into two groups: a control group (n=200) and an experimental group (n=200).
[00253] The control group is administered inflixumab at a dose of 3mg/kg by slow infusion on days 0, 14, 28, 42, 56, 70, 84, 98, 112, 126, 140, 154, and 168.
[00254] The experimental group is administered inflixumab at a dose of 3mg/kg by slow infusion on days 0, 14, 28, 42, 56, 70, 84, 98, 112, 126, 140, 154, and 168. The experimental group is coadministered HC3- 1496 (15 mg/m2) on days 0, 14, 28, 42, 56, 70, 84, 98, 112, 126, 140, 154, and 168. Inclusion Criteria
[00255] Male or female
[00256] Between the ages of 18 and 60
[00257] Use of for contraceptive women of childbearing potential
[00258] Diagnosis of RA based on ACR criteria
[00259] Presence of at least four of the following criteria a. Morning stiffness last at least an hour b. Swelling of at least 3 out of the following joints i. Wrist ii. Elbow iii. Knee iv. Ankle v. Metacarpophalangeal joints vi. Metatarsophalangeal joints vii. Interphalngeal joints c. Swelling of at least one joint of the hand d. Presence of rheumatoid nodules e. Positive rheumatoid factor
[00260] Onset of RA symptoms within 12 months prior to day=0
Example 3: Human Clinical Trial of Natalizumab in combination with HC3-1496 for Treatment of Crohn's Disease Design
[00261] Multicenter, randomized, double-blind, parallel group study in subjects with moderately to severely active CD (based on clinical evaluation and CDAI score >220 to <450) and elevated CRP levels (defined as >2.87 mg/L, the upper limit of normal [ULN]) as assessed by the study central laboratory at the screening visit. Objectives and Endpoints
[00262] The primary objective is: a. To compare the ability of natalizumab alone versus natalizumab in combination with HC3- 1496 to induce a clinical response in subjects with moderately to severely active Crohn's disease (CD) and elevated serum C-reactive protein (CRP), as determined by the proportion (%) of subjects with a >70-point decrease from baseline (Week 0) in Crohn's Disease Activity Index (CDAI) score at both Weeks 8 and 12. b. To evaluate the safety and tolerability of natalizumab alone versus natalizumab in combination with HC3-1496 in subjects with moderately to severely active CD and elevated CRP as determined by the occurrence of and/or changes in: i. adverse events; ii. clinical laboratory parameters; iii. vital signs; and iv. physical examination data. [00263] The secondary objective is: a. To compare the ability of natalizumab alone versus natalizumab in combination with HC3- 1496 to induce a clinical remission in subjects with moderately to severely active CD and elevated CRP, as determined by the proportion (%) of subjects with a CDAI score < 150 at both Weeks 8 and 12. b. To compare the ability of natalizumab alone versus natalizumab in combination with HC3- 1496 to induce a clinical response or remission at Week 12, as determined by: i. the proportion (%) of subjects with a clinical response (>70-point decrease from baseline [Week 0] in CDAI score) at Week 12; and ii. the proportion (%) of subjects in clinical remission (CDAI score <150) at Week 12 c. To compare the ability of natalizumab alone versus natalizumab in combination with HC3- 1496 to induce a clinical effect at various time points as determined by: i. the proportion (%) of subjects achieving a >100-point decrease from baseline (Week 0) in CDAI score at both Weeks 8 and 12; ii. the proportion (%) of subjects achieving a clinical response at both Weeks 4 and 8; iii. the proportion (%) of subjects achieving a clinical remission at both Weeks 4 and 8; iv. time to clinical response; v. time to clinical remission; vi. the proportion (%) of subjects achieving a clinical response at Week 8; vii. the proportion (%) of subjects achieving a clinical remission at Week 8; and viii. mean change from baseline (Week 0) in CDAI score at Weeks 4, 8, and 12. d. To compare the effects of natalizumab alone versus natalizumab in combination with HC3- 1496 on changes in inflammatory markers, including CRP and platelet count, as determined by: i. mean change from baseline (Week 0) in platelet count at Weeks 4, 8, and 12; and ii. mean change from baseline (Week 0) in CRP level at Weeks 4, 8, and 12. e. To evaluate the effects of natalizumab alone versus natalizumab in combination with HC3- 1496 on quality of life (QOL), as measured by the Inflammatory Bowel Disease Questionnaire (IBDQ) and Short Form-36 (SF-36) health survey questionnaire, as determined by: i. mean change from baseline (Week 0) in IBDQ score and its dimensions at Week 12; and ii. mean change from baseline (Week 0) in SF-36 score and its components at Week 12. Treatment Regime
[00264] Individuals are randomized into two groups: a control group (n=200) and an experimental group (n=200).
[00265] The control group receives monthly (defined as a 4-week period) intravenous (IV) infusions of natalizumab 300 mg at Weeks 0, 4, and 8 (IV infusion of 300 mg over approximately 60 minutes at a flow rate of 2 mL/min). [00266] The experimental group receives monthly (defined as a 4-week period) intravenous (IV) infusions of natalizumab 300 mg in combination with HC3-1496 (15 mg/m2) at Weeks 0, 4, and 8.
[00267] Subjects return to the clinic for safety and efficacy assessments at Weeks 4, 8, 12, and 20. Inclusion Criteria
[00268] Male or female subjects
[00269] >18 years of age
[00270] At least a 6-month clinical history of Crohn's Disease a. Clinical evidence of active (symptomatic) CD based on clinical history and radiological or endoscopic findings within the previous 36 months, a CDAI score >220 to <450 at Week 0, and a CRP level above the ULN (>2.87 mg/L) performed by the central laboratory at the screening visit.
[00271] Use of for contraceptive women of childbearing potential Exclusion Criteria
[00272] Subjects with colostomy, ileostomy, or colectomy with ileorectal anastomosis, clinical short bowel syndrome, irritable bowel syndrome, draining fistulae, any history of neoplastic disease or other clinically significant illness, or subjects deemed likely to require emergency surgery were not allowed in the study.
While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims

CLAIMS WHAT IS CLAIMED IS:
1. A method of treating an inflammatory disorder, comprising administering to an individual in need thereof a synergistically-effective amount of (a) an antibody or fusion protein comprising an Fc domain; and (b) a second agent selected from: a complement depleting agent, an agent that inhibits the formation and/or activity of C3b, or a combination thereof; and wherein the production of C5a is not stimulated, or is only minimally stimulated.
2. The method of claim 1, wherein the antibody is a monoclonal antibody.
3. The method of claim 1, wherein the antibody is an anti-CD3 antibody, an anti-CD 1 la§ antibody, an anti-CD-20 antibody, an anti-CD22 antibody, an anti-CD-25 antibody, an anti-CD52 antibody, anti- TNF antibody, anti- IgE antibody, an anti- α4-integrin antibody, or combinations thereof.
4. The method of claim 1 , wherein the second agent is GR-2II, AGIIa, AGIIb- 1 , AR-2IIa, AR- 21Ib, AR-2IIc, AR-2IH, CVF, hybrid human C3-CVF protein, rC3, HC3-1496, HC3-1496-2, HC3-1496-3, HC3-1496-4, HC3-1496/1617, HC3-1496-8, HC3-1496-9, HC3-1496-10, HC3-1496-11, HC3-1496-12, HC3- 1496-13, HC3-1496-14, HC3-1496-15, HC3-1496-16, HC3-1496-17, complement receptor 1, sCRl, APT070, TPlO, TP20, sCRl [desLHR-A]), sCRl-SLex, Crry, Crry-Ig, fucan, BS8, complestatin, Ecb, Efb, compstatin, rosmarinic acid, CRIT, CRIT-H17, glycyrrhetinic acid, CD55, sCD55, a CD55/MCP fusion protein, BCX- 1470, FUT- 175, Factor I, MCP, sMCP, heparin, an anti-properdin antibody, or combinations thereof.
5. The method of claim 1; wherein the inflammatory disorder is Atopic dermatitis; Acute disseminated encephalomyelitis; Acute leukocyte-mediated lung injury; Addison's disease; AIDS dementia; Allergic rhinitis; Ankylosing spondylitis; Antiphospholipid antibody syndrome; Alzheimer's disorder; Asthma; Adult respiratory distress syndrome; Autoimmune hemolytic anemia; Autoimmune hepatitis; Autoimmune inner ear disease; Behcet's syndrome; Bronchitis; Bullous pemphigoid; Chagas disease; Chronic obstructive pulmonary disease; Coagulative Necrosis; Coeliac disease; Crohn's disorder; Collagenous colitis; Conjunctivitis; Cystic fibrosis; Dermatomyositis; Dermatitis; Diabetes mellitus type 1; Diabetes mellitus type 2; Distal proctitis; Diversion colitis; Encephalitis; Endometriosis; Endotoxin shock; Eczema; Epilepsy; Fibromyalgia; Fibrinoid Necrosis; Goodpasture's syndrome; Gouty arthritis; Graves' disease; Guillain-Barre syndrome; Hashimoto's disease; Idiopathic thrombocytopenic purpura; Indeterminate colitis; Infective colitis; Inflammatory liver disorder; Interstitial cystitis; Ischaemic colitis; Liquefactive Necrosis; Lymphocytic colitis; Meningitis; Multiple sclerosis; Myasthenia gravis; Myocarditis; Narcolepsy; Nephritis; Pancreatitis; Parkinson's disorder; Pemphigus Vulgaris; Periodontal gingivitis; Pernicious anaemia; Polymyositis; Polymyalgia rheumatica; Psoriasis; Primary biliary cirrhosis; Retinitis; Rheumatoid arthritis; Rheumatoid spondylitis; Schizophrenia; Scleroderma; Shingles; Sjogren's syndrome; Smooth muscle proliferation disorders; Systemic lupus erythematosus (SLE); Transplant complication; Hyperacute transplant rejection; Acute transplant rejection; Chronic transplant rejection,; Acute graft-versus-host disease; Chronic graft- versus-host disease; Tuberculosis; Ulcerative colitis; Uveitis; Vasculitis; Vitiligo; Wegener's granulomatosis; or combinations thereof.
6. A method of enhancing an immune response against a pathogen, comprising administering to an individual in need thereof a synergistically-effective amount of (a) an antibody or fusion protein comprising an Fc domain; and (b) a second agent selected from: a complement depleting agent, an agent that inhibits the formation and/or activity of C3b, or a combination thereof; and wherein the production of C5a is not stimulated, or is only minimally stimulated.
7. The method of claim 6, wherein the pathogen is a bacterium, a virus, a fungus, a prion, a protozoan, or a combination thereof.
8. The method of claim 6, wherein the pathogen is RSV, Vibrio cholerae, Plasmodium falciparum, Plasmodium vivax, Plasmodium ovale, Plasmodium malariae, Mycobacterium tuberculosis, a influenza, HIV, E. coli O157:H7, a hepatitis virus, Poliovirus, Mycobacterium leprae, Yersinia pestis, Rickettsia prowazekii, Borrelia burgdorferi, Borrelia afzelii, Borrelia garinii, or combinations thereof.
9. The method of claim 6, wherein the IgG is IgGl, IgG2, IgG3, IgG4, or combinations thereof.
10. The method of claim 6, wherein the antibody is an anti- RSV gpF antibody.
11. The method of claim 6, wherein the antibody is an anti- Pfs25 (Plasmodium falciparum) antibody, an anti- Pfs28 (Plasmodium falciparum), or combinations thereof.
12. The method of claim 6, wherein the antibody is an anti- phenolic glycolipid TB (PGL-Tb) antibody.
13. The method of claim 6, wherein the antibody is an anti- influenza matrix 2 protein (M2e) antibody.
14. The method of claim 6, wherein the antibody is an anti- Y. pestis capsular Fl (Cafl) antibody.
15. The method of claim 6, wherein the second agent is: GR-2II, AGIIa, AGIIb- 1 , AR-2IIa, AR- 21Ib, AR-2IIc, AR-2IId, CVF, a hybrid human C3-CVF protein, rC3, HC3-1496, HC3-1496-2, HC3-1496-3, HC3-1496-4, HC3-1496/1617, HC3-1496-8, HC3-1496-9, HC3-1496-10, HC3-1496-11, HC3-1496-12, HC3- 1496-13, HC3-1496-14, HC3-1496-15, HC3-1496-16, HC3-1496-17, complement receptor 1, sCRl, APT070, TPlO, TP20, sCRl [desLHR-A]), sCRl-SLex, Crry, Crry-Ig, fucan, BS8, complestatin, Ecb, Efb, compstatin, rosmarinic acid, CRIT, CRIT-H 17, glycyrrhetinic acid, CD55, sCD55, a CD55/MCP fusion protein, BCX- 1470, FUT- 175, Factor I, MCP, sMCP, heparin, an anti-properdin antibody, or combinations thereof.
16. A modified human complement C3 protein comprising a human C3 protein, wherein one or more amino acid residues in the human C3 protein are substituted with a corresponding portion of a CVF protein; wherein the protein does not comprise C3a, and wherein the protein does not activate C5.
17. The modified human complement C3 protein of claim 16, wherein the amino acid residues in the human C3 protein corresponding to amino acid residues 1496 to 1663 of SEQ ID NO: 1 are substituted with a corresponding portion of a CVF protein.
18. The modified human complement C3 protein of claim 16, wherein the protein is at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, or at least about 95% homologous with a human C3 sequence.
19. The modified human complement C3 protein of claim 16, wherein one or more amino acid residues in the CVF portion is modified.
20. The modified human complement C3 protein of claim 16, wherein one or more amino acid residues in the CVF portion is modified, and wherein the modification is selected from the group consisting of: (a) T1499D and L1501K; (b) I1507R, G1508D, N1509E, and V1510L; (c) S1519F, S1520I, L1521Q, N1522K, H1523S, and Q1524D; (d) D1528T, V1529L, P1530E, L1531E, and Q1532R; (e) 1519-1550 replaced with corresponding amino acid residues of SEQ ID NO: 1; (f) Q 157 IS, T 1573 S, N 1576V, P1577Q, and R1578V; (g) 1596-1617 replaced with corresponding amino acid residues of SEQ ID NO: 1; (h) 1596- 1611 replaced with corresponding amino acid residues of SEQ ID NO:1; (i) V1598L, N1599D, D1600N, S1607L, and R1608S; (j) V1598L, N1599D, and D1600N; (k) S1607L, and R1608S, or combinations thereof.
21. The modified human complement C3 protein of claim 16, wherein an amino acid of human C3 is substituted or deleted in a domain selected from: MGl, MG2, MG6, alpha-NT', TED, MG7, or combinations thereof.
22. The modified human complement C3 protein of claim 16, wherein Glutamine (Q) 163 of human C3 is substituted.
23. The modified human complement C3 protein of claim 16, wherein Glutamine (Q) 163 of human C3 is deleted.
24. The modified human complement C3 protein of claim 16, wherein Glutamine (Q) 163 of human C3 is deleted, the amino acid residue immediately preceding Glutamine 163 is deleted, and the three amino acid residues immediately following Glutamine 163 are deleted.
25. The modified human complement C3 protein of claim 16, wherein Serine (S) 1075 of human C3 is substituted.
26. The modified human complement C3 protein of claim 16, wherein Serine (S) 1075 of human C3 is substituted with Histidine (H).
27. The modified human complement C3 protein of claim 16, wherein Serine (S) 1075 of human C3 is substituted with Histidine (H) and the two preceding amino acid residues are deleted.
28. The modified human complement C3 protein of claim 16, wherein Asparagine (N) 738 of human C3 is substituted.
29. The modified human complement C3 protein of claim 16, wherein Asparagine (N) 738 of human C3 is substituted with Aspartic Acid.
30. The modified human complement C3 protein of claim 16, wherein Asparagine (N) 738 of human C3 is substituted with Aspartic Acid and the two preceding Glutamic acid residues (EE) are substituted with aspartic acid and serine (DS).
31. The modified human complement C3 protein of claim 16, wherein Asparagine (N) 738 of human C3 is substituted with Aspartic acid and the immediately preceding Glutamic acid (E) is substituted with Serine (S).
32. The modified human complement C3 protein of claim 16, wherein the Glutamine (Q) at position 1139 of human C3 is substituted with a Lysine (K) or a Tyrosine (Y).
PCT/US2009/046331 2008-11-17 2009-06-04 Method and composition for modulating the immune system and various inflammatory conditions comprising complement depletors WO2010056399A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11548108P 2008-11-17 2008-11-17
US61/115,481 2008-11-17

Publications (1)

Publication Number Publication Date
WO2010056399A1 true WO2010056399A1 (en) 2010-05-20

Family

ID=42170235

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/046331 WO2010056399A1 (en) 2008-11-17 2009-06-04 Method and composition for modulating the immune system and various inflammatory conditions comprising complement depletors

Country Status (1)

Country Link
WO (1) WO2010056399A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012125680A1 (en) 2011-03-16 2012-09-20 Novartis Ag Methods of treating vasculitis using an il-17 binding molecule
US8548237B2 (en) 2010-10-18 2013-10-01 Hewlett-Packard Development Company, L.P. Ordinal and spatial local feature vector based image representation
US20130344082A1 (en) * 2012-06-20 2013-12-26 The Trustees Of The University Of Pennsylvania Methods of treating or preventing periodontitis and diseases associated with periodontitis
WO2014031676A1 (en) * 2012-08-20 2014-02-27 O-Ray Pharma, Inc. Process for manufacturing drug delivery formulations
WO2018140121A1 (en) * 2017-01-30 2018-08-02 Janssen Biotech, Inc. Anti-tnf antibodies, compositions, and methods for the treatment of active psoriatic arthritis
WO2019217657A1 (en) * 2018-05-10 2019-11-14 TympoBio, Inc. Compositions and methods for treating autoimmune inner ear disease
US10772886B2 (en) 2016-03-11 2020-09-15 Ardea Biosciences, Inc. CXCR-2 inhibitors for treating crystal arthropathy disorders
WO2020234432A1 (en) * 2019-05-21 2020-11-26 Argenx Bvba Antagonists of the complement system for use in methods of treating paraproteinemic neuropathies
US11524050B2 (en) 2018-01-15 2022-12-13 Complement Therapeutics Limited C3B binding polypeptide
US11591386B2 (en) 2018-12-13 2023-02-28 argenx BV Antibodies to human complement C2B
US11780911B2 (en) 2019-05-23 2023-10-10 Janssen Biotech, Inc. Method of treating inflammatory bowel disease with a combination therapy of antibodies to IL-23 and TNF alpha

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5849297A (en) * 1994-09-08 1998-12-15 Imutran Limited Modified human C3 proteins
US20050079585A1 (en) * 2003-07-03 2005-04-14 Johanna Kolln Complement depletion using recombinant human C3-derivatives
WO2005107785A2 (en) * 2004-04-30 2005-11-17 University Of Hawaii Human complement c3 derivates with cobra venom factor-like function
WO2008060634A2 (en) * 2006-11-15 2008-05-22 University Of Hawaii Human complement c3 derivatives with cobra venom factor-like function

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5849297A (en) * 1994-09-08 1998-12-15 Imutran Limited Modified human C3 proteins
US20050079585A1 (en) * 2003-07-03 2005-04-14 Johanna Kolln Complement depletion using recombinant human C3-derivatives
WO2005107785A2 (en) * 2004-04-30 2005-11-17 University Of Hawaii Human complement c3 derivates with cobra venom factor-like function
WO2008060634A2 (en) * 2006-11-15 2008-05-22 University Of Hawaii Human complement c3 derivatives with cobra venom factor-like function

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KOLLN, J. ET AL.: "Complement inactivation by recombinant human C3 derivatives", J. IMMUNOL., vol. 173, no. 9, 1 November 2004 (2004-11-01), pages 5540 - 5545 *
KOLLN, J. ET AL.: "Engineering of human complement component C3 for catalytic inhibition of complement", IMMUNOL. LETT., vol. 98, no. 1, 15 April 2005 (2005-04-15), pages 49 - 56 *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8548237B2 (en) 2010-10-18 2013-10-01 Hewlett-Packard Development Company, L.P. Ordinal and spatial local feature vector based image representation
WO2012125680A1 (en) 2011-03-16 2012-09-20 Novartis Ag Methods of treating vasculitis using an il-17 binding molecule
US9579360B2 (en) * 2012-06-20 2017-02-28 The Trustees Of The University Of Pennsylvania Methods of treating or preventing periodontitis and diseases associated with periodontitis
US20130344082A1 (en) * 2012-06-20 2013-12-26 The Trustees Of The University Of Pennsylvania Methods of treating or preventing periodontitis and diseases associated with periodontitis
US10668135B2 (en) 2012-06-20 2020-06-02 The Trustees Of The University Of Pennsylvania Methods of treating or preventing periodontitis and diseases associated with periodontitis
CN104582782A (en) * 2012-08-20 2015-04-29 奥雷制药有限公司 Process for manufacturing drug delivery formulations
WO2014031676A1 (en) * 2012-08-20 2014-02-27 O-Ray Pharma, Inc. Process for manufacturing drug delivery formulations
US10772886B2 (en) 2016-03-11 2020-09-15 Ardea Biosciences, Inc. CXCR-2 inhibitors for treating crystal arthropathy disorders
WO2018140121A1 (en) * 2017-01-30 2018-08-02 Janssen Biotech, Inc. Anti-tnf antibodies, compositions, and methods for the treatment of active psoriatic arthritis
US11041020B2 (en) 2017-01-30 2021-06-22 Janssen Biotech, Inc. Methods for the treatment of active Psoriatic Arthritis
US11524050B2 (en) 2018-01-15 2022-12-13 Complement Therapeutics Limited C3B binding polypeptide
WO2019217657A1 (en) * 2018-05-10 2019-11-14 TympoBio, Inc. Compositions and methods for treating autoimmune inner ear disease
US11591386B2 (en) 2018-12-13 2023-02-28 argenx BV Antibodies to human complement C2B
US11708403B2 (en) 2018-12-13 2023-07-25 argenx BV Antibodies to human complement factor C2B and methods of use
WO2020234432A1 (en) * 2019-05-21 2020-11-26 Argenx Bvba Antagonists of the complement system for use in methods of treating paraproteinemic neuropathies
US11780911B2 (en) 2019-05-23 2023-10-10 Janssen Biotech, Inc. Method of treating inflammatory bowel disease with a combination therapy of antibodies to IL-23 and TNF alpha

Similar Documents

Publication Publication Date Title
WO2010056399A1 (en) Method and composition for modulating the immune system and various inflammatory conditions comprising complement depletors
JP7143452B2 (en) ANTIBODY THAT CAN BLOCK THE INTERACTION BETWEEN CD47 AND SIRPa, AND ITS APPLICATION
AU2010208125B2 (en) Human anti-IL-6 antibodies with extended in vivo half-life and their use in treatment of oncology, autoimmune diseases and inflammatory diseases
EP3167888B1 (en) Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
SG186397A1 (en) Antibodies to the c3d fragment of complement component 3
EP1871807A2 (en) Antibodies against cxcr4 and methods of use thereof
JP2020511950A (en) Anti-LILRB3 antibody and methods of use thereof
CN114173819A (en) Methods of treating allergy and enhancing allergen-specific immunotherapy by administering IL-4R antagonists
WO2009149306A2 (en) Complement depleting compounds and methods of treating cancer comprising monoclonal antibody therapy and said complement depleting compounds
JP2021500865A (en) Staphylococcus bispecific antigen-binding molecule that binds to target antigens and complement components and their use
WO2014160121A1 (en) Humanized antibodies specific for staphylococcal enterotoxin b
US20220153819A1 (en) Compositions and methods for using bispecific antibodies to bind complement and a target antigen
JP2023534779A (en) anti-ceramide antibody
WO2022044010A1 (en) Anti-t-cell immunoglobulin and itim domain (tigit) antibodies for the treatment of fungal infections
AU2015202502A1 (en) Human anti-il-6 antibodies with extended in vivo half-life and their use in treatment of oncology, autoimmune diseases and inflammatory diseases
WO2022034044A1 (en) Sars-cov-2 antibodies for treatment and prevention of covid-19
EA040553B1 (en) ANTIBODY TO CD154 WITH IMPROVED BINDING, FUNCTIONALITY AND SAFETY AND ITS USE IN HUMAN IMMUNOTHERAPY
AU2013206214A1 (en) Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09826477

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09826477

Country of ref document: EP

Kind code of ref document: A1