US20100247546A1 - Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria - Google Patents

Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria Download PDF

Info

Publication number
US20100247546A1
US20100247546A1 US12/572,410 US57241009A US2010247546A1 US 20100247546 A1 US20100247546 A1 US 20100247546A1 US 57241009 A US57241009 A US 57241009A US 2010247546 A1 US2010247546 A1 US 2010247546A1
Authority
US
United States
Prior art keywords
antibody
lta
antibodies
human
positive bacteria
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/572,410
Inventor
Gerald Walter Fischer
Richard F. Schuman
Hing Wong
Jeffrey R. Stinson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Henry M Jackson Foundation for Advancedment of Military Medicine Inc
ImmunityBio Inc
Original Assignee
Altor Bioscience Corp
Henry M Jackson Foundation for Advancedment of Military Medicine 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
Priority claimed from US09/097,055 external-priority patent/US6610293B1/en
Application filed by Altor Bioscience Corp, Henry M Jackson Foundation for Advancedment of Military Medicine Inc filed Critical Altor Bioscience Corp
Priority to US12/572,410 priority Critical patent/US20100247546A1/en
Assigned to ALTOR BIOSCIENCE CORPORATION reassignment ALTOR BIOSCIENCE CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: STINSON, JEFFREY R., WONG, HING
Assigned to THE HENRY M. JACKSON FOUNDATION FOR THE ADVANCEMENT OF MILITARY MEDICINE, INC. reassignment THE HENRY M. JACKSON FOUNDATION FOR THE ADVANCEMENT OF MILITARY MEDICINE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SCHUMAN, RICHARD, FISCHER, GERALD
Publication of US20100247546A1 publication Critical patent/US20100247546A1/en
Priority to US15/081,297 priority patent/USRE48475E1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • C07K16/1271Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria from Micrococcaceae (F), e.g. Staphylococcus
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • This invention in the fields of immunology and infectious diseases relates to antibodies that are specific for Gram positive bacteria, particularly to bacteria that bear lipoteichoic acids on their surfaces.
  • the invention includes monoclonal and chimeric antibodies, as well as fragments, regions and derivatives thereof.
  • This invention further relates to sequences of the variable region that enhance the antibody's opsonic activity.
  • the antibodies of the invention may be used for diagnostic, prophylactic and therapeutic applications.
  • Staphylococci are particularly worrisome because they commonly colonize humans and animals and are an important cause of human morbidity and mortality. Because of their prevalence on the skin and mucosal linings, staphylococci are ideally situated to produce both localized and systemic infections. Of the staphylococci, both S. aureus , a coagulase positive bacteria, and S. epidermidis , a coagulase negative species, are the most problematic. In fact, S. aureus is the most virulent Staphylococcus , producing severe and often fatal disease in both normal and immunocompromised hosts. S.
  • S. epidermidis has become one of the major causes of nosocomial (hospital acquired) infection in patients with impaired immune responses or those whose treatments involve the placement of foreign objects into the body, such as patients who receive continuous ambulatory peritoneal dialysis and patients receiving parenteral nutrition through central venous catheters (25).
  • S. epidermidis is now recognized as a common cause of neonatal nosocomial sepsis, and infections frequently occur in premature infants that have received parenteral nutrition.
  • the involvement of S. epidermidis in neonatal infection has increased dramatically. Indeed, for every 10 babies diagnosed with bacterial sepsis seven or more days after birth (indicative of post-partum bacterial exposure), six of those are infected with S.
  • Staphylococcus infections in newborns can result in multiple organ failure and death in two to three days.
  • Antibiotics are only partially effective and, unfortunately, the rise in multiply drug resistant strains of Staphylococcus renders antibiotic treatments less and less effective.
  • the other possible route of treatment is the administration of antibodies.
  • Antibodies protect against bacterial attack by recognizing and binding to antigens on the bacteria to thereby facilitate the removal or “clearance” of the bacteria by a process called phagocytosis, wherein phagocytic cells (predominantly neutrophils and macrophages) identify, engulf, and subsequently destroy the invading bacteria.
  • bacteria have developed mechanisms to avoid phagocytosis, such as the production of a “capsule” to which phagocytes cannot adhere or the production of toxins that actually poison the encroaching phagocytes.
  • Antibodies overcome these defenses by, for example, binding to the toxins to thereby neutralize them. More significantly, antibodies may themselves bind to the capsule to coat it, in a process called opsonization, to make the bacteria extremely attractive to phagocytes and to enhance their rate of clearance from the bloodstream.
  • the present invention encompasses broadly reactive, opsonic, and protective monoclonal and chimeric antibodies that bind to lipoteichoic acid (LTA) of Gram positive bacteria.
  • LTA lipoteichoic acid
  • the antibodies also bind to whole bacteria and enhance phagocytosis and killing of the bacteria in vitro and enhance protection from lethal infection in vivo.
  • the present invention further encompasses opsonic antibodies to LTA that share a high degree of sequence homology.
  • the present invention also encompasses antibodies having variable regions derived from two or more different anti-LTA antibodies.
  • FIG. 1 provides a schematic representation of lipoteichoic acid (LTA) in the Gram positive bacterial cell wall.
  • LTA lipoteichoic acid
  • FIG. 2 depicts antibody regions, including the heavy chain constant region (C H ), the heavy chain variable region (V H ), the light chain constant region (C L ), and the light chain variable region (V L ).
  • the complementarity determining regions (CDRs) within the variable regions are shown as black bars.
  • FIG. 3 shows the cDNA cloning strategy for the heavy and light chain variable regions of A120.
  • FIG. 4 shows the oligonucleotide primers used to amplify the variable region fragments. (SEQ ID NOs: 1-9 and 18)
  • FIG. 5 shows the amino acid sequence (SEQ ID NO: 10) and the polynucleotide sequence (SEQ ID NO: 11) of the A120 light chain variable region.
  • FIG. 6 shows the amino acid sequence (SEQ ID NO: 12) and the polynucleotide sequence (SEQ ID NO: 13) of the A120 heavy chain variable region.
  • FIG. 7 depicts the pJSB23-1 plasmid that expresses the A120 heavy chain.
  • FIG. 8 depicts the pJSB24 plasmid that expresses the A120 light chain.
  • FIG. 9 shows an alignment of (A) the A110 light chain variable region cDNA (SEQ ID NO: 14), the A120 light chain variable region cDNA (SEQ ID NO: 11) and the 391.4 light chain variable region cDNA (SEQ ID NO: 19) and (B) the A110 heavy chain variable region cDNA (SEQ ID NO: 15), the A120 heavy chain variable region cDNA (SEQ ID NO: 13) and the 391.4 heavy chain variable region cDNA (SEQ ID NO: 20).
  • the nucleotides that differ between any two sequences are boxed.
  • FIG. 10A shows an alignment of the A110 light chain variable region polypeptide sequence (SEQ ID NO: 16), the A120 light chain variable region polypeptide sequence (SEQ ID NO: 10) and the 391.4 light chain variable region polypeptide sequence (SEQ ID NO: 21).
  • FIG. 10B shows an alignment of the A110 heavy chain variable region polypeptide sequence (SEQ ID NO: 17), the A120 heavy chain variable region polypeptide sequence (SEQ ID NO: 12) and the 391.4 heavy chain variable region polypeptide sequence (SEQ ID NO: 22).
  • the complementarity determining regions (CDRs) are underlined and the amino acids that differ between any two sequences are boxed.
  • FIG. 11 depicts the pJRS354 bi-cistronic plasmid that expresses the A110 heavy chain and light chain variable regions.
  • FIG. 12 depicts the pJSB25-3 bi-cistronic plasmid that expresses the A110 heavy chain variable region and the A120 light chain variable region.
  • FIG. 13 depicts the pJSB26 bi-cistronic plasmid that expresses the A120 heavy chain and light chain variable regions.
  • FIG. 14 depicts the pJSB27 bi-cistronic plasmid that expresses the A120 heavy chain variable region and the A110 light chain variable region.
  • FIG. 15 provides the results of the chimeric antibody production ELISA. All antibodies shown are human/mouse chimeras. A110 contains both the heavy and light chain variable regions from A110. A120 contains both the heavy and light chain variable regions from A120. A120a contains the heavy chain variable region from A110 and the light chain variable region from A120. A120b contains the heavy chain variable region from A120 and the light chain variable region from A110.
  • FIG. 16 provides the results of the experiment to determine chimeric antibody binding to the S. aureus LTA.
  • the antibodies used are the same as in FIG. 15 .
  • antibody includes full-length antibodies and portions thereof.
  • a full-length antibody has one pair or, more commonly, two pairs of polypeptide chains, each pair comprising a light and a heavy chain. Each heavy or light chain is divided into two regions, the variable region (which confers antigen recognition and binding) and the constant region (associated with localization and cellular interactions).
  • a full-length antibody commonly contains two heavy chain constant regions (H c , or C H ), two heavy chain variable regions (H v or V H ), two light chain constant regions (Lc or Cl_), and two light chain variable regions (L v or V L ) ( FIG. 2 ).
  • the light chains or chain may be either a lambda or a kappa chain.
  • the antibodies include at least one heavy chain variable region and one light chain variable region, such that the antibody binds antigen.
  • variable region that comprises alternating complementarity determining regions, or CDRs, and framework regions, or FRs.
  • CDRs are the sequences within the variable region that generally confer antigen specificity.
  • the invention also encompasses portions of antibodies that comprise sufficient variable region sequence to confer antigen binding.
  • Portions of antibodies include, but are not limited to Fab, Fab′, F(ab′) 2 , Fv, SFv, scFv (single-chain Fv), whether produced by proteolytic cleavage of intact antibodies, such as papain or pepsin cleavage, or by recombinant methods, in which the cDNAs for the intact heavy and light chains are manipulated to produce fragments of the heavy and light chains, either separately, or as part of the same polypeptide.
  • MAbs within the scope of the invention include sequences corresponding to human antibodies, animal antibodies, and combinations thereof.
  • One type of chimeric antibodies, “humanized antibodies”, have had the variable regions altered (through mutagenesis or CDR grafting) to match (as much as possible) the known sequence of human variable regions.
  • CDR grafting involves grafting the CDRs from an antibody with desired specificity onto the FRs of a human antibody, thereby replacing much of the non-human sequence with human sequence.
  • Humanized antibodies therefore, more closely match (in amino acid sequence) the sequence of known human antibodies.
  • HAMA human anti-mouse antibody
  • the invention further includes fully human antibodies which would avoid, as much a possible, the HAMA response.
  • Modified antibodies include, for example, the proteins or peptides encoded by truncated or modified antibody-encoding genes. Such proteins or peptides may function similarly to the antibodies of the invention.
  • Other modifications such as the addition of other sequences that may enhance the effector function, which includes the ability to block or alleviate nasal colonization by staphylococci, are also within the present invention.
  • Such modifications include, for example, the addition of amino acids to the antibody's amino acid sequence, deletion of amino acids in the antibody's amino acid sequence, substitution of one or more amino acids in the antibody amino acid sequence with alternate amino acids, isotype switching, and class switching.
  • an antibody may be modified in its Fc region to prevent binding to bacterial proteins.
  • the Fc region normally provides binding sites for neutrophils, macrophages, other accessory cells, complement components, and, receptors of the immune system.
  • accessory cells recognize the coated bacteria and respond to infection.
  • a bacterial protein binds to the Fc region near the places where accessory cells bind, the normal function of these cells is inhibited.
  • Protein A a bacterial protein found in the cell membrane of S. aureus , binds to the Fc region of IgG near accessory cell binding sites. In doing so, Protein A inhibits the function of these accessory cells, thus interfering with clearance of the bacterium.
  • the Fc portion of the antibody of the invention may be modified to prevent nonspecific binding of Protein A while retaining binding to accessory cells (15).
  • the antibodies of the invention include clones of full length antibodies, antibody portions, chimeric antibodies, humanized antibodies, fully human antibodies, and modified antibodies. Collectively, these will be referred to as “MAbs” or monoclonal antibodies unless otherwise indicated.
  • epitope refers to a region, or regions, of LTA that is bound by an antibody to LTA.
  • the regions that are bound may or may not represent a contiguous portion of the molecule.
  • an antigen refers to a polypeptide sequence, a non-proteinaceous molecule, or any molecule that can be recognized by the immune system.
  • An antigen may be a full-sized staphylococcal protein or molecule, or a fragment thereof, wherein the fragment is either produced from a recombinant cDNA encoding less than the full-length protein or derived from the full-sized molecule or protein. Such fragments may be produced via enzymatic processing, such as proteolysis.
  • An antigen may also be a polypeptide sequence that encompasses an epitope of a staphylococcal protein, wherein the epitope may not be contiguous with the linear polypeptide sequence of the protein.
  • the DNA sequence encoding an antigen may be identified, isolated, cloned, and transferred to a prokaryotic or eukaryotic cell for expression by procedures well-known in the art (25).
  • An antigen, or epitope thereof may be 100% identical to a region of the staphylococcal molecule or protein amino acid sequence, or it may be at least 95% identical, or at least 90% identical, or at least 85% identical.
  • An antigen may also have less than 95%, 90% or 85% identity with the staphylococcal molecule or protein amino acid sequence, provided that it still be able to elicit antibodies the bind to a native staphylococcal molecule or protein.
  • the percent identity of a peptide antigen can be determined, for example, by comparing the sequence of the target antigen or epitope to the analogous portion of staphylococcal sequence using the GAP computer program, version 6.0 described by Devereux et al. (Nucl. Acids Res.
  • the GAP program utilizes the alignment method of Needleman and Wunsch (J. Mol. Biol. 48:443, 1970), as revised by Smith and Waterman (Adv. Appl. Math 2:482, 1981), and is applicable to determining the percent identity of protein or nucleotide sequences referenced herein.
  • the preferred default parameters for the GAP program include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non-identities) for nucleotides, and the weighted comparison matrix of Gribskov and Burgess, Nucl. Acids Res.
  • the percent identity over a defined region of peptide or nucleotide sequence may by determined by dividing the number of matching amino acids or nucleotides by the total length of the aligned sequences, multiplied by 100%. Where an insertion or gap of one, two, or three amino acids occurs in a MAb chain, for example in or abutting a CDR, the insertion or gap is counted as single amino acid mismatch.
  • Antigens may be surface antigens and/or virulence antigens and/or adherance antigens.
  • Surface antigens are antigens that are accessible to an antibody when the antigen is in the configuration of the whole intact bacterium, i.e., the antigen is not inside the cell cytoplasm.
  • Virulence antigens are antigens that are involved in the pathogenic process, causing disease in a host. Virulence antigens include, for example, LTA, peptidoglycan, toxins, fimbria, flagella, and adherence antigens.
  • Adherence antigens mediate the ability of a staphylococcal bacterium to adhere to an epithelial surface, such as the epithelial surface of the anterior nares.
  • An antigen may be a non-proteinaceous component of staphylococci such as a carbohydrate or lipid.
  • peptidoglycan and lipoteichoic acid are two non-proteinaceous antigens found in the cell wall of staphylococci.
  • Antigens may comprise or include fragments of non-proteinaceous molecules as long as they elicit an immune response.
  • antigens include molecules that can elicit an antibody response to LTA.
  • An antigen may be LTA itself, or a fragment or portion thereof.
  • An antigen may also be an unrelated molecule, which, through some structural similarity, is able to elicit antibodies that bind to LTA. Binding to LTA may thus be assessed by binding to such peptide epitope mimics, as described, for example, in U.S. Patent Application Publication No. 2002/0082395, which is incorporated herein by reference.
  • an antigen elicits antibodies that bind to LTA on the surface of bacteria.
  • an antigen is any molecule that can specifically bind to an antibody, including antibodies specific for LTA.
  • Antigens of the invention thus include antigens that bind to any of monoclonal antibodies MAb-391.4, M110, M120, A110, A120, A120a, and A120b, described herein.
  • An antibody is said to specifically bind to an antigen, epitope, or protein, if the antibody gives a signal by an assay such as an ELISA assay that is at least two fold, at least three fold, at least five fold, or at least ten fold greater than the background signal, i.e., at least two fold, at least three fold, at least five fold, or at least ten fold greater than the signal ascribed to non-specific binding.
  • An antibody is said to specifically bind to a bacterium if the antibody gives a signal by MeOH-fixed bacteria ELISA or live bacteria ELISA, or other assay, that is at least 1.5 fold, 2 fold, or 3 fold greater than the background signal.
  • Enhanced phagocytosis means an increase in phagocytosis over a background level as assayed by the methods in this application, or another comparable assay. The level deemed valuable may well vary depending on the specific circumstances of the infection, including the type of bacteria and the severity of the infection. For example, for enhanced phagocytic activity, in one embodiment, an enhanced response is equal to or greater than 75% over background phagocytosis. In another embodiment, an enhanced response is equal to or greater than 80% or 85% over background phagocytosis. In another embodiment, an enhanced response is equal to or greater than 90% or 95% over background phagocytosis.
  • Enhanced phagocytosis may also be equal to or greater than 50%, 55%, 60%, 65%, or 70% over background phagocytosis.
  • enhanced phagocytosis comprises a statistically significant increase in phagocytic activity as compared to background phagocytosis or phagocytosis with a non-specific or non-opsonic control antibody.
  • a MAb has “opsonic activity” if it can bind to an antigen to promote attachment of the antigen to the phagocyte and thereby enhance phagocytosis. As used herein, opsonic activity may also be assessed by assays that measure neutrophil mediated opsonophagocytotic bactericidal activity.
  • the MAb's of the invention are useful for the treatment of systemic and local staphylococcal infections.
  • treatment encompasses any reduction, amelioration, or “alleviation” of existing infection as well as “blocking” or prophylaxis against future infection.
  • treatment with a MAb of the invention is said to “alleviate” staphylococcal nasal colonization if it is able to decrease the number of colonies in the nares of a mammal when the MAb is administered before, concurrently with, or after exposure to staphylococci, whether that exposure results from the intentional instillation of staphylococcus or from general exposure.
  • a MAb or collection of MAbs is considered to alleviate colonization if the extent of colonization, or the number of bacterial colonies that can be grown from a sample of nasal tissue, is decreased after administering the MAb or collection of MAbs.
  • a MAb or collection of MAbs alleviates colonization in the nasal colonization assays described herein when it reduces the number of colonies by at least 50%, at least 60%, at least 75%, at least 80%, or at least 90%. 100% alleviation may also be referred to as eradication.
  • a MAb is said to “block” staphylococcal colonization if it is able to prevent the nasal colonization of a human or non-human mammal when it is administered prior to, or concurrently with, exposure to staphylococci, whether by intentional instillation or otherwise into the nares.
  • a MAb blocks colonization, as in the nasal colonization assay described herein, if no staphylococcal colonies can be grown from a sample of nasal tissue taken from a mammal treated with the MAb of the invention for an extended period such as 12 hours or longer or 24 hours or longer compared to control mammals.
  • a MAb also blocks colonization in the nasal colonization assay described herein if it causes a reduction in the number of animals that are colonized relative to control animals.
  • a MAb is considered to block colonization if the number of animals that are colonized after administering the material and the Gram-positive bacteria is reduced by at least 25%, at least 50%, and at least 75%, relative to control animals or if no colonies can be grown from a sample taken from a treated individual for an extended period such as 12 hours or 24 hours or longer.
  • a MAb “blocks” colonization if it prevents future colonization in human patients who show no signs of prior colonization for an extended period of 12 or 24 hours or longer.
  • a MAb “alleviates” colonization if it causes a discernable decrease in the number of positive cultures taken from a human patient who is already positive for staphylococci before the MAbs of the invention are administered.
  • a vaccine is considered to confer a protective immune response if it stimulates the production of opsonic antibodies to gram-positive bacteria.
  • Production of opsonic antibodies may be measured by the presence of such antibodies in the serum of a test subject that has been administered the vaccine, relative to a control that has not received the vaccine.
  • the presence of opsonic antibodies in the serum may be measured by the activity assays described herein, or by other equivalent assays. If an opsonophagocytic bactericidal assay is used, then killing by the test serum of at least 50% more bacteria, 75% more bacteria, and at least 100% more bacteria, relative to the control serum, is considered to be enhanced immunity.
  • the present invention provides murine antibodies, including monoclonal antibodies, and chimeric, humanized and fully human antibodies, fragments, derivatives, and regions thereof, which bind to lipoteichoic acid (LTA) of Gram positive staphylococci.
  • Gram positive bacteria unlike Gram negative bacteria, take up the Gram stain as a result of a difference in the structure of the cell wall.
  • the cell walls of Gram negative bacteria are made up of a unique outer membrane of two opposing phospholipid-protein leaflets, with an ordinary phospholipid in the inner leaflet but the extremely toxic lipopolysaccharide in the outer leaflet.
  • the cell walls of Gram positive bacteria seem much simpler in comparison, containing two major components, peptidoglycan and teichoic acids plus additional carbohydrates and proteins depending on the species.
  • the antibodies of the claimed invention are broadly reactive against Gram positive bacteria. This broad reactivity permits the antibodies of the invention to block the binding of Gram positive bacteria to epithelial cells, such as human epithelial cells (50-54). Finally, these antibodies exhibit broad opsonic activity and consequently enhance phagocytosis and killing of Gram positive bacteria. Accordingly, the invention provides broadly reactive, opsonic, and protective antibodies for the diagnosis, prevention, and/or treatment of bacterial infections caused by Gram positive bacteria.
  • Gram positive Staphylococci against which the antibodies of the invention are directed are S. aureus (a coagulase positive bacteria) and S. epidermidis (a coagulase negative bacteria).
  • M110, M120, and MAb-391.4 bind strongly to LTA.
  • M110 and M120 also exhibit high opsonic activity for S. epidermidis
  • MAb-391.4 is also opsonic for S. epidermidis
  • M120 is also highly opsonic against S. aureus .
  • M110 was derived from mice immunized with whole S. epidermidis strain Hay as described in detail in U.S. Pat. No. 6,610,293, which is incorporated herein by reference.
  • hybridomas In screening for hybridomas, the antibodies of one clone (hybridoma line 96-105CE11 IF6, which produces antibody M110) were found to bind very strongly to Gram positive bacteria such as strain Hay, all three serotypes of S. epidermidis, S. hemolyticus, S. hominus , and two serotypes of S. aureus , but not to the Gram negative control, Haemophilus influenza (see U.S. Pat. No. 6,610,293).
  • M120 was derived from mice immunized with conjugates of S. aureus LTA.
  • the antibodies of one clone (00-107GG12 ID12, which produces antibody M120) were found to bind strongly to LTA, and were opsonic for S. aureus type 5 and S. epidermidis strain Hay.
  • MAb-391.4 is from QED Biosciences, and was derived from mice immunized with whole UV-killed S. aureus.
  • variable regions of M110, M120, and 391.4 were sequenced and compared, revealing a surprising 88% identity (203/230) at the amino acid level. Further, the level of identity was found to be 96% (220/230) between the antibodies that are highly opsonic for S. epidermidis , M110 and M120. We believe that this level of homology between three monoclonal antibodies that were raised in three different mice, using three different antigen preparations from two different types of bacteria, is unprecedented. To understand how unexpected this finding is, one need only consider how vast and diverse is the collection of antibodies in the immune system.
  • the immune system is made up of a large number of B cells, each bearing antibodies of a different specificity, but only about 1 in 10,000 to 1 in 1,000,000 B cells is specific for a particular antigen.
  • a foreign antigen such as is found on the surface of a bacteria
  • the appropriate B cell recognizes that antigen and then enters a lymph node where it undergoes rapid division to produce many progeny bearing the identical specificity.
  • the rapidly dividing B cells also undergo somatic hypermutation. Somatic hypermutation results in about half of the B cells acquiring mutations in their rearranged heavy and light chain genes, with mutation occurring preferentially in complementarity determining regions (CDRs) of the variable regions.
  • CDRs complementarity determining regions
  • B cells that retain their ability to bind antigen continue to secrete antibody, while those that no longer bind antigen undergo apoptosis. As the antigen is cleared from the host, only B cells that have very high antigen affinity survive in a process called affinity maturation. The surviving activated B cells differentiate into plasma cells, which are short-lived and secrete antibody, and memory B cells, which are long-lived lymphocytes bearing membrane-bound antibody that can be rapidly stimulated when the antigen is re-introduced.
  • M110, M120, and MAb-391.4 were not only raised in different mice, but with different immunogens: M110 was raised to whole S. epidermidis , M120 was raised to purified and conjugated S. aureus LTA, and MAb-391.4 was raised to whole UV-killed S. aureus . Yet, though these antibodies were raised against different immunogen preparations in different mice, they share 88% identity at the amino acid level in both the heavy and light chain variable regions. This high degree of homology suggests that LTA contains a highly antigenic, and highly conserved, epitope which is bound by the three antibodies in a very similar manner. This epitope and mode of binding may be responsible for the high opsonic activity of the monoclonal antibodies.
  • MAb A110 is currently being manufactured under GMP conditions in preparation for clinical trials. Additional disclosure regarding the MAb A110 is provided in U.S. Provisional Application Ser. No. 60/341,806, and in related application Methods for Blocking or Alleviating Staphylococcal Nasal Colonizaton by Intranasal Application of Monoclonal Antibodies, filed concurrently herewith, both of which are expressly incorporated by reference.
  • one aspect of the invention relates to antibodies that bind to the LTA of Gram positive bacteria, including both coagulase negative ( S. epidermidis ) and coagulase positive ( S. aureus ) bacteria, and that enhance the opsonization of such bacteria.
  • anti-LTA antibodies include monoclonal antibodies, such as M110, M120, and MAb-391.4, chimeric monoclonal antibodies A110, A120, A120a, and A120b, and other monoclonal antibodies including, chimeric, humanized, fully human antibodies, antibody fragments, and modified antibodies.
  • the antibody is a chimeric mouse/human antibody made up of regions from the anti-LTA antibodies of the invention together with regions of human antibodies.
  • Chimeric or other monoclonal antibodies are advantageous in that they avoid the development of anti-murine antibodies.
  • patients administered murine anti-TNF (tumor necrosis factor) monoclonal antibodies developed anti-murine antibody responses to the administered antibody (5).
  • This type of immune response to the treatment regimen commonly referred to as the human anti-mouse antibody response, or the HAMA response, decreases the effectiveness of the treatment and may even render the treatment completely ineffective.
  • Humanized or chimeric human/mouse monoclonal antibodies have been shown to significantly decrease the HAMA response and to increase the therapeutic effectiveness (19).
  • a chimeric heavy chain can comprise the antigen binding region of the heavy chain variable region of the anti-LTA antibody of the invention linked to at least a portion of a human heavy chain IgG, IgA, IgM, or IgD constant region.
  • This humanized or chimeric heavy chain may be combined with a chimeric light chain that comprises the antigen binding region of the light chain variable region of the anti-LTA antibody linked to at least a portion of the human light chain kappa or lambda constant region.
  • Exemplary embodiments include, but are not limited to, an antibody having a mouse heavy chain variable region fused to a human IgG i constant region, and a mouse light chain variable region fused to a human kappa light chain constant region.
  • the chimeric antibodies and other MAb's of the invention may be monovalent, divalent, or polyvalent immunoglobulins.
  • a monovalent chimeric antibody is a dimer (HL) formed by a chimeric H chain associated through disulfide bridges with a chimeric L chain, as noted above.
  • a divalent chimeric antibody is a tetramer (H2 L 2 ) formed by two HL dimers associated through at least one disulfide bridge.
  • a polyvalent or multivalent chimeric antibody may be based on an aggregation of chains, with or without a carrier or scaffold.
  • the MAbs of the invention include antibodies that contain heavy and light chain variable regions derived from two different antibodies.
  • the heavy and light chain variable regions are derived from two antibodies that bind to the same molecule, e.g. LTA.
  • Exemplary embodiments include A120a, which is a human/mouse chimeric antibody that has a heavy chain variable region from A110 and a light chain variable region from A120; and A120b, which is a human/mouse chimeric antibody that has a heavy chain variable region from A120 and a light chain variable region from A110.
  • Additional exemplary embodiments include antibodies that comprise a heavy chain variable region from MAb-391.4, and a light chain variable region from either of A110 or A120, and antibodies that comprise a light chain variable region from MAb-391.4, and a heavy chain variable region from either of A110 or A120.
  • the invention is a collection of opsonic monoclonal antibodies that bind to LTA and that exhibit a high degree of homology in the variable regions at either the amino acid or nucleic acid level, or both.
  • this collection comprises one or more of M110, M120, their human/mouse chimeric counterparts, A110, A120, and MAb-391.4.
  • the amino acid sequences of the variable regions are at least 75% identical, at least 80% identical, at least 85% identical, at least 88% identical, at least 90% identical, or at least 95% identical as defined above.
  • the present invention also encompasses the DNA sequences of the genes coding for the antibodies (see, e.g., FIGS. 5 , 6 , and 9 ; SEQ ID NOs: 11, 13-15, 19, and 20) as well as the polypeptides encoded by the DNA (see, e.g., FIGS. 5 , 6 , and 10 ; SEQ ID NOs: 10, 12, 16, 17, 21, and 22).
  • Those figures provide the variable regions of the heavy and light chains of A110, A120, and MAb-391.4, including the complementarity determining regions (CDRs), the hypervariable amino acid sequences within antibody variable regions that usually interact with the antigen.
  • CDRs complementarity determining regions
  • the DNA and amino acid sequence homology between A110 and A120 is striking. There is a 94% homology (216/229) at the amino acid level and a 96% homology (662/687) at the DNA level between the antibodies. This suggests that these antibodies share a sequence and structural similarity.
  • the invention includes peptide sequences for, and DNA sequences encoding, full-length antibodies and portions thereof, as well as CDRs and FRs relating to these MAbs.
  • the invention further includes DNA and peptide sequences that are homologous to these sequences. In one embodiment, these homologous DNAs and peptide sequences are about 70% identical, although other embodiments include sequences that are about 75%, 80%, 85%, 88%, 90%, and 95% or more identical. As indicated above, determining levels of identity for both the DNA and peptide sequence is well within the routine skill of those in the art.
  • alignment of the A110, A120, and 391.4 light chain variable regions shows identical amino acids in 95 of 106 amino acids, or more than 89% identity overall.
  • the percent identity is about 93% (68 out of 73 amino acids). It is predicted that light chain variable regions with a somewhat lower overall identity would still form MAbs that specifically bind LTA, and are therefore within the scope of the invention.
  • the CDRs themselves show at least 88% identity, in particular, CDR1 (amino acids 24-33), CDR2 (amino acids 49-55), and CDR3 (amino acids 88-96), show 9/10, 7/7, and 8/9 identical amino acids.
  • the framework regions (FRs) surrounding the CDRs are also highly conserved: amino acids 1-23 of SEQ ID Nos. 16, 10, and 21 show greater than 86% identity (20/23 matching amino acids); amino acids 34-48 show about 93% identity (14/15); amino acids 56-87 show about 93% identity (68/73); and amino acids 97-106 show 70% identity.
  • FIG. 10B alignment of the A110, A120, and 391.4 heavy chain variable regions (Seq. ID Nos. 17, 12, and 22, respectively) also shows a high degree of sequence identity. Counting single amino acid gaps and insertions as single-point mis-matches, Seq. ID Nos. 17, 12, and 22 show 86% identity overall (108/125 identical amino acids). It is predicted that heavy chain variable regions with a somewhat lower overall identity would still form MAbs that specifically bind LTA, and are therefore within the scope of the invention. The degree of identity is particularly high in the FR region preceding CDR1 through the FR region preceding CDR3, in particular, the 96 base region from amino acid 16 to 101 of Seq. ID Nos.
  • CDR1 itself, shows 90% identity over 10 amino acids (amino acids 26-35), and CDR2 (amino acids 50-69) shows about 89% identity over 19 amino acids.
  • the framework regions surrounding the CDRs are also highly conserved.
  • Amino acids 1-25 of SEQ ID Nos. 17, 12, and 22 show 92% identity (23/25 matching amino acids); amino acids 36-49 show 100% identity over 14 amino acids; the FR region between CDR2 and CDR3 (amino acids 70 to about 101) shows about 87% identity (over 31-32 amino acids); and amino acids 115-125 show 90% identity.
  • the invention encompasses polypeptides (including regions of larger polypeptides, such as MAbs) that 1) exhibit high sequence homology to Seq. ID Nos. 10, 12, 16, 17, 21, or 22, or defined regions thereof, and 2) are capable of functioning as all or part of the variable region of a MAb that specifically binds LTA.
  • polypeptides comprise, or are at least 70%, 75%, 77% 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 93%, 95% identical to, any of Seq. ID Nos. 10, 12, 16, 17, 21, or 22.
  • polypeptides within the scope of the invention may be less than 100%, 99%, 95%, 90%, 80% or less identical to Seq. ID Nos. 10, 12, 16, 17, 21, or 22 provided that they are capable of functioning as all or part of the variable region of a MAb that specifically binds to LTA.
  • polypeptides within the scope of the invention comprise, or are at least 70%, 75%, 77%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 93%, 95% identical to, amino acids 24 to 96 of any of Seq. ID Nos. 10, 16, or 21.
  • such polypeptides comprise, or are at least 70%, 75%, 77%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 93%, 95% identical to, 1) amino acids 24-33, 49-55, and 88-73 of Seq. ID Nos. 10, 16, or 21, or 2) amino acids 26-35 or 50-69 of Seq. ID Nos.
  • such polypeptides comprise, or are at least 70%, 75%, 77%, 80%, 81%, 82%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 93%, 95% identical to, 1) amino acids 1-23, 34-48, 56-87, and 97-106 of Seq. ID Nos. 10, 12, 16, 17, 21, or 22, or 2) amino acids 1-25, 36-49, 70-101, or 115-125 of Seq. ID Nos. 12,17, or 22; and are capable of functioning as a framework region, or portion thereof, in a MAb that specifically binds to LTA.
  • the invention further comprises collections of a multiplicity of any of the above sequences capable of functioning as all or part of the variable region of a MAb that specifically binds to LTA, as part of a larger polypeptide, MAb, collection of MAbs or aggregation of MAbs; and the use thereof in prophylaxis, treatment, and for the production of pharmaceutical compounds or medicaments.
  • the invention further comprises any non-naturally occurring RNA, DNA, or vector thereof, encoding any of the above sequences capable of functioning as all or part of the variable region of a MAb that specifically binds to LTA, as well as plasmids, viruses, bacteria, yeast, microorganisms, cell lines, transgenic plants or animals harboring or expressing such nucleic acids.
  • the invention contemplates production systems for Mabs, light chains, heavy chains, and portions thereof, comprising 1) a cell (including bacteria, yeast, microorganisms, eukaryotic cell lines, transgenic plant or animal) in connection with 2) at least one recombinant nucleic acid capable of directing the expression of any of the Mabs or related polypeptides of the invention.
  • the invention thus further comprises a general method of identifying highly antigenic and highly conserved epitopes by raising antibodies against different immunogen preparations in different mice, sequencing the variable regions of the antibodies, comparing the variable regions, and identifying antibodies that share a high degree of homology in the variable regions.
  • the DNA sequences of the invention can be identified, isolated, cloned, and transferred to a prokaryotic or eukaryotic cell for expression by procedures well-known in the art. Such procedures are generally described in Molecular Cloning: A Laboratory Manual , as well as Current Protocols in Molecular Biology (44, 45), which are incorporated by reference. Guidance relating more specifically to the manipulation of sequences of the invention may be found in Antibody Engineering , and Antibodies: A Laboratory Manual (64, 65), both of which are incorporated by reference in their entirety.
  • a CDR can be grafted onto any human antibody framework region using techniques standard in the art, in such a manner that the CDR maintains the same binding specificity as in the intact antibody.
  • an antibody that has its CDRs grafted onto a human framework region is said to be “humanized”.
  • Humanized, and fully human antibodies generally also include human constant regions, thus maximizing the percentage of the antibody that is human-derived, and potentially minimizing the HAMA response.
  • DNA and peptide sequences of the antibodies of the invention may form the basis of antibody “derivatives,” which include, for example, the proteins or peptides encoded by truncated or modified genes. Such proteins or peptides may function similarly to the antibodies of the invention. Other modifications, such as the addition of other sequences that may enhance the effector function, which includes phagocytosis and/or killing of the bacteria, are also within the present invention.
  • the present invention also discloses a pharmaceutical composition
  • a pharmaceutical composition comprising the antibodies, whether monoclonal or chimeric, humanized, or fully human, together with a pharmaceutically acceptable carrier.
  • the pharmaceutical compositions of the invention may alternatively comprise the isolated antigen, epitope, or portions thereof, together with a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers can be sterile liquids, such as water, oils, including petroleum oil, animal oil, vegetable oil, peanut oil, soybean oil, mineral oil, sesame oil, and the like. Saline solutions, aqueous dextrose, and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, 18 th Edition (13), which is herein incorporated by reference.
  • the invention may be practiced with various delivery vehicles and/or carriers.
  • Such vehicles may increase the half-life of the MAbs in storage and upon administration including, but not limited to, application to skin, wounds, eyes, lungs, or mucus membranes of the nasal or gastrointestinal tract, or upon inhalation or instillation into the nares.
  • These carriers comprise natural polymers, semi-synthetic polymers, synthetic polymers, lipososmes, and semi-solid dosage forms (21, 29, 33, 35, 36, 46).
  • Natural polymers include, for example, proteins and polysaccharides.
  • Semi-synthetic polymers are modified natural polymers such as chitosan, which is the deacetylated form of the natural polysaccharide, chitin.
  • Synthetic polymers include, for example, polyphosphoesters, polyethylene glycol, poly(lactic acid), polystyrene sulfonate, and poly(lactide coglycolide).
  • Semi-solid dosage forms include, for example, dendrimers, creams, ointments, gels, and lotions. These carriers can also be used to microencapsulate the MAbs or be covalently linked to the MAbs.
  • the present invention provides methods for treating a patient infected with, or suspected of being infected with, a Gram-positive bacteria such as a staphylococcal organism.
  • the method comprises administering a therapeutically effective amount of a pharmaceutical composition comprising the anti-LTA immunoglobulin (whether monoclonal, chimeric, humanized, or fully human, including fragments, regions, and derivatives thereof) and a pharmaceutically acceptable carrier.
  • a patient can be any human or non-human mammal in need of prophylaxis or other treatment. Representative patients include any mammal subject to S.
  • aureus or other staphylococcal or Gram-positive infection or carriage including humans and non-human animals such as mice, rats, rabbits, dogs, cats, pigs, sheep, goats, horses, primates, ruminants including beef and milk cattle, buffalo, camels, as well as fur-bearing animals, herd animals, laboratory, zoo, and farm animals, kenneled and stabled animals, domestic pets, and veterinary animals.
  • non-human animals such as mice, rats, rabbits, dogs, cats, pigs, sheep, goats, horses, primates, ruminants including beef and milk cattle, buffalo, camels, as well as fur-bearing animals, herd animals, laboratory, zoo, and farm animals, kenneled and stabled animals, domestic pets, and veterinary animals.
  • a therapeutically effective amount is an amount reasonably believed to provide some measure of relief, assistance, prophylaxis, or preventative effect in the treatment of the infection.
  • a therapeutically effective amount may be an amount believed to be sufficient to block a bacterial infection.
  • a therapeutically effective amount may be an amount believed to be sufficient to alleviate a bacterial infection.
  • Such therapy as above or as described below may be primary or supplemental to additional treatment, such as antibiotic therapy, for a staphylococcal infection, an infection caused by a different agent, or an unrelated disease. Indeed, combination therapy with other antibodies is expressly contemplated within the invention.
  • a further embodiment of the present invention is a method of preventing such infections, comprising administering a prophylactically effective amount of a pharmaceutical composition comprising the anti-LTA antibody (whether monoclonal, chimeric, humanized, or fully human) and a pharmaceutically acceptable carrier.
  • a prophylactically effective amount is an amount reasonably believed to provide some measure of prevention of infection by Gram positive bacteria.
  • Such therapy as above or as described below may be primary or supplemental to additional treatment, such as antibiotic therapy, for a staphylococcal infection, an infection caused by a different agent, or an unrelated disease. Indeed, combination therapy with other antibodies is expressly contemplated within the invention.
  • the antibodies and the pharmaceutical compositions of the invention may be administered by intravenous, intraperitoneal, intracorporeal injection, intra-articular, intraventricular, intrathecal, intramuscular or subcutaneous injection, or intranasally, dermally, intradermally, intravaginally, orally, or by any other effective method of administration.
  • the composition may also be given locally, such as by injection to the particular area infected, either intramuscularly or subcutaneously.
  • Administration can comprise administering the pharmaceutical composition by swabbing, immersing, soaking, or wiping directly to a patient.
  • the treatment can also be applied to objects to be placed within a patient, such as dwelling catheters, cardiac valves, cerebrospinal fluid shunts, joint prostheses, other implants into the body, or any other objects, instruments, or appliances at risk of becoming infected with a Gram positive bacteria, or at risk of introducing such an infection into a patient.
  • objects to be placed within a patient such as dwelling catheters, cardiac valves, cerebrospinal fluid shunts, joint prostheses, other implants into the body, or any other objects, instruments, or appliances at risk of becoming infected with a Gram positive bacteria, or at risk of introducing such an infection into a patient.
  • compositions of the invention may be the reduction in cytokine release that results from the introduction of the LTA of a Gram positive bacteria (49).
  • LTA induces cytokines, including, for example, tumor necrosis factor alpha, interleukin 6, and interferon gamma (see, e.g., (37)).
  • compositions of the invention may enhance protection at three levels: (1) by binding to LTA on the bacteria and thereby blocking the initial binding to epithelial cells and preventing subsequent invasion of the bacteria; (2) by binding to LTA on bacteria and thereby enhancing opsonization of the bacteria and clearance of the bacteria from tissues and/or blood; and/or (3) by binding to LTA and partially or fully blocking cytokine release and modulating the inflammatory responses to prevent shock and tissue destruction.
  • S. aureus , type 5 is deposited at the ATCC under Accession No. 49521.
  • Hybridoma 96-105CE11 IF6 (M110) was deposited at the American Type Culture Collection (ATCC), located at P.O. Box 1549, Manassas, Va. 20108, U.S.A., on Jun. 13, 1997, under ATCC Accession No. HB-12368.
  • Hybridoma 00-107GG12 ID12 was deposited at the American Type Culture Collection (ATCC), located at P.O. Box 1549, Manassas, Va. 20108, U.S.A., on Aug. 16, 2001, under ATCC Accession No. PTA-3644.
  • ATCC American Type Culture Collection
  • Hybridoma 391.4 was deposited at the American Type Culture Collection (ATCC), located at P.O. Box 1549, Manassas, Va. 20108, U.S.A., on Dec. 18, 2001, under ATCC Accession No. PTA-3932.
  • ATCC American Type Culture Collection
  • Isotype was determined using a mouse immunoglobulin isotype kit obtained from Zymed Laboratories (Cat. No. 90-6550).
  • immunoglobulin is incubated with a preparation of whole cell staphylococci or with a preparation of bacterial cell wall components such as LTA or PepG.
  • the binding assay may be an agglutination assay, a coagulation assay, a colorimetric assay, a fluorescent binding assay, or any other suitable binding assay that is known in the art.
  • a particularly suitable assay is either an enzyme-linked immunosorbent assay (ELISA) or a radio-immunoassay (RIA). Binding is detected directly and can also be detected indirectly by using competitive or noncompetitive binding procedures known in the art.
  • the whole cell staphylococcus preparation, LTA preparation, PepG preparation, or a combination of those preparations, may be fixed using standard techniques to a suitable solid support, including, but not limited to, a plate, a well, a bead, a micro-bead, a paddle, a propeller, or a stick.
  • Solid supports may be comprised of, for example, glass or plastic.
  • the solid support is a microtiter plate.
  • a binding assay requires the following steps. First, the fixed preparation is incubated with an immunoglobulin source.
  • the immunoglobulin source is, for example, tissue culture supernatant or a biological sample such as ascites, plasma, serum, whole blood, or body tissue.
  • the immunoglobulin may be further isolated or purified from its source by means known in the art, and the purified or isolated immunoglobulin is subsequently used in the assay.
  • the amount of binding is determined by comparing the binding in a test sample to the binding in a negative control.
  • a negative control is defined as any sample that does not contain antigen-specific immunoglobulin.
  • a positive binding reaction results when the amount of binding observed for the test sample is greater than the amount of binding for a negative control.
  • Positive binding may be determined from a single positive/negative binding reaction or from the average of a series of binding reactions.
  • the series of binding reactions may include samples containing a measured amount of immunoglobulin that specifically binds to the fixed antigen, thereby creating a standard curve. This standard curve may be used to quantitate the amount of antigen-specific immunoglobulin in an unknown sample.
  • antibodies are fixed to a solid support and an unknown immunoglobulin sample is characterized by its ability to bind a bacterial preparation.
  • LBE Live Bacteria ELISA
  • the bacteria were again pelleted by centrifugation, the supernatant discarded and the bacteria resuspended in PBS/BSA to a percent transmittance (% T) of 65%-70% at 650 nm. From this suspension the bacteria were further diluted 15-fold in sterile 0.9% sodium chloride (Sigma cat. no. S8776, or equivalent), and 100 ⁇ l of this suspension was added to replicate wells of a flat-bottomed, sterile 96-well plate.
  • % T percent transmittance
  • Each antibody to be tested was diluted to the desired concentration in PBS/BSA containing 0.05% Tween-20 and horse radish peroxidase-conjugated Protein A (Protein A-HRP, Zymed Laboratories) at a 1:8000 dilution (PBS/BSA/Tween/Prot A-HRP).
  • the Protein A-HRP was allowed to bind to the antibodies for 30-60 minutes at room temperature before use, thereby generating an antibody-Protein A-HRP complex to minimize the potential non-specific binding of the antibodies to the Protein A found on the surface of S. aureus .
  • several dilutions of test antibody were used in each assay.
  • the bacteria were pelleted in the plate by centrifugation at 1800-2000 ⁇ g for 10-15 minutes at room temperature. The supernatant was carefully removed from the wells and 200 ⁇ l of PBS/BSA containing 0.05% Tween-20 (PBS/BSA/Tween) was added to all wells to dilute unbound reagents. The bacteria were again pelleted by centrifugation and the supernatant was removed. Two hundred ⁇ l of PBS/BSA/Tween was again added to all wells and the bacteria were again pelleted by centrifugation as described above. The supernatant was removed and 100 ⁇ l of TMB substrate (BioFx, Inc. cat. no.
  • TMBW-0100-01, or equivalent was added to each well and the hydrolysis of the substrate was allowed to proceed for 15 minutes at room temperature. The reactions were stopped by adding 100 ⁇ l of TMB stop reagent (450 nm Stop Reagent; BioFx, Inc. catalog no. STPR-0100-01, or equivalent). The absorbance of each well was determined using a microplate reader fitted with a 450 nm filter.
  • the bacteria-coated plates were washed four times with phosphate buffered saline containing 0.05% Tween-20 (PBS-T) as follows. Approximately 250 microliters of PBS-T was added to each well. The buffer was removed by flicking the plate over the sink and the remaining buffer removed by inverting the plate and tapping it on absorbent paper. The antibody was diluted in PBS-T and then added to the wells. Supernatants, ascites, or purified antibodies were tested at the dilutions indicated in the Examples. Control wells received PBS-T alone. After addition of the antibody, the wells were incubated at room temperature for 30-60 minutes in a draft-free environment.
  • PBS-T phosphate buffered saline containing 0.05% Tween-20
  • the wells were again washed four times with PBS-T.
  • Ninety-five microliters of detection antibody was then added to each well.
  • the detection antibody was one of the following: rabbit anti-mouse IgG3, rabbit anti-mouse IgM, or goat anti-human IgG (gamma-specific), all conjugated to horse radish peroxidase (HRP) and diluted 1:6000 in PBS-T (Zymed catalog numbers 61-0420, 61-6820 and 62-8420, respectively).
  • Immunoassay with Protein A In order to evaluate the binding of the MAbs to S. aureus , the immunoassay procedure was modified for methanol-fixed bacteria, described above. Because S. aureus expresses Protein A on its surface, and Protein A binds strongly to the constant region of the heavy chains of gamma-globulins, it is possible that false positive results may be obtained from non-specific binding of the antibodies to Protein A. To overcome this difficulty, the immunoassay wells were coated with bacteria as described above, but prior to the addition of the antibodies to the bacteria-coated wells, the MAbs were incubated with a solution of recombinant Protein A conjugated to HRP (Zymed Laboratories Cat. No.
  • Protein A-HRP-MAb solutions were allowed to bind in the coated wells for 30-60 minutes at room temperature. The wells were then washed with PBS-T and TMB substrate solution was added and the assay completed as described above.
  • Immunoassay on LTA The binding of the MAbs to LTA was measured by immunoassay on wells coated with S. aureus LTA (Sigma Cat. No. 2515). One hundred microliters of a 1 ⁇ g/ml LTA solution in PBS was distributed into replicate Nunc Maxisorp Stripwells and incubated overnight at room temperature. The unbound material was removed from the wells by washing four times with PBS-T. Antibody, diluted in PBS-T, was then added to the wells and the assay continued as described above for the Immunoassay on Methanol-Fixed Bacteria.
  • Antibodies that bind to an antigen may not necessarily enhance opsonization or enhance protection from infection. Therefore, an opsonization assay was used to determine the functional activities of the antibodies.
  • An opsonization assay can be a colorimetric assay, a chemiluminescent assay, a fluorescent or radiolabel uptake assay, a cell-mediated bactericidal assay, or any other appropriate assay known in the art which measures the opsonic potential of a substance and thereby identifies reactive immunoglobulin.
  • an infectious agent, a eukaryotic cell, and the opsonizing substance to be tested, or an opsonizing substance plus a purported opsonizing enhancing substance are incubated together.
  • the opsonization assay is a cell-mediated bactericidal assay.
  • an infectious agent such as a bacterium, a phagocytic cell, and an opsonizing substance such as immunoglobulin, are incubated together.
  • Any eukaryotic cell with phagocytic or binding ability may be used in a cell-mediated bactericidal assay.
  • phagocytic cells are macrophages, monocytes, neutrophils, or any combination of these cells. Complement proteins may be included to promote opsonization by both the classical and alternate pathways.
  • the opsonic ability of an antibody is determined by the amount or number of infectious agents remaining after incubation. The fewer the number of infectious agents that remain after incubation, the greater the opsonic activity of the antibody tested.
  • opsonic activity is measured by comparing the number of surviving bacteria between two similar assays, only one of which contains the antibody being tested.
  • opsonic activity is determined by measuring the number of viable organisms before and after incubation with a sample antibody. A reduced number of bacteria after incubation in the presence of antibody indicates a positive opsonizing activity.
  • positive opsonization is determined by culturing the incubation mixture under appropriate bacterial growth conditions. Any reduction in the number of viable bacteria comparing pre-incubation and post-incubation samples, or between samples which contain immunoglobulin and those that do not, is a positive reaction.
  • Neutrophil-Mediated Opsonophagocytic Bactericidal Assay The assay was performed using neutrophils isolated from adult venous blood by sedimentation using PMN Separation Medium (Robbins Scientific catalog no. 1068-00-0). Forty microliters of antibody, serum, or other immunoglobulin source, was added at various dilutions to replicate wells of a round-bottom microtiter plate. Forty microliters of neutrophils (approximately 2 ⁇ 10 6 cells per well) was then added to each well, followed immediately by approximately 3 ⁇ 10 4 mid-log phase bacteria ( S. epidermidis strain Hay, ATCC 55133 or S. aureus type 5, gift from S.
  • each aliquot was diluted 20-fold in a solution of 0.1% BSA in water (to lyse the PMNs), mixed vigorously by rapid pipetting, and cultured on blood agar plates (Remel, cat. no. 01-202, or equivalent) overnight at 37° C.
  • the opsonic activity was measured by comparing the number of bacterial colonies observed from the sample taken at two hours with the nurjnber of bacterial colonies observed from the sample taken at time zero. Colonies were enumerated using an IPI Minicount Colony Counter.
  • Nasal Colonization Assay The mouse nasal colonization model for S. aureus was based on the work of Kiser et al. (47). Briefly, streptomycin resistant S. aureus type 5 is grown on high salt Columbia agar (Difco) to promote capsule formation. The bacteria are washed with sterile saline (0.9% NaCI in water) to remove media components and resuspended at ⁇ 10 8 bacteria/animal dose in saline (0.9% NaCl in water) containing various concentrations and combinations of anti-staphylococcal or irrelevant control MAbs.
  • the bacteria are pelleted and resuspended in a final volume of 10 ⁇ l per animal dose in either saline or saline containing antibody.
  • Mice that have been maintained on streptomycin-containing water for 24 hours are sedated with anesthesia. Staphylococci are injected into the nares of the mice by pipetting without contacting the nose.
  • LTA conjugates LTA/PspA, LTA/SIA/TT, and LTA/GMBS/TT, prepared as set forth below, were used.
  • LTA was first derivatized with thiol groups as follows.
  • S. aureus LTA (Sigma Chemical Co.) was purified essentially as described in Fischer et al. (9). The purified LTA was diluted to 4 mg/ml with water.
  • the reaction was incubated for 4 hours at room temperature, and then 55 ⁇ l of 0.5 M DTT was added and the solution was dialyzed overnight at 4° C. against 2 mM EDTA, pH 5 (2 ⁇ 1 L).
  • the reaction resulted in 0.27 mM thiol (LTA-SH) in a 1.2 mL volume, or 0.32 ⁇ mol thiol, as determined by DTNB assay (2).
  • LTA/PspA conjugate was prepared as follows. Three milligrams of pneumococcal surface protein A (PspA; 188 ⁇ l of a 16 mg/ml solution in PBS; prepared essentially as described in Wortham et al. (43) was combined with 25 ⁇ l of 0.75 M HEPES, 10 mM EDTA, pH 7.3 and 17 ul 0.1 M N-hydroxysuccinimidyl iodoacetate (SIA; Bioaffinity Systems) and incubated for 2 hours at room temperature.
  • PspA pneumococcal surface protein A
  • SIA N-hydroxysuccinimidyl iodoacetate
  • the volume of the solution was then made up to 2 ml with 10 mM sodium acetate, 0.15 M NaCl, 2 mM EDTA, and then concentrated to a final volume of about 150 ⁇ l using an Ultrafree 4 device (30 kDa cutoff; Amicon).
  • the resulting iodoacetyl PspA was then combined with 400 ⁇ l of LTA-SH.
  • the pH was raised to 8 with 1 M HEPES pH 8, and the reaction proceeded overnight at 4° C.
  • the solution was then fractionated on a 1 ⁇ 60 cm S-200HR column, which had been equilibrated with 0.1% deoxycholate (DOC) in PBS.
  • DOC deoxycholate
  • the void volume fractions were pooled, dialyzed into saline (0.15 M NaCl) to remove DOC and PBS, and had an optical density of 0.14 at 280 nm.
  • the concentration of protein in the conjugate solution was 0.66 mg/ml by BCA assay (Pierce Chemical Company), and the concentration of phosphate in the conjugate solution was 0.88 mM by phosphate assay (1).
  • LTA/SIA/TT conjugate was prepared as follows. Four milligrams of tetanus toxoid (TT; 280 ⁇ l of 14.5 mg/ml; SmithKline Beecham), diluted to 4 ml with 2 M NaCl was concentrated to 50 ⁇ l using an Ultrafree 4 centrifugal filter with a 30 kD cutoff (Millipore). The resulting solution was diluted to 250 ⁇ l with 2 M NaCl (TT/2 M NaCl).
  • iodoacetylated TT was combined with 400 ⁇ l of LTA-SH. The reaction is incubated overnight at 4° C. The reaction was fractionated on a 1 ⁇ 60 cm Sephacryl S-200HR column (Pharmacia), equilibrated with 0.1% deoxycholate in PBS. The void volume fractions, containing the LTA/SIA/TT conjugate, were pooled, dialyzed into saline to remove DOC and PBS, and had an optical density of 0.77 at 280 nm. The yield of TT in the conjugate was 0.77 mg/ml by BCA assay (Pierce Chemical Co.). The concentration of phosphate in the conjugate solution was 0.77 mM by phosphate assay (1).
  • LTA/GMBS/TT conjugate was prepared as follows. Four milligrams of TT (280 ⁇ l of 14.5 mg/ml; SmithKline Beecham), diluted to 4 ml with 2 M NaCl was concentrated to 50 ⁇ l using an Ultrafree 4 centrifugal filter with a 30 kD cutoff (Millipore). The resulting solution was diluted to 250 ul with 2 M NaCl (TT/2 M NaCl).
  • mice Twenty-four approximately 4 month old female BALB/c mice were separated into six groups and immunized with 10 ⁇ g (groups A, C, and E) or 1 ⁇ g (groups B, D, and F) of LTA/PspA (groups A and B), LTA/SIA/TT (groups C and D), or LTA/GMBS/TT (groups E and F; Table 1).
  • mice All immunizations were administered subcutaneously in 50% RIBI adjuvant.
  • the mice received a boost 21 days after the primary immunization, and a second boost 79 days after the primary immunization.
  • Boosts were performed as described for the primary immunizations. Eyebleeds were taken at 0 days, 21 days, 35 days, 79 days, 94 days, and 119 days after the primary immunization. Serum collected at 21 days and 35 days was tested by ELISA for antibodies to LTA (Table 2).
  • Serum collected at 35 days and 79 days was also tested for antibodies to LTA by ELISA, and serum collected at 94 days was tested by ELISA, and in an LBE assay against S. epidermidis strain Hay and S. aureus (Table 3).
  • Mouse 1396 which had been immunized with LTA/GMBS/TT, was selected because serum from the mouse showed a strong signal by anti-LTA ELISA, and was opsonic against S. aureus .
  • Mouse 1396 was boosted one more time at day 134, and then sacrificed on day 141, and spleen removed and used to make hybridomas.
  • Hybridomas were prepared by the general methods of Shulman, Wilde and Kohler; and Bartal and Hirshaut (34, 48). A total of 2.08 ⁇ 10 8 spleenocytes from mouse 1396 were mixed with 2.00 ⁇ 10 7 SP2/0 mouse myeloma cells (ATCC Catalog number CRL1581) and pelleted by centrifugation (400 ⁇ g, 10 minutes at room temperature) and washed in serum free medium.
  • the supernatant was removed to near-dryness and fusion of the cell mixture was accomplished in a sterile 50 ml centrifuge conical by the addition of 1 ml of warm (37° C.) polyethylene glycol (PEG; mw 1400; Boehringer Mannheim) over a period of 60-90 seconds.
  • PEG polyethylene glycol
  • the PEG was diluted by slow addition of serum-free medium in successive volumes of 1, 2, 4, 8, 16 and 19 mls.
  • the hybridoma cell suspension was gently resuspended into the medium and the cells pelleted by centrifugation (500 ⁇ g, 10 minutes at room temperature).
  • the supernatant was removed and the cells resuspended in medium RPM11640, supplemented with 10% heat-inactivated fetal bovine serum, 0.05 mM hypoxanthine and 16 ⁇ M thymidine (HT medium).
  • medium RPM11640 supplemented with 10% heat-inactivated fetal bovine serum, 0.05 mM hypoxanthine and 16 ⁇ M thymidine (HT medium).
  • HT medium 16 ⁇ M thymidine
  • One hundred ⁇ l of the hybridoma cells were planted into 952 wells of 96-well tissue culture plates. Eight wells (column 1 of plate A) received approximately 2.5 ⁇ 10 4 SP2/0 cells in 100 ⁇ l. The SP2/0 cells served as a control for killing by the selection medium added 24 hours later.
  • the absorbance values for eleven of the culture supernatants were less than 0.100. However, the absorbance value obtained with the supernatant from hybridoma culture 00-107GG12 was 4.000. This culture was expanded for further evaluation and cloned into two 96-well culture dishes. Cloning was accomplished by diluting the cell suspension into 4.5 viable cells per ml in RPM11640, supplemented with 15% fetal bovine serum, 5% Hybridoma SFM (Life Technologies) and 100 ⁇ g/ml of kanamycin.
  • Hybridoma clone 00-107GG12 ID12 was expanded and cryopreserved. Isotype determination revealed that both the original hybridoma (00-107GG12) and its clone (00-107GG12 ID12) were mouse IgG2a heavy chains with kappa light chains.
  • the monoclonal antibody produced by hybridoma 00-107GG12 ID12 was designated M120.
  • Opsonic assays were carried out substantially as described above under the heading “Neutrophil-mediated Opsonophagocytic Bacteriacidal Assay”.
  • M120 was purified from ascites essentially as described by the manufacturer of MEP Hypercel gel (BiSepra). Thirty-three ml of buffer A (50 mM Tris, 5 mM EDTA, pH 8) was added to 17 ml of mouse ascites, and then centrifuged for 15 minutes at 4000 rpm in an Eppendorf model 5810R centrifuge using rotor A462. The solution was filtered using Whatman GD/XP PES 0.45 ⁇ membrane (cat. no.
  • the volume of diluted ascites was 47 ml after filtering.
  • the solution was loaded onto a 1 cm ⁇ 7 cm MEP hypercel column that had been equilibrated with buffer A, at a rate of 1.8 ml/min.
  • the column was washed with buffer A, and then with buffer A+25 mM sodium caprylate until the OD280 was ⁇ 0.05.
  • the column was then washed with water until the OD 280 ⁇ 0.05.
  • the column was eluted with buffer B (50 mM sodium acetate, 5 mM EDTA, pH 4) and the eluent collected at a rate of 70 drops/min.
  • pool A contained 3.2 mg/ml antibody
  • pool B contained 0.25 mg/ml antibody.
  • MAb-391.4 which was raised against UV-killed S. aureus , has strong opsonic activity against S. epidermidis strain Hay.
  • FIG. 3 shows the strategy for cloning the variable region genes.
  • the total RNA (2 ⁇ g) was converted to cDNA by using Superscript II-MMLV Reverse Transcriptase (Life Technologies) and mouse Kappa chain-specific primer (JSBX-18; SEQ ID NO: 5) and a mouse heavy chain-specific primer (JSBX-25A; SEQ ID NO: 6) according to the manufacturer's procedures (see FIG. 4 for primer sequences).
  • the first strand cDNA synthesis products were then purified using a Centricon-30 concentrator device (Amicon). Of the 40 ⁇ l of cDNA recovered, 5 ⁇ l was used as template DNA for PCR.
  • Typical PCR amplification reactions contained template DNA, 30 pmoles of the appropriate primers (JSBX-9A, 11 A, and 18 for light chains, SEQ ID NOs: 3-5; JSBX-1, 4 and JSBX-25A for heavy chains, SEQ ID NOs: 1, 2, and 6), 2.5 units of ExTaq polymerase (PanVera), 1 ⁇ ExTaq reaction buffer, 200 ⁇ M dNTP, 2 mM MgC12.
  • the template was denatured by an initial incubation at 96° C. for 3 min.
  • the products were amplified by 30 thermal cycles of 96° C. for 1 min., 60° C. for 30 sec, 72° C. for 30 seconds.
  • the PCR products from the successful reactions were purified using the Nucleospin PCR Purification system (Clontech) as per manufacturer's procedure.
  • PCR products (approximately 400 base pairs each) were then cloned into a bacterial vector, pGEM T (Promega) for DNA sequence determination.
  • PCR fragments were ligated into pGEM T, a T/A style cloning vector, following the manufacturer's procedures using a 3:1 insert to vector molar ratio.
  • One half (5 ⁇ l) of the ligation reactions were used to transform Ultracompetent XL1 Blue cells (Stratagene) as per the manufacturer's procedure.
  • Bacterial clones containing plasmids with DNA inserts were identified using diagnostic restriction enzyme digestions with Dralll and BsiWI (for heavy chain clones) or Dralll and EcoRV (for light chain clones) (New England Biolabs). The DNA sequences of plasmids containing inserts of the appropriate size ( ⁇ 400 bp) were then determined. The plasmid containing the A120 heavy chain sequence was designated pJSB16-6 and the plasmid containing the A120 light chain variable region was designated pJSB17-23. The final consensus DNA sequences of the light chain and heavy chain variable regions are shown in FIG. 5 and FIG. 6 , respectively.
  • the heavy and light chain variable regions were then subcloned into a mammalian expression plasmid vector for production of recombinant chimeric mouse/human antibody molecules.
  • the human/mouse chimera of the M120 antibody is designated A120
  • the human/mouse chimera of the M110 antibody is designated A110 (see U.S. Pat. No. 6,610,293, filed Jun. 15, 1998).
  • vectors were designed that express recombinant antibody molecules under the control of CMV transcriptional promoters.
  • the chimeric heavy chains are expressed as a fusion of a heavy chain variable region and a human IgG1 constant domain.
  • the chimeric light chains are expressed as a fusion of a light chain variable region and a human kappa chain constant region.
  • the chimeric light chain cDNA contains a mouse kappa intron between the variable region and the human kappa constant region. After splicing, the variable region becomes fused to a human Kappa constant region exon.
  • the selectable marker for the vector in mammalian cells is Neomycin resistance (resistance to G418).
  • variable region gene fragments of M120 were re-amplified by PCR using primers that adapted the fragments for cloning into the expression vector ⁇ see FIG. 4 , JSBX-46 through JSBX-49, SEQ ID NOs: 7-9 and 18).
  • the heavy chain front primer (JSBX-46; SEQ ID NO: 7) includes a 5′ tail that encodes the C-terminus of the heavy chain leader and a Ss/WI restriction site for cloning, while the heavy chain reverse primer (JSBX-47; SEQ ID NO: 8) adds a 3′ EcoRI restriction site for cloning.
  • the light chain front primer (JSBX-48; SEQ ID NO: 9) introduces a 5′ tail that encodes the two C-terminal amino acids of the light chain leader and an Age/restriction site for cloning purposes.
  • the light chain reverse primer (JSBX-49; SEQ ID NO: 18) adds a 3′ DNA sequence for the joining region-Kappa exon splice junction followed by a BstBI restriction site for cloning.
  • variable regions were re-amplified from the plasmid DNA using vector pJSB16-6 for the heavy chain variable region and vector pJSB17-23 for the light chain variable region.
  • PCR reactions were performed as described above. Following a 3 minute incubation at 96° C., the PCR parameters were 30 thermal cycles of 58° C. for 30 seconds, 70° C. for 30 seconds, and 96° C. for 1 minute.
  • the heavy chain variable region PCR product was digested with BsiWI and EcoRI (New England Biolabs), purified using a Nucleospin PCR Purification column (Clontech), as described by the manufacturer, and ligated into BsiWI/EcoRI/PfIMI-digested and gel-purified pJRS383 vector using the Takara Ligation Kit (Panvera) following the manufacturer's procedure.
  • the ligation mix was then transformed into XL1Blue cells (Stratagene), resulting in plasmid mammalian expression vector pJSB23-1 ( FIG. 7 ).
  • the light chain variable region PCR product (approximately 350 bp) was digested with AgeI and BstBI (New England Biolabs), and purified using a Nucleospin PCR Purification column (Clontech) as described by the manufacturer.
  • the light chain variable region fragment was ligated into pJRS384 that had been AgeI/BstBI/Xcml-digested and gel-purified using the Takara Ligation Kit (Panvera) following the manufacturer's procedure.
  • the ligation mix was transformed into XL1Blue cells (Stratagene), resulting in mammalian expression plasmid pJSB24 ( FIG. 8 ).
  • mammalian cell expression plasmids that contained the A120 heavy and light chain variable regions in a bi-cistronic plasmid, as well as plasmids that combined the A110 heavy chain variable region with the A120 light chain variable region, and the A110 light chain variable region with the A120 heavy chain variable region, in order to investigate the binding and opsonic properties of the different antibodies.
  • Construction of the bi-cistronic vectors was done in a step-wise fashion, in which the heavy and light chain variable regions of A120 were cloned into a bi-cistonic expression plasmid already containing the A110 light and heavy chain variable regions (pJRS354, FIG.
  • the plasmid pJRS354 was digested with ClaI and XhoI (New England Biolabs), the digestion products were separated on an agarose gel and the backbone fragment was cut out and gel purified using a Nucleospin Gel Fragment DNA Purification column (Clontech), as described by the manufacturer.
  • the plasmid pJSB24 was digested with ClaI and XhoI (New England Biolabs), the digestion products were separated on an agarose gel and the light chain variable region fragment was cut out and gel purified using a Nucleospin Gel Fragment DNA Purification column (Clontech), as described by the manufacturer.
  • the two other bi-cistronic plasmids were constructed in a similar manner.
  • the plasmid pJSB25-3 was digested with BspEI and NotI (New England Biolabs), the digestion products were separated on an agarose gel and the backbone fragment was cut out and gel purified using a Nucleospin Gel Fragment DNA Purification column (Clontech), as described by the manufacturer.
  • the plasmid pJSB23-1 was digested with BspEl and NotI (New England Biolabs), the digestion products were separated on an agarose gel and the heavy chain variable region fragment was cut out and gel purified using a Nucleospin Gel Fragment DNA Purification column (Clontech), as described by the manufacturer.
  • the plasmid pJRS354 was digested with BspEl and NotI (New England Biolabs), the digestion products were separated on an agarose gel and the backbone fragment was cut out and gel purified using a Nucleospin Gel Fragment DNA Purification column (Clontech), as described by the manufacturer.
  • the plasmid pJSB23-1 was digested with BspEl and NotI (New England Biolabs), the digestion products were separated on an agarose gel and the heavy chain variable region fragment was cut out and gel purified using a Nucleospin Gel Fragment DNA Purification column (Clontech), as described by the manufacturer.
  • Anti-LTA human/mouse chimeric antibody A110 was previously described in U.S. patent application Ser. No. 09/097,055, now U.S. Pat. No. 6,610,293, which is herein incorporated by reference.
  • the binding activities of anti-LTA human/mouse chimeric antibodies A110 and A120 were compared in ELISA assays against LTA.
  • Dilutions of A120 supernatant were compared to dilutions of purified A110 antibody in an immunoassay as described above under the heading “Binding Assays”, subheading “Immunoassay on LTA”. Briefly, the wells of a 96-well plate were coated with 1 ⁇ g/ml of S. aureus LTA for three hours at room temperature. After washing, dilutions of purified A110 antibody or A120 supernatant in PBS-T were added to quadruplicate wells and incubated for 30 to 60 minutes at room temperature.
  • Purified human/mouse chimeric A110 and A120, and mouse M120 MAbs were assayed for their opsonic activity as described above, under the heading “Neutrophil-Mediated Opsonophagocytic Bactericidal Assay.” Briefly, dilutions of purified A110, A120, or M120 antibodies were combined with neutrophils (PMNs) in the wells of a microtiter plate. Mid-log phase bacteria were added to each well, followed by immunoglobulin-depleted human serum, which serves as a source of complement (C). Samples were incubated for 2 hours at 37° C., and were then plated on blood agar and incubated overnight to determine the number of live bacteria remaining.
  • PMNs neutrophils
  • C immunoglobulin-depleted human serum
  • the plasmids pJSB25, pJSB26 and pJSB27 were transected into COS cells grown in IMDM plus 10% fetal bovine serum, using Superfect (Qiagen) in 6 well tissue culture wells as described by the manufacturer. After two days the supernatant was assayed for the production of chimeric antibody and for the capability for the expressed antibody to bind to S. aureus LTA antigen.
  • Antibody production assays were preformed in 8-well strips from 96-well microtiter plates (Maxisorp F8; Nunc, Inc.) coated at a 1:500 dilution with a goat antihuman Fc (Pierce). The plates are covered with pressure sensitive film and incubated overnight at 4° C. Plates were then washed once with Wash solution (Imidazole/NaCl/0.4% Tween-20). One hundred microliters of culture supernatant dilutions were then applied to duplicate wells and allowed to incubate for 60 minutes on a plate rotator at room temperature. The plates were washed seven times with Wash solution.
  • a Goat anti-Human IgG H+L-HRP (Zymed) conjugate was diluted 1:4000 in the sample/conjugate diluent.
  • One hundred microliters was added to the samples, and then incubated on a plate rotator for 60 minutes at room temperature.
  • the samples were washed as above and then incubated with 100 (xL/well of TMB developing substrate (BioFx) for 1 minute at room temperature.
  • the binding reaction was stopped with 100 ⁇ Uwell of Quench buffer (BioFx) and the absorbance value at 450 nm was determined using an automated microtiter plate ELISA reader.
  • This assay (see FIG. 15 ) demonstrates that the transfection of cells with this plasmid construct results in the cells producing a molecule containing both human IgG and Kappa domains.
  • the supernatants were then assayed for the ability of the expressed antibodies to bind to lipoteichoic acid.
  • the activity assays were preformed in 8-well strips from 96-well microtiter plates (Maxisorp F8; Nunc, Inc.) coated at 1 ⁇ g/mL with S. aureus LTA (Sigma) using PBS. The plates were covered and incubated overnight at 4° C. Plates were then washed once with PBS. One hundred microliters of culture supernatant dilutions were then applied to duplicate wells and allowed to incubate for 60 minutes on a plate rotator at room temperature. The plates were washed seven times with Wash solution.
  • Goat anti-Human IgG H+L-HRP (Zymed) was diluted 1:4000 in the sample/conjugate diluent, and 100 ul were added to the samples, and then incubated on a plate rotator for 60 minutes at room temperature. The samples were washed as above and then incubated with 100 ⁇ L/well of TMB developing substrate (BioFx) for 10-15 minutes on a plate rotator at room temperature. The binding reaction was stopped with 100 ⁇ L/well of Quench buffer (BioFx) and the absorbance value at 450 nm was determined using an automated microtiter plate ELISA reader.
  • TMB developing substrate BioFx
  • the binding reaction was stopped with 100 ⁇ L/well of Quench buffer (BioFx) and the absorbance value at 450 nm was determined using an automated microtiter plate ELISA reader.
  • a skilled artisan can isolate a protective anti-LTA antibody that is fully human.
  • a protective anti-LTA antibody that is fully human.
  • human RNA is used to produce a cDNA library of antibody heavy and light chain fragments expressed on the surface of bacteriophage. These libraries can be used to probe against the antigen of interest (i.e., LTA) and the phage that bind, because of the antibody expressed on the surface, are then isolated.
  • the DNA encoding the variable regions is sequenced and cloned for antibody expression.
  • mice Another method of producing human antibodies employs “humanized” mice. These transgenic mice have had their own antibody genes replaced with a portion of the human antibody gene complex so that upon inoculation with antigen, they produce human antibodies (57, 59, 60, 61, 63). The antibody producing cells that result can then be incorporated into the standard hybridoma technology for the establishment of specific monoclonal antibody producing cell lines.
  • Recombinant human antibodies are also produced by isolating antibody-producing B cells from human volunteers that have a robust anti-LTA response. Using fluorescence activated cell sorting (FACS) and fluorescently labeled LTA, cells producing the anti-LTA antibodies can be separated from the other cells. The RNA can then be extracted and the sequence of the reactive antibody variable regions determined (58, 62). The DNA sequence of the functional variable regions can be synthesized or cloned into mammalian expression vectors for large-scale human recombinant antibody production.
  • FACS fluorescence activated cell sorting
  • LTA fluorescently labeled LTA
  • Hybridoma subclone 00-107GG12 ID12 produced an IgG 2a monoclonal antibody with a kappa light chain that bound strongly to LTA.
  • the antibody produced by this hybridoma was designated M120 (Example 1).
  • M120 was tested in an opsonophagocytic bacteriocidal assay for opsonic activity against S. aureus type 5 and S. epidermidis strain Hay.
  • the antibody was mixed with PMNs and complement, which was derived from human serum that had been depleted of anti- S. aureus and anti- S. epidermidis antibodies, and then tested for activity against the bacteria.
  • M120 showed opsonic activity against both S. aureus and S. epidermidis , killing 95% of S. epidermidis and an average of 82% of S. aureus at 200 ⁇ g/ml (Example 2, Tables 5 and 6).
  • MAb-391.4 which was raised to UV-killed S. aureus , was tested for opsonic activity against S. epidermidis strain Hay in a similar assay, and showed 81.9% killing (Table 7).
  • M120 variable regions were then cloned and sequenced, and the sequence compared to another anti-LTA antibody, M110.
  • M110 and M120 were found to share about 94% sequence identity at the amino acid level, and about 96% sequence identity at the nucleotide level.
  • a third anti-LTA antibody, MAb-391.4 was also sequenced compared to the other two. The three antibodies share 88% sequence identity at the amino acid level. This high level of sequence identity may suggest that the antibodies bind to a common epitope on LTA (Example 3, FIGS. 9 and 10 ).
  • Human/mouse chimeric antibodies were then made, fusing the heavy chain variable region of either M120 or M110 to a human IgG1 constant region, and the light chain variable region of either M120 or M110 to a human kappa light chain constant region.
  • the human/mouse chimera of M120 is referred to as A120 and the human/mouse chimera of M110 is referred to as A110.
  • A120a an antibody that contained the heavy chain of A110 and the light chain of A120
  • A120b an antibody that contained the light chain of A110 and the heavy chain of A120
  • the human/mouse chimeric antibodies A120 and A110 were tested for their ability to bind to LTA in an ELISA assay. Both chimeric antibodies bound strongly to LTA, indicating that replacing the mouse constant regions with human constant regions had little effect on the binding properties of the antibodies (Example 5, Tables 8 and 9).
  • the opsonic activity of chimeric antibodies A110 and A120, and of M120 were compared in an opsonic assay against S. epidermidis strain Hay. All three antibodies showed at least 94% killing of S. epidermidis .
  • These results show that the chimeric antibodies are strongly opsonic against S. epidermidis , and because they have a reduced HAMA response in humans, they should be suitable therapeutic molecules for fighting Gram-positive bacterial infections (Example 6, Table 10).
  • the level of homology between the M110, M120, and MAb-391.4 variable regions may indicate that opsonic antibodies to LTA recognize a nearly identical epitope using nearly identical modes of binding, and that this mode of binding is important to their functional activity. Furthermore, the epitope to which the antibodies bind appears to be highly conserved between S. epidermidis and S. aureus , and may be common to most, if not all, Gram-positive bacteria. Monoclonal antibodies to this epitope may, therefore, be broadly opsonic against a wide range of bacteria, allowing researchers to develop a few antibodies that will have broad opsonic and protective activity against many Gram-positive bacteria.

Abstract

The present invention encompasses monoclonal antibodies that bind to lipoteichoic acid (LTA) of Gram positive bacteria. The antibodies also bind to whole bacteria and enhance phagocytosis and killing of the bacteria in vitro. The invention also provides antibodies having human sequences (chimeric, humanized and human antibodies). The invention also sets forth the variable regions of three antibodies within the invention and presents the striking homology between them.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. patent application Ser. No. 11/724,040, which is a divisional of U.S. patent Ser. No. 10/323,927, filed Dec. 20, 2002, now U.S. Pat. No. 7,250,494, which is a continuation-in-part of U.S. patent application Ser. No. 09/097,055, filed Jun. 15, 1998, now U.S. Pat. No. 6,610,293, and is further based on and claims the benefit of U.S. Provisional Application Ser. No. 60/343,503, filed Dec. 21, 2001, now expired.
  • This application also relates to U.S. Pat. No. 5,571,511, U.S. Pat. No. 5,955,074, and U.S. Pat. No. 6,610,293. Each of the above-referenced applications is incorporated herein by reference for all purposes.
  • DESCRIPTION OF THE INVENTION
  • 1. Field of the Invention
  • This invention in the fields of immunology and infectious diseases relates to antibodies that are specific for Gram positive bacteria, particularly to bacteria that bear lipoteichoic acids on their surfaces. The invention includes monoclonal and chimeric antibodies, as well as fragments, regions and derivatives thereof. This invention further relates to sequences of the variable region that enhance the antibody's opsonic activity. The antibodies of the invention may be used for diagnostic, prophylactic and therapeutic applications.
  • 2. Background of the Invention
  • The search for agents to combat bacterial infections has been long and arduous. The development of antibiotics has brought us from the time when sepsis associated with amputation was associated with a 50 percent mortality rate. Today's challenge, however, is the increasing development of bacteria that are resistant to antibiotics, such as members of the genera Staphylococcus.
  • Staphylococci are particularly worrisome because they commonly colonize humans and animals and are an important cause of human morbidity and mortality. Because of their prevalence on the skin and mucosal linings, staphylococci are ideally situated to produce both localized and systemic infections. Of the staphylococci, both S. aureus, a coagulase positive bacteria, and S. epidermidis, a coagulase negative species, are the most problematic. In fact, S. aureus is the most virulent Staphylococcus, producing severe and often fatal disease in both normal and immunocompromised hosts. S. epidermidis has become one of the major causes of nosocomial (hospital acquired) infection in patients with impaired immune responses or those whose treatments involve the placement of foreign objects into the body, such as patients who receive continuous ambulatory peritoneal dialysis and patients receiving parenteral nutrition through central venous catheters (25). Indeed, S. epidermidis is now recognized as a common cause of neonatal nosocomial sepsis, and infections frequently occur in premature infants that have received parenteral nutrition. Moreover, in recent years, the involvement of S. epidermidis in neonatal infection has increased dramatically. Indeed, for every 10 babies diagnosed with bacterial sepsis seven or more days after birth (indicative of post-partum bacterial exposure), six of those are infected with S. epidermidis. Untreated, Staphylococcus infections in newborns can result in multiple organ failure and death in two to three days. Antibiotics are only partially effective and, unfortunately, the rise in multiply drug resistant strains of Staphylococcus renders antibiotic treatments less and less effective.
  • The problems of antibiotic resistance are so significant that they have reached the lay press. See, e.g., The Washington Post “Microbe in Hospital Infections Show Resistance to Antibiotics,” May 29, 1997; The Washington Times, “Deadly bacteria outwits antibiotics,” May 29, 1997. And this concern is borne out by the scientific literature. See L. Garrett, The Coming Plague, “The Revenge of the Germs or Just Keep Inventing New Drugs” Ch. 13, pgs. 411-456, Farrar, Straus and Giroux, N.Y., Eds. (1994). In one study, the majority of staphylococci isolated from blood cultures of septic infants were resistant to multiple antibiotics (10). Another study describes methicillin-resistant S. aureus (31). There is no doubt that the emergence of antibiotic resistance among clinical isolates is making treatment difficult (18).
  • The other possible route of treatment is the administration of antibodies. Antibodies protect against bacterial attack by recognizing and binding to antigens on the bacteria to thereby facilitate the removal or “clearance” of the bacteria by a process called phagocytosis, wherein phagocytic cells (predominantly neutrophils and macrophages) identify, engulf, and subsequently destroy the invading bacteria. However, bacteria have developed mechanisms to avoid phagocytosis, such as the production of a “capsule” to which phagocytes cannot adhere or the production of toxins that actually poison the encroaching phagocytes. Antibodies overcome these defenses by, for example, binding to the toxins to thereby neutralize them. More significantly, antibodies may themselves bind to the capsule to coat it, in a process called opsonization, to make the bacteria extremely attractive to phagocytes and to enhance their rate of clearance from the bloodstream.
  • Confounding the use of administered antibodies, however, are conflicting reports in the literature. For example, the immunization studies of Fattom et al. demonstrated that opsonization of S. epidermidis was related to the specific capsule type, as with S. aureus and other encapsulated Gram positive bacteria such as Streptococcus pneumonia (6). In another study, Timmerman et al. identified a surface protein of S. epidermidis that induced opsonic monoclonal antibodies (39). Timmerman et al. also identified other monoclonal antibodies that bound to non-homologous S. epidermidis strains, but only the monoclonal antibody produced to the homologous strain was opsonic, thus opsonization was enhanced only to the homologous strain but not to heterologous strains. Accordingly, based on the studies of Fattom et al., and Timmerman et al., and others in the field (and in contrast to our own studies as set forth in U.S. Pat. Nos. 5,571,511 and 5,955,074), one would not expect that an antibody that is broadly reactive to multiple strains of S. epidermidis and to S. aureus would have opsonic activity against each strain. This is particularly true for antibodies that bind to both coagulase positive and coagulase negative staphylococci.
  • Further exacerbating the problem, the role of the common surface antigens on staphylococci has been unclear. For example, while lipoteichoic acid and teichoic acid make up the majority of the cell wall of S. aureus, there was no prior appreciation that antibodies to lipoteichoic acid and teichoic acid could be protective. Indeed, anti-teichoic acid antibodies have been often used as controls. For example, Fattom et al. examined the opsonic activity of antibodies induced against a type-specific capsular polysaccharide of S. epidermidis, using as controls antibodies induced against teichoic acids and against S. hominus. While type-specific antibodies were highly opsonic, anti-teichoic acid antibodies were not functionally different from the anti-S. hominus antibodies (6).
  • Similarly, in Kojima et al., the authors assessed the protective effects of antibody to capsular polysaccharide/adhesion against catheter-related bacteremia due to coagulase negative staphylococci and specifically used a strain of S. epidermidis that expresses teichoic acid as a control ((16); see page 436, Materials and Methods, left column, first paragraph; right column, third paragraph). In a later study, Takeda et al. (38), the authors reached a more explicit conclusion against the utility of anti-techoic antibodies:
      • Immunization protocols designed to elicit antibody to techoic acid but not to PS/A afforded no protection against bacteremia or endocarditis (38).
  • Thus, the role of antibodies in the protection against infections by Gram positive bacteria, particularly Staphylococci such as S. aureus and S. epidermidis, has not been clear, and there is a need in the art for monoclonal antibodies to both protect against such bacterial infection and to help elucidate the role of such antibodies against such infection. There is also a need in the art for sequence analysis of such antibodies so that antibodies of enhanced binding and opsonic activity can be identified and/or produced.
  • SUMMARY OF THE INVENTION
  • The present invention encompasses broadly reactive, opsonic, and protective monoclonal and chimeric antibodies that bind to lipoteichoic acid (LTA) of Gram positive bacteria. The antibodies also bind to whole bacteria and enhance phagocytosis and killing of the bacteria in vitro and enhance protection from lethal infection in vivo. The present invention further encompasses opsonic antibodies to LTA that share a high degree of sequence homology. The present invention also encompasses antibodies having variable regions derived from two or more different anti-LTA antibodies.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 provides a schematic representation of lipoteichoic acid (LTA) in the Gram positive bacterial cell wall.
  • FIG. 2 depicts antibody regions, including the heavy chain constant region (CH), the heavy chain variable region (VH), the light chain constant region (CL), and the light chain variable region (VL). The complementarity determining regions (CDRs) within the variable regions are shown as black bars.
  • FIG. 3 shows the cDNA cloning strategy for the heavy and light chain variable regions of A120.
  • FIG. 4 shows the oligonucleotide primers used to amplify the variable region fragments. (SEQ ID NOs: 1-9 and 18)
  • FIG. 5 shows the amino acid sequence (SEQ ID NO: 10) and the polynucleotide sequence (SEQ ID NO: 11) of the A120 light chain variable region.
  • FIG. 6 shows the amino acid sequence (SEQ ID NO: 12) and the polynucleotide sequence (SEQ ID NO: 13) of the A120 heavy chain variable region.
  • FIG. 7 depicts the pJSB23-1 plasmid that expresses the A120 heavy chain.
  • FIG. 8 depicts the pJSB24 plasmid that expresses the A120 light chain.
  • FIG. 9 shows an alignment of (A) the A110 light chain variable region cDNA (SEQ ID NO: 14), the A120 light chain variable region cDNA (SEQ ID NO: 11) and the 391.4 light chain variable region cDNA (SEQ ID NO: 19) and (B) the A110 heavy chain variable region cDNA (SEQ ID NO: 15), the A120 heavy chain variable region cDNA (SEQ ID NO: 13) and the 391.4 heavy chain variable region cDNA (SEQ ID NO: 20). The nucleotides that differ between any two sequences are boxed.
  • FIG. 10A shows an alignment of the A110 light chain variable region polypeptide sequence (SEQ ID NO: 16), the A120 light chain variable region polypeptide sequence (SEQ ID NO: 10) and the 391.4 light chain variable region polypeptide sequence (SEQ ID NO: 21). FIG. 10B shows an alignment of the A110 heavy chain variable region polypeptide sequence (SEQ ID NO: 17), the A120 heavy chain variable region polypeptide sequence (SEQ ID NO: 12) and the 391.4 heavy chain variable region polypeptide sequence (SEQ ID NO: 22). The complementarity determining regions (CDRs) are underlined and the amino acids that differ between any two sequences are boxed.
  • FIG. 11 depicts the pJRS354 bi-cistronic plasmid that expresses the A110 heavy chain and light chain variable regions.
  • FIG. 12 depicts the pJSB25-3 bi-cistronic plasmid that expresses the A110 heavy chain variable region and the A120 light chain variable region.
  • FIG. 13 depicts the pJSB26 bi-cistronic plasmid that expresses the A120 heavy chain and light chain variable regions.
  • FIG. 14 depicts the pJSB27 bi-cistronic plasmid that expresses the A120 heavy chain variable region and the A110 light chain variable region.
  • FIG. 15 provides the results of the chimeric antibody production ELISA. All antibodies shown are human/mouse chimeras. A110 contains both the heavy and light chain variable regions from A110. A120 contains both the heavy and light chain variable regions from A120. A120a contains the heavy chain variable region from A110 and the light chain variable region from A120. A120b contains the heavy chain variable region from A120 and the light chain variable region from A110.
  • FIG. 16 provides the results of the experiment to determine chimeric antibody binding to the S. aureus LTA. The antibodies used are the same as in FIG. 15.
  • DETAILED DESCRIPTION OF THE INVENTION Definitions
  • The term “antibody”, as used herein, includes full-length antibodies and portions thereof. A full-length antibody has one pair or, more commonly, two pairs of polypeptide chains, each pair comprising a light and a heavy chain. Each heavy or light chain is divided into two regions, the variable region (which confers antigen recognition and binding) and the constant region (associated with localization and cellular interactions). Thus, a full-length antibody commonly contains two heavy chain constant regions (Hc, or CH), two heavy chain variable regions (Hv or VH), two light chain constant regions (Lc or Cl_), and two light chain variable regions (Lv or VL) (FIG. 2). The light chains or chain, may be either a lambda or a kappa chain. Thus, in an embodiment of the invention, the antibodies include at least one heavy chain variable region and one light chain variable region, such that the antibody binds antigen.
  • Another aspect of the invention involves the variable region that comprises alternating complementarity determining regions, or CDRs, and framework regions, or FRs. The CDRs are the sequences within the variable region that generally confer antigen specificity.
  • The invention also encompasses portions of antibodies that comprise sufficient variable region sequence to confer antigen binding. Portions of antibodies include, but are not limited to Fab, Fab′, F(ab′)2, Fv, SFv, scFv (single-chain Fv), whether produced by proteolytic cleavage of intact antibodies, such as papain or pepsin cleavage, or by recombinant methods, in which the cDNAs for the intact heavy and light chains are manipulated to produce fragments of the heavy and light chains, either separately, or as part of the same polypeptide.
  • MAbs within the scope of the invention include sequences corresponding to human antibodies, animal antibodies, and combinations thereof. The term “chimeric antibody,” as used herein, includes antibodies that have variable regions derived from an animal antibody, such as a rat or mouse antibody, fused to another molecule, for example, the constant domains derived from a human antibody. One type of chimeric antibodies, “humanized antibodies”, have had the variable regions altered (through mutagenesis or CDR grafting) to match (as much as possible) the known sequence of human variable regions. CDR grafting involves grafting the CDRs from an antibody with desired specificity onto the FRs of a human antibody, thereby replacing much of the non-human sequence with human sequence. Humanized antibodies, therefore, more closely match (in amino acid sequence) the sequence of known human antibodies. By humanizing mouse monoclonal antibodies, the severity of the human anti-mouse antibody, or HAMA, response is diminished. The invention further includes fully human antibodies which would avoid, as much a possible, the HAMA response.
  • Modified antibodies include, for example, the proteins or peptides encoded by truncated or modified antibody-encoding genes. Such proteins or peptides may function similarly to the antibodies of the invention. Other modifications, such as the addition of other sequences that may enhance the effector function, which includes the ability to block or alleviate nasal colonization by staphylococci, are also within the present invention. Such modifications include, for example, the addition of amino acids to the antibody's amino acid sequence, deletion of amino acids in the antibody's amino acid sequence, substitution of one or more amino acids in the antibody amino acid sequence with alternate amino acids, isotype switching, and class switching.
  • In certain embodiments, an antibody may be modified in its Fc region to prevent binding to bacterial proteins. The Fc region normally provides binding sites for neutrophils, macrophages, other accessory cells, complement components, and, receptors of the immune system. As the antibodies bind to bacteria and opsonize them, accessory cells recognize the coated bacteria and respond to infection. When a bacterial protein binds to the Fc region near the places where accessory cells bind, the normal function of these cells is inhibited. For example, Protein A, a bacterial protein found in the cell membrane of S. aureus, binds to the Fc region of IgG near accessory cell binding sites. In doing so, Protein A inhibits the function of these accessory cells, thus interfering with clearance of the bacterium. To circumvent this interference with the antibacterial immune response, the Fc portion of the antibody of the invention may be modified to prevent nonspecific binding of Protein A while retaining binding to accessory cells (15).
  • In light of these various forms, the antibodies of the invention include clones of full length antibodies, antibody portions, chimeric antibodies, humanized antibodies, fully human antibodies, and modified antibodies. Collectively, these will be referred to as “MAbs” or monoclonal antibodies unless otherwise indicated.
  • The term “epitope”, as used herein, refers to a region, or regions, of LTA that is bound by an antibody to LTA. The regions that are bound may or may not represent a contiguous portion of the molecule.
  • The term “antigen”, as used herein, refers to a polypeptide sequence, a non-proteinaceous molecule, or any molecule that can be recognized by the immune system. An antigen may be a full-sized staphylococcal protein or molecule, or a fragment thereof, wherein the fragment is either produced from a recombinant cDNA encoding less than the full-length protein or derived from the full-sized molecule or protein. Such fragments may be produced via enzymatic processing, such as proteolysis. An antigen may also be a polypeptide sequence that encompasses an epitope of a staphylococcal protein, wherein the epitope may not be contiguous with the linear polypeptide sequence of the protein. The DNA sequence encoding an antigen may be identified, isolated, cloned, and transferred to a prokaryotic or eukaryotic cell for expression by procedures well-known in the art (25).
  • An antigen, or epitope thereof, may be 100% identical to a region of the staphylococcal molecule or protein amino acid sequence, or it may be at least 95% identical, or at least 90% identical, or at least 85% identical. An antigen may also have less than 95%, 90% or 85% identity with the staphylococcal molecule or protein amino acid sequence, provided that it still be able to elicit antibodies the bind to a native staphylococcal molecule or protein. The percent identity of a peptide antigen can be determined, for example, by comparing the sequence of the target antigen or epitope to the analogous portion of staphylococcal sequence using the GAP computer program, version 6.0 described by Devereux et al. (Nucl. Acids Res. 12:387, 1984) and available from the University of Wisconsin Genetics Computer Group (UWGCG). The GAP program utilizes the alignment method of Needleman and Wunsch (J. Mol. Biol. 48:443, 1970), as revised by Smith and Waterman (Adv. Appl. Math 2:482, 1981), and is applicable to determining the percent identity of protein or nucleotide sequences referenced herein. The preferred default parameters for the GAP program include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non-identities) for nucleotides, and the weighted comparison matrix of Gribskov and Burgess, Nucl. Acids Res. 14:6745, 1986, as described by Schwartz and Dayhoff, eds., Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, pp. 353-358, 1979; (2) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap; and (3) no penalty for end gaps.
  • Alternatively, for simple comparisons over short regions up to 10 or 20 units, or regions of relatively high homology, for example between antibody sequences, the percent identity over a defined region of peptide or nucleotide sequence may by determined by dividing the number of matching amino acids or nucleotides by the total length of the aligned sequences, multiplied by 100%. Where an insertion or gap of one, two, or three amino acids occurs in a MAb chain, for example in or abutting a CDR, the insertion or gap is counted as single amino acid mismatch.
  • Antigens may be surface antigens and/or virulence antigens and/or adherance antigens. Surface antigens are antigens that are accessible to an antibody when the antigen is in the configuration of the whole intact bacterium, i.e., the antigen is not inside the cell cytoplasm. Virulence antigens are antigens that are involved in the pathogenic process, causing disease in a host. Virulence antigens include, for example, LTA, peptidoglycan, toxins, fimbria, flagella, and adherence antigens. Adherence antigens mediate the ability of a staphylococcal bacterium to adhere to an epithelial surface, such as the epithelial surface of the anterior nares. An antigen may be a non-proteinaceous component of staphylococci such as a carbohydrate or lipid. For example, peptidoglycan and lipoteichoic acid are two non-proteinaceous antigens found in the cell wall of staphylococci. Antigens may comprise or include fragments of non-proteinaceous molecules as long as they elicit an immune response.
  • As used herein, antigens include molecules that can elicit an antibody response to LTA. An antigen may be LTA itself, or a fragment or portion thereof. An antigen may also be an unrelated molecule, which, through some structural similarity, is able to elicit antibodies that bind to LTA. Binding to LTA may thus be assessed by binding to such peptide epitope mimics, as described, for example, in U.S. Patent Application Publication No. 2002/0082395, which is incorporated herein by reference. In certain embodiments of the invention, an antigen elicits antibodies that bind to LTA on the surface of bacteria.
  • As specifically used herein, an antigen is any molecule that can specifically bind to an antibody, including antibodies specific for LTA. Antigens of the invention thus include antigens that bind to any of monoclonal antibodies MAb-391.4, M110, M120, A110, A120, A120a, and A120b, described herein.
  • An antibody is said to specifically bind to an antigen, epitope, or protein, if the antibody gives a signal by an assay such as an ELISA assay that is at least two fold, at least three fold, at least five fold, or at least ten fold greater than the background signal, i.e., at least two fold, at least three fold, at least five fold, or at least ten fold greater than the signal ascribed to non-specific binding. An antibody is said to specifically bind to a bacterium if the antibody gives a signal by MeOH-fixed bacteria ELISA or live bacteria ELISA, or other assay, that is at least 1.5 fold, 2 fold, or 3 fold greater than the background signal.
  • “Enhanced phagocytosis”, as used herein, means an increase in phagocytosis over a background level as assayed by the methods in this application, or another comparable assay. The level deemed valuable may well vary depending on the specific circumstances of the infection, including the type of bacteria and the severity of the infection. For example, for enhanced phagocytic activity, in one embodiment, an enhanced response is equal to or greater than 75% over background phagocytosis. In another embodiment, an enhanced response is equal to or greater than 80% or 85% over background phagocytosis. In another embodiment, an enhanced response is equal to or greater than 90% or 95% over background phagocytosis. Enhanced phagocytosis may also be equal to or greater than 50%, 55%, 60%, 65%, or 70% over background phagocytosis. In another embodiment, enhanced phagocytosis comprises a statistically significant increase in phagocytic activity as compared to background phagocytosis or phagocytosis with a non-specific or non-opsonic control antibody.
  • The specific determination or identification of a “statistically significant” result will depend on the exact statistical test used. One of ordinary skill in the art can readily recognize a statistically significant result in the context of any statistical test employed, as determined by the parameters of the test itself. Examples of these well-known statistical tests include, but are not limited to, X2 Test (Chi-Squared Test), Students t Test, F Test, M test, Fisher Exact Text, Binomial Exact Test, Poisson Exact Test one way or two way repeated measures analysis of variance, and calculation of correlation efficient (Pearson and Spearman).
  • A MAb has “opsonic activity” if it can bind to an antigen to promote attachment of the antigen to the phagocyte and thereby enhance phagocytosis. As used herein, opsonic activity may also be assessed by assays that measure neutrophil mediated opsonophagocytotic bactericidal activity.
  • The MAb's of the invention are useful for the treatment of systemic and local staphylococcal infections. As used herein, “treatment” encompasses any reduction, amelioration, or “alleviation” of existing infection as well as “blocking” or prophylaxis against future infection. In this respect, treatment with a MAb of the invention is said to “alleviate” staphylococcal nasal colonization if it is able to decrease the number of colonies in the nares of a mammal when the MAb is administered before, concurrently with, or after exposure to staphylococci, whether that exposure results from the intentional instillation of staphylococcus or from general exposure. For instance, in the nasal colonization animal model described below, a MAb or collection of MAbs is considered to alleviate colonization if the extent of colonization, or the number of bacterial colonies that can be grown from a sample of nasal tissue, is decreased after administering the MAb or collection of MAbs. A MAb or collection of MAbs alleviates colonization in the nasal colonization assays described herein when it reduces the number of colonies by at least 50%, at least 60%, at least 75%, at least 80%, or at least 90%. 100% alleviation may also be referred to as eradication.
  • A MAb is said to “block” staphylococcal colonization if it is able to prevent the nasal colonization of a human or non-human mammal when it is administered prior to, or concurrently with, exposure to staphylococci, whether by intentional instillation or otherwise into the nares. A MAb blocks colonization, as in the nasal colonization assay described herein, if no staphylococcal colonies can be grown from a sample of nasal tissue taken from a mammal treated with the MAb of the invention for an extended period such as 12 hours or longer or 24 hours or longer compared to control mammals. A MAb also blocks colonization in the nasal colonization assay described herein if it causes a reduction in the number of animals that are colonized relative to control animals. For instance, a MAb is considered to block colonization if the number of animals that are colonized after administering the material and the Gram-positive bacteria is reduced by at least 25%, at least 50%, and at least 75%, relative to control animals or if no colonies can be grown from a sample taken from a treated individual for an extended period such as 12 hours or 24 hours or longer.
  • In a clinical setting, the presence or absence of nasal colonization in a human patient is determined by culturing nasal swabs on an appropriate bacterial medium. These cultures are scored for the presence or absence of staphylococcal colonies. In this type of qualitative assay system, it may be difficult to distinguish between blocking and alleviation of staphylococcal colonization. Thus, for the purposes of qualitative assays, such as nasal swabs, a MAb “blocks” colonization if it prevents future colonization in human patients who show no signs of prior colonization for an extended period of 12 or 24 hours or longer. A MAb “alleviates” colonization if it causes a discernable decrease in the number of positive cultures taken from a human patient who is already positive for staphylococci before the MAbs of the invention are administered.
  • A vaccine is considered to confer a protective immune response if it stimulates the production of opsonic antibodies to gram-positive bacteria. Production of opsonic antibodies may be measured by the presence of such antibodies in the serum of a test subject that has been administered the vaccine, relative to a control that has not received the vaccine. The presence of opsonic antibodies in the serum may be measured by the activity assays described herein, or by other equivalent assays. If an opsonophagocytic bactericidal assay is used, then killing by the test serum of at least 50% more bacteria, 75% more bacteria, and at least 100% more bacteria, relative to the control serum, is considered to be enhanced immunity.
  • Detailed Description of the Invention
  • The present invention provides murine antibodies, including monoclonal antibodies, and chimeric, humanized and fully human antibodies, fragments, derivatives, and regions thereof, which bind to lipoteichoic acid (LTA) of Gram positive staphylococci. Gram positive bacteria, unlike Gram negative bacteria, take up the Gram stain as a result of a difference in the structure of the cell wall. The cell walls of Gram negative bacteria are made up of a unique outer membrane of two opposing phospholipid-protein leaflets, with an ordinary phospholipid in the inner leaflet but the extremely toxic lipopolysaccharide in the outer leaflet. The cell walls of Gram positive bacteria seem much simpler in comparison, containing two major components, peptidoglycan and teichoic acids plus additional carbohydrates and proteins depending on the species.
  • Moreover, because the basis of the binding to Gram positive bacteria is the presence of LTA and because LTA is a major component of the cell walls of Gram positive bacteria and is highly conserved, the antibodies of the claimed invention are broadly reactive against Gram positive bacteria. This broad reactivity permits the antibodies of the invention to block the binding of Gram positive bacteria to epithelial cells, such as human epithelial cells (50-54). Finally, these antibodies exhibit broad opsonic activity and consequently enhance phagocytosis and killing of Gram positive bacteria. Accordingly, the invention provides broadly reactive, opsonic, and protective antibodies for the diagnosis, prevention, and/or treatment of bacterial infections caused by Gram positive bacteria.
  • Among the Gram positive Staphylococci against which the antibodies of the invention are directed are S. aureus (a coagulase positive bacteria) and S. epidermidis (a coagulase negative bacteria).
  • Three of the monoclonal antibodies of the invention (M110, M120, and MAb-391.4) bind strongly to LTA. M110 and M120 also exhibit high opsonic activity for S. epidermidis, while MAb-391.4 is also opsonic for S. epidermidis, but less so. M120 is also highly opsonic against S. aureus. M110 was derived from mice immunized with whole S. epidermidis strain Hay as described in detail in U.S. Pat. No. 6,610,293, which is incorporated herein by reference. In screening for hybridomas, the antibodies of one clone (hybridoma line 96-105CE11 IF6, which produces antibody M110) were found to bind very strongly to Gram positive bacteria such as strain Hay, all three serotypes of S. epidermidis, S. hemolyticus, S. hominus, and two serotypes of S. aureus, but not to the Gram negative control, Haemophilus influenza (see U.S. Pat. No. 6,610,293).
  • M120 was derived from mice immunized with conjugates of S. aureus LTA. The antibodies of one clone (00-107GG12 ID12, which produces antibody M120) were found to bind strongly to LTA, and were opsonic for S. aureus type 5 and S. epidermidis strain Hay.
  • MAb-391.4 is from QED Biosciences, and was derived from mice immunized with whole UV-killed S. aureus.
  • The variable regions of M110, M120, and 391.4 were sequenced and compared, revealing a surprising 88% identity (203/230) at the amino acid level. Further, the level of identity was found to be 96% (220/230) between the antibodies that are highly opsonic for S. epidermidis, M110 and M120. We believe that this level of homology between three monoclonal antibodies that were raised in three different mice, using three different antigen preparations from two different types of bacteria, is unprecedented. To understand how unexpected this finding is, one need only consider how vast and diverse is the collection of antibodies in the immune system.
  • The immune system is made up of a large number of B cells, each bearing antibodies of a different specificity, but only about 1 in 10,000 to 1 in 1,000,000 B cells is specific for a particular antigen. When a foreign antigen, such as is found on the surface of a bacteria, enters the blood stream, the appropriate B cell recognizes that antigen and then enters a lymph node where it undergoes rapid division to produce many progeny bearing the identical specificity. However, the rapidly dividing B cells also undergo somatic hypermutation. Somatic hypermutation results in about half of the B cells acquiring mutations in their rearranged heavy and light chain genes, with mutation occurring preferentially in complementarity determining regions (CDRs) of the variable regions. Mutated B cells that retain their ability to bind antigen continue to secrete antibody, while those that no longer bind antigen undergo apoptosis. As the antigen is cleared from the host, only B cells that have very high antigen affinity survive in a process called affinity maturation. The surviving activated B cells differentiate into plasma cells, which are short-lived and secrete antibody, and memory B cells, which are long-lived lymphocytes bearing membrane-bound antibody that can be rapidly stimulated when the antigen is re-introduced.
  • The processes of somatic hypermutation and affinity maturation result in progeny B cells that are of higher affinity and have immunoglobulins of different amino acid sequence than the original activated B cell. Therefore, a single B cell that is activated by a foreign antigen can produce many progeny of differing affinity and immunoglobulin amino acid sequence.
  • Because of these processes, it is generally believed that two animals immunized with the same antigen will produce vastly different antibody repertoires. Nickerson and colleagues demonstrated this concept when they showed that a mouse monoclonal antibody and a human monoclonal antibody that showed nearly identical binding to the same blood group A antigen shared only 15% and 37% identity in their heavy and light chain CDRs (55). X-ray crystallography studies of two antibodies that both bind to hemagglutinin of influenza virus, reveal that, although they share only 56% sequence identity, they both bind with similar affinities and in the same orientation to the same epitope (56).
  • It has been postulated that the immune system has evolved to provide a maximum range of antigen specificities and redundancy, rather than to bind to specific antigens (55). It follows, therefore, that antibodies derived from the same mouse may be of high specificity, but low homology, because any number of progenitor B cells may be specific for the immunized antigen. Amplification and somatic mutation of those progenitors may, however, result in groups of antibodies that are of higher homology within the group, although they are of very low homology between groups. Antibodies raised against the same immunogen in two or more different mice will necessarily be even less homologous, because they do not share progenitor B cells.
  • Three specific antibodies of the present invention, M110, M120, and MAb-391.4, were not only raised in different mice, but with different immunogens: M110 was raised to whole S. epidermidis, M120 was raised to purified and conjugated S. aureus LTA, and MAb-391.4 was raised to whole UV-killed S. aureus. Yet, though these antibodies were raised against different immunogen preparations in different mice, they share 88% identity at the amino acid level in both the heavy and light chain variable regions. This high degree of homology suggests that LTA contains a highly antigenic, and highly conserved, epitope which is bound by the three antibodies in a very similar manner. This epitope and mode of binding may be responsible for the high opsonic activity of the monoclonal antibodies.
  • MAb-391.4 and human/mouse chimeric antibodies of M110 and M120, designated A110 and A120, respectively, were tested for opsonic activity. MAb-391 .4, A110, and A120 each demonstrated a high level of opsonic activity against S. epidermidis strain Hay. (see also U.S. Pat. No. 6,610,293).
  • MAb A110 is currently being manufactured under GMP conditions in preparation for clinical trials. Additional disclosure regarding the MAb A110 is provided in U.S. Provisional Application Ser. No. 60/341,806, and in related application Methods for Blocking or Alleviating Staphylococcal Nasal Colonizaton by Intranasal Application of Monoclonal Antibodies, filed concurrently herewith, both of which are expressly incorporated by reference.
  • Thus, one aspect of the invention relates to antibodies that bind to the LTA of Gram positive bacteria, including both coagulase negative (S. epidermidis) and coagulase positive (S. aureus) bacteria, and that enhance the opsonization of such bacteria. These anti-LTA antibodies include monoclonal antibodies, such as M110, M120, and MAb-391.4, chimeric monoclonal antibodies A110, A120, A120a, and A120b, and other monoclonal antibodies including, chimeric, humanized, fully human antibodies, antibody fragments, and modified antibodies.
  • In a one aspect of the invention, as noted above, the antibody is a chimeric mouse/human antibody made up of regions from the anti-LTA antibodies of the invention together with regions of human antibodies. Chimeric or other monoclonal antibodies are advantageous in that they avoid the development of anti-murine antibodies. In at least one study, patients administered murine anti-TNF (tumor necrosis factor) monoclonal antibodies developed anti-murine antibody responses to the administered antibody (5). This type of immune response to the treatment regimen, commonly referred to as the human anti-mouse antibody response, or the HAMA response, decreases the effectiveness of the treatment and may even render the treatment completely ineffective. Humanized or chimeric human/mouse monoclonal antibodies have been shown to significantly decrease the HAMA response and to increase the therapeutic effectiveness (19).
  • Thus, in one aspect of the invention, a chimeric heavy chain can comprise the antigen binding region of the heavy chain variable region of the anti-LTA antibody of the invention linked to at least a portion of a human heavy chain IgG, IgA, IgM, or IgD constant region. This humanized or chimeric heavy chain may be combined with a chimeric light chain that comprises the antigen binding region of the light chain variable region of the anti-LTA antibody linked to at least a portion of the human light chain kappa or lambda constant region. Exemplary embodiments include, but are not limited to, an antibody having a mouse heavy chain variable region fused to a human IgGi constant region, and a mouse light chain variable region fused to a human kappa light chain constant region.
  • The chimeric antibodies and other MAb's of the invention may be monovalent, divalent, or polyvalent immunoglobulins. For example, a monovalent chimeric antibody is a dimer (HL) formed by a chimeric H chain associated through disulfide bridges with a chimeric L chain, as noted above. A divalent chimeric antibody is a tetramer (H2 L2) formed by two HL dimers associated through at least one disulfide bridge. A polyvalent or multivalent chimeric antibody may be based on an aggregation of chains, with or without a carrier or scaffold.
  • The MAbs of the invention include antibodies that contain heavy and light chain variable regions derived from two different antibodies. In one embodiment, the heavy and light chain variable regions are derived from two antibodies that bind to the same molecule, e.g. LTA. Exemplary embodiments include A120a, which is a human/mouse chimeric antibody that has a heavy chain variable region from A110 and a light chain variable region from A120; and A120b, which is a human/mouse chimeric antibody that has a heavy chain variable region from A120 and a light chain variable region from A110. Additional exemplary embodiments include antibodies that comprise a heavy chain variable region from MAb-391.4, and a light chain variable region from either of A110 or A120, and antibodies that comprise a light chain variable region from MAb-391.4, and a heavy chain variable region from either of A110 or A120.
  • In yet another aspect, the invention is a collection of opsonic monoclonal antibodies that bind to LTA and that exhibit a high degree of homology in the variable regions at either the amino acid or nucleic acid level, or both. In one embodiment, this collection comprises one or more of M110, M120, their human/mouse chimeric counterparts, A110, A120, and MAb-391.4. In one aspect, the amino acid sequences of the variable regions are at least 75% identical, at least 80% identical, at least 85% identical, at least 88% identical, at least 90% identical, or at least 95% identical as defined above.
  • In addition to the antibodies, the present invention also encompasses the DNA sequences of the genes coding for the antibodies (see, e.g., FIGS. 5, 6, and 9; SEQ ID NOs: 11, 13-15, 19, and 20) as well as the polypeptides encoded by the DNA (see, e.g., FIGS. 5, 6, and 10; SEQ ID NOs: 10, 12, 16, 17, 21, and 22). Those figures provide the variable regions of the heavy and light chains of A110, A120, and MAb-391.4, including the complementarity determining regions (CDRs), the hypervariable amino acid sequences within antibody variable regions that usually interact with the antigen. As noted above, the DNA and amino acid sequence homology between A110 and A120 is striking. There is a 94% homology (216/229) at the amino acid level and a 96% homology (662/687) at the DNA level between the antibodies. This suggests that these antibodies share a sequence and structural similarity.
  • The invention includes peptide sequences for, and DNA sequences encoding, full-length antibodies and portions thereof, as well as CDRs and FRs relating to these MAbs. The invention further includes DNA and peptide sequences that are homologous to these sequences. In one embodiment, these homologous DNAs and peptide sequences are about 70% identical, although other embodiments include sequences that are about 75%, 80%, 85%, 88%, 90%, and 95% or more identical. As indicated above, determining levels of identity for both the DNA and peptide sequence is well within the routine skill of those in the art.
  • As shown in FIG. 10A, alignment of the A110, A120, and 391.4 light chain variable regions (Seq. ID Nos. 16, 10, and 21, respectively) shows identical amino acids in 95 of 106 amino acids, or more than 89% identity overall. Within the region spanning the CDRs (amino acids 24 to 96 of the light chain variable regions) the percent identity is about 93% (68 out of 73 amino acids). It is predicted that light chain variable regions with a somewhat lower overall identity would still form MAbs that specifically bind LTA, and are therefore within the scope of the invention. The CDRs themselves show at least 88% identity, in particular, CDR1 (amino acids 24-33), CDR2 (amino acids 49-55), and CDR3 (amino acids 88-96), show 9/10, 7/7, and 8/9 identical amino acids. Likewise, the framework regions (FRs) surrounding the CDRs are also highly conserved: amino acids 1-23 of SEQ ID Nos. 16, 10, and 21 show greater than 86% identity (20/23 matching amino acids); amino acids 34-48 show about 93% identity (14/15); amino acids 56-87 show about 93% identity (68/73); and amino acids 97-106 show 70% identity.
  • Similarly, in FIG. 10B, alignment of the A110, A120, and 391.4 heavy chain variable regions (Seq. ID Nos. 17, 12, and 22, respectively) also shows a high degree of sequence identity. Counting single amino acid gaps and insertions as single-point mis-matches, Seq. ID Nos. 17, 12, and 22 show 86% identity overall (108/125 identical amino acids). It is predicted that heavy chain variable regions with a somewhat lower overall identity would still form MAbs that specifically bind LTA, and are therefore within the scope of the invention. The degree of identity is particularly high in the FR region preceding CDR1 through the FR region preceding CDR3, in particular, the 96 base region from amino acid 16 to 101 of Seq. ID Nos. 17, 12, and 22 shows 8 mismatches or approximately 91% identity. CDR1, itself, shows 90% identity over 10 amino acids (amino acids 26-35), and CDR2 (amino acids 50-69) shows about 89% identity over 19 amino acids. The framework regions surrounding the CDRs are also highly conserved. Amino acids 1-25 of SEQ ID Nos. 17, 12, and 22 show 92% identity (23/25 matching amino acids); amino acids 36-49 show 100% identity over 14 amino acids; the FR region between CDR2 and CDR3 (amino acids 70 to about 101) shows about 87% identity (over 31-32 amino acids); and amino acids 115-125 show 90% identity.
  • Thus, in one aspect, the invention encompasses polypeptides (including regions of larger polypeptides, such as MAbs) that 1) exhibit high sequence homology to Seq. ID Nos. 10, 12, 16, 17, 21, or 22, or defined regions thereof, and 2) are capable of functioning as all or part of the variable region of a MAb that specifically binds LTA. In one embodiment, such polypeptides comprise, or are at least 70%, 75%, 77% 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 93%, 95% identical to, any of Seq. ID Nos. 10, 12, 16, 17, 21, or 22. Conversely, polypeptides within the scope of the invention may be less than 100%, 99%, 95%, 90%, 80% or less identical to Seq. ID Nos. 10, 12, 16, 17, 21, or 22 provided that they are capable of functioning as all or part of the variable region of a MAb that specifically binds to LTA.
  • In another embodiment, polypeptides within the scope of the invention comprise, or are at least 70%, 75%, 77%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 93%, 95% identical to, amino acids 24 to 96 of any of Seq. ID Nos. 10, 16, or 21. In another embodiment, such polypeptides comprise, or are at least 70%, 75%, 77%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 93%, 95% identical to, 1) amino acids 24-33, 49-55, and 88-73 of Seq. ID Nos. 10, 16, or 21, or 2) amino acids 26-35 or 50-69 of Seq. ID Nos. 12, 17, or 22; and are capable of functioning as a CDR, or portion thereof, in a MAb that specifically binds to LTA. In another embodiment, such polypeptides comprise, or are at least 70%, 75%, 77%, 80%, 81%, 82%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 93%, 95% identical to, 1) amino acids 1-23, 34-48, 56-87, and 97-106 of Seq. ID Nos. 10, 12, 16, 17, 21, or 22, or 2) amino acids 1-25, 36-49, 70-101, or 115-125 of Seq. ID Nos. 12,17, or 22; and are capable of functioning as a framework region, or portion thereof, in a MAb that specifically binds to LTA.
  • The invention further comprises collections of a multiplicity of any of the above sequences capable of functioning as all or part of the variable region of a MAb that specifically binds to LTA, as part of a larger polypeptide, MAb, collection of MAbs or aggregation of MAbs; and the use thereof in prophylaxis, treatment, and for the production of pharmaceutical compounds or medicaments. The invention further comprises any non-naturally occurring RNA, DNA, or vector thereof, encoding any of the above sequences capable of functioning as all or part of the variable region of a MAb that specifically binds to LTA, as well as plasmids, viruses, bacteria, yeast, microorganisms, cell lines, transgenic plants or animals harboring or expressing such nucleic acids. Thus, the invention contemplates production systems for Mabs, light chains, heavy chains, and portions thereof, comprising 1) a cell (including bacteria, yeast, microorganisms, eukaryotic cell lines, transgenic plant or animal) in connection with 2) at least one recombinant nucleic acid capable of directing the expression of any of the Mabs or related polypeptides of the invention.
  • The invention thus further comprises a general method of identifying highly antigenic and highly conserved epitopes by raising antibodies against different immunogen preparations in different mice, sequencing the variable regions of the antibodies, comparing the variable regions, and identifying antibodies that share a high degree of homology in the variable regions.
  • The DNA sequences of the invention can be identified, isolated, cloned, and transferred to a prokaryotic or eukaryotic cell for expression by procedures well-known in the art. Such procedures are generally described in Molecular Cloning: A Laboratory Manual, as well as Current Protocols in Molecular Biology (44, 45), which are incorporated by reference. Guidance relating more specifically to the manipulation of sequences of the invention may be found in Antibody Engineering, and Antibodies: A Laboratory Manual (64, 65), both of which are incorporated by reference in their entirety. In certain embodiments, a CDR can be grafted onto any human antibody framework region using techniques standard in the art, in such a manner that the CDR maintains the same binding specificity as in the intact antibody. As noted as above, an antibody that has its CDRs grafted onto a human framework region is said to be “humanized”. Humanized, and fully human antibodies generally also include human constant regions, thus maximizing the percentage of the antibody that is human-derived, and potentially minimizing the HAMA response.
  • In addition, the DNA and peptide sequences of the antibodies of the invention, including both monoclonal and chimeric antibodies, humanized and fully human antibodies, may form the basis of antibody “derivatives,” which include, for example, the proteins or peptides encoded by truncated or modified genes. Such proteins or peptides may function similarly to the antibodies of the invention. Other modifications, such as the addition of other sequences that may enhance the effector function, which includes phagocytosis and/or killing of the bacteria, are also within the present invention.
  • The present invention also discloses a pharmaceutical composition comprising the antibodies, whether monoclonal or chimeric, humanized, or fully human, together with a pharmaceutically acceptable carrier. The pharmaceutical compositions of the invention may alternatively comprise the isolated antigen, epitope, or portions thereof, together with a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers can be sterile liquids, such as water, oils, including petroleum oil, animal oil, vegetable oil, peanut oil, soybean oil, mineral oil, sesame oil, and the like. Saline solutions, aqueous dextrose, and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical carriers are described in Remington's Pharmaceutical Sciences, 18th Edition (13), which is herein incorporated by reference.
  • Additionally, the invention may be practiced with various delivery vehicles and/or carriers. Such vehicles may increase the half-life of the MAbs in storage and upon administration including, but not limited to, application to skin, wounds, eyes, lungs, or mucus membranes of the nasal or gastrointestinal tract, or upon inhalation or instillation into the nares. These carriers comprise natural polymers, semi-synthetic polymers, synthetic polymers, lipososmes, and semi-solid dosage forms (21, 29, 33, 35, 36, 46). Natural polymers include, for example, proteins and polysaccharides. Semi-synthetic polymers are modified natural polymers such as chitosan, which is the deacetylated form of the natural polysaccharide, chitin. Synthetic polymers include, for example, polyphosphoesters, polyethylene glycol, poly(lactic acid), polystyrene sulfonate, and poly(lactide coglycolide). Semi-solid dosage forms include, for example, dendrimers, creams, ointments, gels, and lotions. These carriers can also be used to microencapsulate the MAbs or be covalently linked to the MAbs.
  • Finally, the present invention provides methods for treating a patient infected with, or suspected of being infected with, a Gram-positive bacteria such as a staphylococcal organism. The method comprises administering a therapeutically effective amount of a pharmaceutical composition comprising the anti-LTA immunoglobulin (whether monoclonal, chimeric, humanized, or fully human, including fragments, regions, and derivatives thereof) and a pharmaceutically acceptable carrier. A patient can be any human or non-human mammal in need of prophylaxis or other treatment. Representative patients include any mammal subject to S. aureus or other staphylococcal or Gram-positive infection or carriage, including humans and non-human animals such as mice, rats, rabbits, dogs, cats, pigs, sheep, goats, horses, primates, ruminants including beef and milk cattle, buffalo, camels, as well as fur-bearing animals, herd animals, laboratory, zoo, and farm animals, kenneled and stabled animals, domestic pets, and veterinary animals.
  • A therapeutically effective amount is an amount reasonably believed to provide some measure of relief, assistance, prophylaxis, or preventative effect in the treatment of the infection. A therapeutically effective amount may be an amount believed to be sufficient to block a bacterial infection. Similarly, a therapeutically effective amount may be an amount believed to be sufficient to alleviate a bacterial infection. Such therapy as above or as described below may be primary or supplemental to additional treatment, such as antibiotic therapy, for a staphylococcal infection, an infection caused by a different agent, or an unrelated disease. Indeed, combination therapy with other antibodies is expressly contemplated within the invention.
  • A further embodiment of the present invention is a method of preventing such infections, comprising administering a prophylactically effective amount of a pharmaceutical composition comprising the anti-LTA antibody (whether monoclonal, chimeric, humanized, or fully human) and a pharmaceutically acceptable carrier.
  • A prophylactically effective amount is an amount reasonably believed to provide some measure of prevention of infection by Gram positive bacteria. Such therapy as above or as described below may be primary or supplemental to additional treatment, such as antibiotic therapy, for a staphylococcal infection, an infection caused by a different agent, or an unrelated disease. Indeed, combination therapy with other antibodies is expressly contemplated within the invention.
  • The antibodies and the pharmaceutical compositions of the invention may be administered by intravenous, intraperitoneal, intracorporeal injection, intra-articular, intraventricular, intrathecal, intramuscular or subcutaneous injection, or intranasally, dermally, intradermally, intravaginally, orally, or by any other effective method of administration. The composition may also be given locally, such as by injection to the particular area infected, either intramuscularly or subcutaneously. Administration can comprise administering the pharmaceutical composition by swabbing, immersing, soaking, or wiping directly to a patient. The treatment can also be applied to objects to be placed within a patient, such as dwelling catheters, cardiac valves, cerebrospinal fluid shunts, joint prostheses, other implants into the body, or any other objects, instruments, or appliances at risk of becoming infected with a Gram positive bacteria, or at risk of introducing such an infection into a patient.
  • As a particularly valuable corollary of treatment with the compositions of the invention (pharmaceutical compositions comprising anti-LTA antibodies, whether, monoclonal, chimeric, humanized or fully human) may be the reduction in cytokine release that results from the introduction of the LTA of a Gram positive bacteria (49). As is now recognized in the art, LTA induces cytokines, including, for example, tumor necrosis factor alpha, interleukin 6, and interferon gamma (see, e.g., (37)). Accordingly, the compositions of the invention may enhance protection at three levels: (1) by binding to LTA on the bacteria and thereby blocking the initial binding to epithelial cells and preventing subsequent invasion of the bacteria; (2) by binding to LTA on bacteria and thereby enhancing opsonization of the bacteria and clearance of the bacteria from tissues and/or blood; and/or (3) by binding to LTA and partially or fully blocking cytokine release and modulating the inflammatory responses to prevent shock and tissue destruction.
  • Having generally described the invention, it is clear that the invention overcomes some of the potentially serious problems described in the Background section regarding the development of antibiotic resistant Gram positive bacteria. As set forth above, Staphylococci and Streptococci (such as S. faecalis) have become increasingly resistant to antibiotics and, with the recent spread of vancomycin resistant strains, antibiotic therapy may become totally ineffective.
  • Particular aspects of the invention are now presented in the form of the following Materials and Methods, as well as the specific Examples. Of course, these are included only for purposes of illustration and are not intended to be limiting of the present invention.
  • Materials and Methods Bacteria
  • S. aureus, type 5, is deposited at the ATCC under Accession No. 49521.
  • S. epidermidis, strain Hay, was deposited at the ATCC on Dec. 19, 1990 under Accession No. 55133.
  • Hybridoma
  • Hybridoma 96-105CE11 IF6 (M110) was deposited at the American Type Culture Collection (ATCC), located at P.O. Box 1549, Manassas, Va. 20108, U.S.A., on Jun. 13, 1997, under ATCC Accession No. HB-12368.
  • Hybridoma 00-107GG12 ID12 (M120) was deposited at the American Type Culture Collection (ATCC), located at P.O. Box 1549, Manassas, Va. 20108, U.S.A., on Aug. 16, 2001, under ATCC Accession No. PTA-3644.
  • Hybridoma 391.4 was deposited at the American Type Culture Collection (ATCC), located at P.O. Box 1549, Manassas, Va. 20108, U.S.A., on Dec. 18, 2001, under ATCC Accession No. PTA-3932.
  • Isotype Determination Assay
  • Isotype was determined using a mouse immunoglobulin isotype kit obtained from Zymed Laboratories (Cat. No. 90-6550).
  • Binding Assays
  • In the binding assays of the invention, immunoglobulin is incubated with a preparation of whole cell staphylococci or with a preparation of bacterial cell wall components such as LTA or PepG. The binding assay may be an agglutination assay, a coagulation assay, a colorimetric assay, a fluorescent binding assay, or any other suitable binding assay that is known in the art. A particularly suitable assay is either an enzyme-linked immunosorbent assay (ELISA) or a radio-immunoassay (RIA). Binding is detected directly and can also be detected indirectly by using competitive or noncompetitive binding procedures known in the art.
  • The whole cell staphylococcus preparation, LTA preparation, PepG preparation, or a combination of those preparations, may be fixed using standard techniques to a suitable solid support, including, but not limited to, a plate, a well, a bead, a micro-bead, a paddle, a propeller, or a stick. Solid supports may be comprised of, for example, glass or plastic. In certain embodiments of the invention, the solid support is a microtiter plate.
  • Generally, a binding assay requires the following steps. First, the fixed preparation is incubated with an immunoglobulin source. In one embodiment of the assay, the immunoglobulin source is, for example, tissue culture supernatant or a biological sample such as ascites, plasma, serum, whole blood, or body tissue. In another embodiment, the immunoglobulin may be further isolated or purified from its source by means known in the art, and the purified or isolated immunoglobulin is subsequently used in the assay. The amount of binding is determined by comparing the binding in a test sample to the binding in a negative control. A negative control is defined as any sample that does not contain antigen-specific immunoglobulin. In the binding assay, a positive binding reaction results when the amount of binding observed for the test sample is greater than the amount of binding for a negative control. Positive binding may be determined from a single positive/negative binding reaction or from the average of a series of binding reactions. The series of binding reactions may include samples containing a measured amount of immunoglobulin that specifically binds to the fixed antigen, thereby creating a standard curve. This standard curve may be used to quantitate the amount of antigen-specific immunoglobulin in an unknown sample.
  • In an alternate embodiment of the assay, antibodies are fixed to a solid support and an unknown immunoglobulin sample is characterized by its ability to bind a bacterial preparation. The other aspects of the assays discussed above apply where appropriate.
  • The specific binding assays used in the Examples are set forth below:
  • Live Bacteria ELISA (LBE): The LBE assay was performed to measure the ability of antibodies to bind to live bacteria. Various types of bacteria may be used in this assay, including S. aureus type 5, type 5-USU, type 8, S. epidermidis strain Hay, and S. hemolyticus. Bacteria from an overnight plate culture were transferred to 35 mis of Tryptic Soy Broth (TSB) and grown with gentle shaking for 1.5-2.0 hours at 37° C. The bacteria were then pelleted by centrifugation at 1800-2000×g for 15 minutes at room temperature. The supernatant was removed and the bacteria were resuspended in 35-45 mis of phosphate buffered saline containing 0.1% bovine serum albumin (PBS/BSA). The bacteria were again pelleted by centrifugation, the supernatant discarded and the bacteria resuspended in PBS/BSA to a percent transmittance (% T) of 65%-70% at 650 nm. From this suspension the bacteria were further diluted 15-fold in sterile 0.9% sodium chloride (Sigma cat. no. S8776, or equivalent), and 100 μl of this suspension was added to replicate wells of a flat-bottomed, sterile 96-well plate.
  • Each antibody to be tested was diluted to the desired concentration in PBS/BSA containing 0.05% Tween-20 and horse radish peroxidase-conjugated Protein A (Protein A-HRP, Zymed Laboratories) at a 1:8000 dilution (PBS/BSA/Tween/Prot A-HRP). The Protein A-HRP was allowed to bind to the antibodies for 30-60 minutes at room temperature before use, thereby generating an antibody-Protein A-HRP complex to minimize the potential non-specific binding of the antibodies to the Protein A found on the surface of S. aureus. Generally, several dilutions of test antibody were used in each assay. From each antibody dilution, 50 μl of the antibody-Protein A-HRP complex was added to replicate wells and the mixture of bacteria and antibody-Protein A-HRP complex was incubated at 37° C. for 30-60 minutes with gentle rotation (50-75 rpm) on an orbital shaker.
  • Following the incubation, the bacteria were pelleted in the plate by centrifugation at 1800-2000×g for 10-15 minutes at room temperature. The supernatant was carefully removed from the wells and 200 μl of PBS/BSA containing 0.05% Tween-20 (PBS/BSA/Tween) was added to all wells to dilute unbound reagents. The bacteria were again pelleted by centrifugation and the supernatant was removed. Two hundred μl of PBS/BSA/Tween was again added to all wells and the bacteria were again pelleted by centrifugation as described above. The supernatant was removed and 100 μl of TMB substrate (BioFx, Inc. cat. no. TMBW-0100-01, or equivalent) was added to each well and the hydrolysis of the substrate was allowed to proceed for 15 minutes at room temperature. The reactions were stopped by adding 100 μl of TMB stop reagent (450 nm Stop Reagent; BioFx, Inc. catalog no. STPR-0100-01, or equivalent). The absorbance of each well was determined using a microplate reader fitted with a 450 nm filter.
  • In this assay, the intensity of the color development was directly proportional to the binding of the antibodies to the bacteria. Control wells contained bacteria and Protein A-HRP without antibody.
  • Immunoassay on Methanol-Fixed Bacteria: Heat-killed bacteria were suspended in sterile 0.9% sodium chloride (Sigma cat. no. S8776, or equivalent) at a % transmittance (% T) of 70-75% at 650 nm. Ten milliliters the bacterial suspension was diluted 15-fold in sterile 0.9% sodium chloride and then pelleted by centrifugation at 1800×g for 15 minutes at 10-15 C. The supernatant was discarded and the pellet was resuspended in 15 milliliters of methanol (MeOH). One hundred microliters of the bacteria-MeOH suspension was distributed into each well of Nunc Maxisorp Stripwells (Nunc catalog no. 469949). The MeOH was allowed to evaporate, fixing the bacteria to the plastic wells. The bacteria-coated stripwells were stored in plastic bags in the dark at room temperature and used within 2 months of preparation.
  • For evaluation of antibodies, the bacteria-coated plates were washed four times with phosphate buffered saline containing 0.05% Tween-20 (PBS-T) as follows. Approximately 250 microliters of PBS-T was added to each well. The buffer was removed by flicking the plate over the sink and the remaining buffer removed by inverting the plate and tapping it on absorbent paper. The antibody was diluted in PBS-T and then added to the wells. Supernatants, ascites, or purified antibodies were tested at the dilutions indicated in the Examples. Control wells received PBS-T alone. After addition of the antibody, the wells were incubated at room temperature for 30-60 minutes in a draft-free environment. The wells were again washed four times with PBS-T. Ninety-five microliters of detection antibody was then added to each well. The detection antibody was one of the following: rabbit anti-mouse IgG3, rabbit anti-mouse IgM, or goat anti-human IgG (gamma-specific), all conjugated to horse radish peroxidase (HRP) and diluted 1:6000 in PBS-T (Zymed catalog numbers 61-0420, 61-6820 and 62-8420, respectively).
  • Following another 30-60 minute incubation at room temperature, the wells were washed four times with PBS-T and each well received 100 ul of TMB substrate solution (BioFx #TMBW-0100-01). Plates were incubated in the dark at room temperature for 15 minutes and the binding reactions were stopped by the addition of 100 μl of TMB stop solution (BioFx #STPR-0100-01). The absorbance of each well was measured at 450 nm using a Molecular Devices Vmax plate reader.
  • Immunoassay with Protein A: In order to evaluate the binding of the MAbs to S. aureus, the immunoassay procedure was modified for methanol-fixed bacteria, described above. Because S. aureus expresses Protein A on its surface, and Protein A binds strongly to the constant region of the heavy chains of gamma-globulins, it is possible that false positive results may be obtained from non-specific binding of the antibodies to Protein A. To overcome this difficulty, the immunoassay wells were coated with bacteria as described above, but prior to the addition of the antibodies to the bacteria-coated wells, the MAbs were incubated with a solution of recombinant Protein A conjugated to HRP (Zymed Laboratories Cat. No. 10-1123), diluted 1:10,000 in PBS-T. The binding reaction was allowed to proceed for 30 minutes at room temperature. The wells were washed four times with PBS-T and 100 μl of the solution of each Protein A-HRP-MAb combination was added to the wells. The presence of the Protein A-HRP from the pretreatment minimized the binding of the MAbs to the Protein A on the S. aureus. Furthermore, the binding of the Protein A-HRP to the constant region of the heavy chain did not interfere with the antibody binding site on the MAbs, thereby allowing evaluation of the MAbs on S. aureus and other bacteria.
  • The Protein A-HRP-MAb solutions were allowed to bind in the coated wells for 30-60 minutes at room temperature. The wells were then washed with PBS-T and TMB substrate solution was added and the assay completed as described above.
  • Immunoassay on LTA: The binding of the MAbs to LTA was measured by immunoassay on wells coated with S. aureus LTA (Sigma Cat. No. 2515). One hundred microliters of a 1 μg/ml LTA solution in PBS was distributed into replicate Nunc Maxisorp Stripwells and incubated overnight at room temperature. The unbound material was removed from the wells by washing four times with PBS-T. Antibody, diluted in PBS-T, was then added to the wells and the assay continued as described above for the Immunoassay on Methanol-Fixed Bacteria.
  • For immunoassays on PepG, Nunc Maxisorp Stripwell plates were coated with 100 ul of a 5-10 μg/ml solution of PepG (gift of S. Foster) in 0.1 M carbonate buffer (pH 9.2-9.6) overnight at room temperature. Unbound antigen was removed from the plate by washing four times with PBS-T. Antibody, diluted in PBS-T, was then added to the wells and the assay continued as described above for the Immunoassay on Methanol-Fixed Bacteria.
  • Activity Assays
  • Antibodies that bind to an antigen may not necessarily enhance opsonization or enhance protection from infection. Therefore, an opsonization assay was used to determine the functional activities of the antibodies.
  • An opsonization assay can be a colorimetric assay, a chemiluminescent assay, a fluorescent or radiolabel uptake assay, a cell-mediated bactericidal assay, or any other appropriate assay known in the art which measures the opsonic potential of a substance and thereby identifies reactive immunoglobulin. In an opsonization assay, an infectious agent, a eukaryotic cell, and the opsonizing substance to be tested, or an opsonizing substance plus a purported opsonizing enhancing substance, are incubated together.
  • In certain embodiments, the opsonization assay is a cell-mediated bactericidal assay. In this in vitro assay, an infectious agent such as a bacterium, a phagocytic cell, and an opsonizing substance such as immunoglobulin, are incubated together. Any eukaryotic cell with phagocytic or binding ability may be used in a cell-mediated bactericidal assay. In certain embodiments, phagocytic cells are macrophages, monocytes, neutrophils, or any combination of these cells. Complement proteins may be included to promote opsonization by both the classical and alternate pathways.
  • The opsonic ability of an antibody is determined by the amount or number of infectious agents remaining after incubation. The fewer the number of infectious agents that remain after incubation, the greater the opsonic activity of the antibody tested. In a cell-mediated bactericidal assay, opsonic activity is measured by comparing the number of surviving bacteria between two similar assays, only one of which contains the antibody being tested. Alternatively, opsonic activity is determined by measuring the number of viable organisms before and after incubation with a sample antibody. A reduced number of bacteria after incubation in the presence of antibody indicates a positive opsonizing activity. In the cell-mediated bactericidal assay, positive opsonization is determined by culturing the incubation mixture under appropriate bacterial growth conditions. Any reduction in the number of viable bacteria comparing pre-incubation and post-incubation samples, or between samples which contain immunoglobulin and those that do not, is a positive reaction.
  • Neutrophil-Mediated Opsonophagocytic Bactericidal Assay: The assay was performed using neutrophils isolated from adult venous blood by sedimentation using PMN Separation Medium (Robbins Scientific catalog no. 1068-00-0). Forty microliters of antibody, serum, or other immunoglobulin source, was added at various dilutions to replicate wells of a round-bottom microtiter plate. Forty microliters of neutrophils (approximately 2×106 cells per well) was then added to each well, followed immediately by approximately 3×104 mid-log phase bacteria (S. epidermidis strain Hay, ATCC 55133 or S. aureus type 5, gift from S. Wilson, Uniformed Services University of the Health Sciences) in 10 ul Tryptic Soy Broth (Difco cat. no. 9063-74, or equivalent). Finally, 10 μl of immunoglobulin-depleted human serum was added as a source of active complement. (Immunoglobulins were removed from human serum complement by preincubating the serum with Protein G-agarose and Protein L-agarose before use in the assay, this depletion of immunoglobulins minimized the concentrations of anti-staphylococcal antibodies in the complement, thereby reducing bacterial killing caused by inherent antibodies in the complement solution.)
  • The plates were incubated at 37° C. with constant, vigorous shaking. Aliquots of 10 μl were taken from each well at zero time, when the sample antibody was first added, and after 2 hours of incubation. To determine the number of viable bacteria in each aliquot harvested from each sample well, each aliquot was diluted 20-fold in a solution of 0.1% BSA in water (to lyse the PMNs), mixed vigorously by rapid pipetting, and cultured on blood agar plates (Remel, cat. no. 01-202, or equivalent) overnight at 37° C. The opsonic activity was measured by comparing the number of bacterial colonies observed from the sample taken at two hours with the nurjnber of bacterial colonies observed from the sample taken at time zero. Colonies were enumerated using an IPI Minicount Colony Counter.
  • Nasal Colonization Assay: The mouse nasal colonization model for S. aureus was based on the work of Kiser et al. (47). Briefly, streptomycin resistant S. aureus type 5 is grown on high salt Columbia agar (Difco) to promote capsule formation. The bacteria are washed with sterile saline (0.9% NaCI in water) to remove media components and resuspended at ˜108 bacteria/animal dose in saline (0.9% NaCl in water) containing various concentrations and combinations of anti-staphylococcal or irrelevant control MAbs. Following one hour preincubation, the bacteria are pelleted and resuspended in a final volume of 10 μl per animal dose in either saline or saline containing antibody. Mice that have been maintained on streptomycin-containing water for 24 hours are sedated with anesthesia. Staphylococci are injected into the nares of the mice by pipetting without contacting the nose.
  • After four to seven days, during which the animals are maintained on streptomycin-containing water, the animals are sacrificed and the noses removed surgically and dissected. Nasal tissue is vortexed vigorously in saline (0.9% NaCl in water) plus 0.5% Tween-20 to release adherent bacteria and the saline is plated on Columbia blood agar (Remel) and tryptic soy agar (Difco) containing streptomycin to determine colonization.
  • The invention, having been described above, may be better understood by reference to examples. The following examples are intended for illustration purposes only, and should not be construed as limiting the scope of the invention in any way.
  • Example 1 The Production of Hybridomas and Monoclonal Antibodies
  • Antibodies were raised against lipoteichoic acid (LTA) from S. aureus by immunizing mice with an LTA conjugate. LTA conjugates LTA/PspA, LTA/SIA/TT, and LTA/GMBS/TT, prepared as set forth below, were used.
  • To prepare each conjugate, LTA was first derivatized with thiol groups as follows. S. aureus LTA (Sigma Chemical Co.) was purified essentially as described in Fischer et al. (9). The purified LTA was diluted to 4 mg/ml with water. One hundred microliters of 0.75 M HEPES, 10 mM EDTA, pH 7.5 and 100 μl of 0.1 M SPDP (Pierce) were added to 1 ml of S. aureus LTA. The reaction was incubated for 4 hours at room temperature, and then 55 μl of 0.5 M DTT was added and the solution was dialyzed overnight at 4° C. against 2 mM EDTA, pH 5 (2×1 L). The reaction resulted in 0.27 mM thiol (LTA-SH) in a 1.2 mL volume, or 0.32 μmol thiol, as determined by DTNB assay (2).
  • LTA/PspA conjugate was prepared as follows. Three milligrams of pneumococcal surface protein A (PspA; 188 μl of a 16 mg/ml solution in PBS; prepared essentially as described in Wortham et al. (43) was combined with 25 μl of 0.75 M HEPES, 10 mM EDTA, pH 7.3 and 17 ul 0.1 M N-hydroxysuccinimidyl iodoacetate (SIA; Bioaffinity Systems) and incubated for 2 hours at room temperature. The volume of the solution was then made up to 2 ml with 10 mM sodium acetate, 0.15 M NaCl, 2 mM EDTA, and then concentrated to a final volume of about 150 μl using an Ultrafree 4 device (30 kDa cutoff; Amicon). The resulting iodoacetyl PspA was then combined with 400 μl of LTA-SH. The pH was raised to 8 with 1 M HEPES pH 8, and the reaction proceeded overnight at 4° C.
  • The solution was then fractionated on a 1×60 cm S-200HR column, which had been equilibrated with 0.1% deoxycholate (DOC) in PBS. The void volume fractions were pooled, dialyzed into saline (0.15 M NaCl) to remove DOC and PBS, and had an optical density of 0.14 at 280 nm. The concentration of protein in the conjugate solution was 0.66 mg/ml by BCA assay (Pierce Chemical Company), and the concentration of phosphate in the conjugate solution was 0.88 mM by phosphate assay (1).
  • LTA/SIA/TT conjugate was prepared as follows. Four milligrams of tetanus toxoid (TT; 280 μl of 14.5 mg/ml; SmithKline Beecham), diluted to 4 ml with 2 M NaCl was concentrated to 50 μl using an Ultrafree 4 centrifugal filter with a 30 kD cutoff (Millipore). The resulting solution was diluted to 250 μl with 2 M NaCl (TT/2 M NaCl). Seventy-five microliters of 0.25 M HEPES, 2 mM EDTA, pH 7.5 and 8 μl N-hydroxysuccinimide iodoacetate (SIA; Bioaffinity Systems, Roscoe, Ill.) were added to 125 μl of TT/2 M NaCl. The reaction was incubated for 2 hours at room temperature and then diluted to about 2 ml with 2 M NaCl. The solution was then concentrated to 150 μl using an Ultrafree 4 centrifugal filter.
  • One hundred and fifty microliters of the resulting product, iodoacetylated TT, was combined with 400 μl of LTA-SH. The reaction is incubated overnight at 4° C. The reaction was fractionated on a 1×60 cm Sephacryl S-200HR column (Pharmacia), equilibrated with 0.1% deoxycholate in PBS. The void volume fractions, containing the LTA/SIA/TT conjugate, were pooled, dialyzed into saline to remove DOC and PBS, and had an optical density of 0.77 at 280 nm. The yield of TT in the conjugate was 0.77 mg/ml by BCA assay (Pierce Chemical Co.). The concentration of phosphate in the conjugate solution was 0.77 mM by phosphate assay (1).
  • LTA/GMBS/TT conjugate was prepared as follows. Four milligrams of TT (280 μl of 14.5 mg/ml; SmithKline Beecham), diluted to 4 ml with 2 M NaCl was concentrated to 50 μl using an Ultrafree 4 centrifugal filter with a 30 kD cutoff (Millipore). The resulting solution was diluted to 250 ul with 2 M NaCl (TT/2 M NaCl). Seventy-five microliters of 0.15 M HEPES, 2 mM EDTA, pH 7.5 and 8 μl N-hydroxysuccinimide gamma butyric maleimide (GMBS; Bioaffinity Systems, Roscoe, Ill.) were added to 125 μl of TT/2 M NaCl. The reaction was incubated for 2 hours at room temperature and then diluted to 2 ml with 2 M NaCl. The solution was then concentrated to 150 μl using an Ultrafree 4 centrifugal filter.
  • Four hundred microliters of LTA-SH was added to the concentrated solution, and the pH was raised to 8 with 1 M HEPES pH 8. The reaction was incubated overnight at 4° C. The reaction was fractionated on a 1×60 cm Sephacryl S-200HR column (Pharmacia), equilibrated with 0.1% deoxycholate in PBS. The void volume fractions, containing the LTA/GMBS/TT conjugate, were pooled and dialyzed into saline to remove DOC and PBS. The yield of TT was 0.83 mg/ml by BCA assay (Pierce Chemical Co.), and the concentration of phosphate in the conjugate solution was 1.45 mM by phosphate assay (1).
  • The presence of LTA in each of the conjugates was confirmed by Western blot following 12% SDS-PAGE electrophoresis of the product.
  • Twenty-four approximately 4 month old female BALB/c mice were separated into six groups and immunized with 10 μg (groups A, C, and E) or 1 μg (groups B, D, and F) of LTA/PspA (groups A and B), LTA/SIA/TT (groups C and D), or LTA/GMBS/TT (groups E and F; Table 1).
  • TABLE 1
    LTA Immunization Groups
    Immun.
    Group Antigen ug/mouse Mouse Ids
    A LTA/PspA 10 1375-1378
    B LTA/PspA 1 1379-1382
    C LTA/SIA/TT 10 1383-1386
    D LTA/SIA/TT 1 1387-1390
    E LTA/GMBS/TT 10 1391-1394
    F LTA/GMBS/TT 1 1395-1398
  • All immunizations were administered subcutaneously in 50% RIBI adjuvant. The mice received a boost 21 days after the primary immunization, and a second boost 79 days after the primary immunization. Boosts were performed as described for the primary immunizations. Eyebleeds were taken at 0 days, 21 days, 35 days, 79 days, 94 days, and 119 days after the primary immunization. Serum collected at 21 days and 35 days was tested by ELISA for antibodies to LTA (Table 2).
  • TABLE 2
    Anti-LTA Titers of Serum Pools
    prebleed 21 day 35 day
    Group Antigen μg/mouse titer* titer* titer*
    A LTA/PSPA 10 77 2885 4748
    B LTA/PSPA 1 57 2668 4667
    C LTA/SIA/TT 10 199 51353 54085
    D LTA/SIA/TT 1 2520 11525 26229
    E LTA/GMBS/TT 10 783 11631 140392
    F LTA/GMBS/TT 1 10 3635 85832
    *titer is the serum dilution required to obtain an absorbance of 0.5 in an anti-LTA ELISA
  • Serum collected at 35 days and 79 days was also tested for antibodies to LTA by ELISA, and serum collected at 94 days was tested by ELISA, and in an LBE assay against S. epidermidis strain Hay and S. aureus (Table 3).
  • TABLE 3
    Comparison of Anti-LTA Titers and LBE Titers
    ELISA* ELISA* ELISA* LBE** LBE***
    Group Antigen μg/mouse 35 day 79 day 94 day S. epi 94 S. aur 94
    A LTA/PSPA 10 16053 4110 65241 531 26
    B LTA/PSPA 1 23505 8806 156343 150 13
    C LTA/SIA/TT 10 153227 39034 279505 520 29
    D LTA/SIAAT 1 98798 20135 313256 980 50
    E LTA/GMBS/TT 10 230410 46859 299995 1903 409
    F LTA/GMBS/TT 1 88756 24338 447440 2475 541
    *The ELISA titer is the serum dilution required to obtain an absorbance of 1.0
    **The LBE titer for S. epidermidis strain Hay is the serum dilution required to obtain an absorbance of 1.0.
    ***The LBE titer for S. aureus is the serum dilution required to obtain an absorbance of 0.5.
  • Based on the results of the ELISA assays and LBE assays, day 94 and day 119 sera from individual mice in groups E and F were tested by ELISA, and in an opsonic assay against S. aureus (Table 4).
  • Mouse 1396, which had been immunized with LTA/GMBS/TT, was selected because serum from the mouse showed a strong signal by anti-LTA ELISA, and was opsonic against S. aureus. Mouse 1396 was boosted one more time at day 134, and then sacrificed on day 141, and spleen removed and used to make hybridomas.
  • TABLE 4
    Anti-LTA ELISA and Opsonic Assay of Individual Mouse Sera
    S. aureus opsonic assay**
    Antigen ELISA* ELISA* Prebleed Prebleed day day day day day
    ID Dose prebleed day 119 neat 1:5 94 94 119 119 119
    1391 10 16364 53 20 68 0 40 14
    1392 10 18142 47915 19 53 0 1 21
    1393 10 18870 65 2 12 37
    1394 10 22 32126 77 25 78 33 29 50 67
    1395 1 29091 7 6 44 16 23 17 39
    1396 1 0 90249 53 20 74 64
    1397 1 0 40833 53 0 33 0
    1398 1 0 16601 60 27 40 31 37
    *serum dilution required to obtain an absorbance of 1.0.
    **numbers are percent killing.
  • Hybridomas were prepared by the general methods of Shulman, Wilde and Kohler; and Bartal and Hirshaut (34, 48). A total of 2.08×108 spleenocytes from mouse 1396 were mixed with 2.00×107 SP2/0 mouse myeloma cells (ATCC Catalog number CRL1581) and pelleted by centrifugation (400×g, 10 minutes at room temperature) and washed in serum free medium. The supernatant was removed to near-dryness and fusion of the cell mixture was accomplished in a sterile 50 ml centrifuge conical by the addition of 1 ml of warm (37° C.) polyethylene glycol (PEG; mw 1400; Boehringer Mannheim) over a period of 60-90 seconds. The PEG was diluted by slow addition of serum-free medium in successive volumes of 1, 2, 4, 8, 16 and 19 mls. The hybridoma cell suspension was gently resuspended into the medium and the cells pelleted by centrifugation (500×g, 10 minutes at room temperature). The supernatant was removed and the cells resuspended in medium RPM11640, supplemented with 10% heat-inactivated fetal bovine serum, 0.05 mM hypoxanthine and 16 μM thymidine (HT medium). One hundred μl of the hybridoma cells were planted into 952 wells of 96-well tissue culture plates. Eight wells (column 1 of plate A) received approximately 2.5×104 SP2/0 cells in 100 μl. The SP2/0 cells served as a control for killing by the selection medium added 24 hours later.
  • Twenty four hours after preparation of the hybridomas, 100 μl of RPMI 1640, supplemented with 10% heat-inactivated fetal bovine serums, 0.1 mM hypoxanthine, 0.8 μM aminopterin and 32 μM thymidine (HAT medium) was added to each well.
  • Forty-eight hours after the preparation of the hybridomas, the SP2/0 cells in plate A, column 1 appeared to be dead, indicating that the HAT selection medium had successfully killed the unfused SP2/0 cells.
  • Ten days after the preparation of the hybridomas, supernatants from all wells were tested by ELISA for the presence of antibodies reactive with methanol-fixed S. aureus LTA. Based on the results of this preliminary assay, cells from 12 wells were transferred to a 24-well culture dish. Three days later, supernatant from these cultures were retested by ELISA for the presence of antibodies that bind to LTA.
  • The absorbance values for eleven of the culture supernatants were less than 0.100. However, the absorbance value obtained with the supernatant from hybridoma culture 00-107GG12 was 4.000. This culture was expanded for further evaluation and cloned into two 96-well culture dishes. Cloning was accomplished by diluting the cell suspension into 4.5 viable cells per ml in RPM11640, supplemented with 15% fetal bovine serum, 5% Hybridoma SFM (Life Technologies) and 100 μg/ml of kanamycin.
  • Ten days later, the supernatants from the hybridoma clones were tested by ELISA for binding to S. aureus LTA. Only one clone, ID12, bound strongly to LTA, with an absorbance of 3.500). In contrast, the absorbance values for the remaining supernatants were less than 0.220. Hybridoma clone 00-107GG12 ID12 was expanded and cryopreserved. Isotype determination revealed that both the original hybridoma (00-107GG12) and its clone (00-107GG12 ID12) were mouse IgG2a heavy chains with kappa light chains. The monoclonal antibody produced by hybridoma 00-107GG12 ID12 was designated M120.
  • Example 2 Opsonic Activity of M120
  • Opsonic assays were carried out substantially as described above under the heading “Neutrophil-mediated Opsonophagocytic Bacteriacidal Assay”. M120 was purified from ascites essentially as described by the manufacturer of MEP Hypercel gel (BiSepra). Thirty-three ml of buffer A (50 mM Tris, 5 mM EDTA, pH 8) was added to 17 ml of mouse ascites, and then centrifuged for 15 minutes at 4000 rpm in an Eppendorf model 5810R centrifuge using rotor A462. The solution was filtered using Whatman GD/XP PES 0.45μ membrane (cat. no. 6994-2504) and the volume of diluted ascites was 47 ml after filtering. The solution was loaded onto a 1 cm×7 cm MEP hypercel column that had been equilibrated with buffer A, at a rate of 1.8 ml/min. The column was washed with buffer A, and then with buffer A+25 mM sodium caprylate until the OD280 was <0.05. The column was then washed with water until the OD280<0.05. The column was eluted with buffer B (50 mM sodium acetate, 5 mM EDTA, pH 4) and the eluent collected at a rate of 70 drops/min. The main peak (pool A) and its trailing end (pool B) were pooled separately and dialyzed against PBS (2×2 L) at 4° C. The dialyzed solution was sterile filtered using a Millex GV device (Millipore). By OD280, pool A contained 3.2 mg/ml antibody, and pool B contained 0.25 mg/ml antibody.
  • First, the opsonic activity of M120 was determined against S. aureus Type 5 (Table 5).
  • TABLE 5
    Opsonic Activity of M120 (200 μg/ml) against S. aureus Type 5
    Description Assay 1 Assay 2 Assay 3 Assay 4
    PMNs alone 0 0 0 0
    C alone 0 9 0 0
    PMNs + C 0 16 25 11
    M120 alone 20 0 0 0
    M120 + PMNs + C′ 73 84 85 85
  • Next, the opsonic activity of M120 was determined against S. epidermidis strain Hay (Table 6). This assay was also performed as described above under the heading “Neutrophil-Mediated Opsonophagocytic Bactericidal Assay”.
  • TABLE 6
    Opsonic activity of M120 against S. epidermidis strain Hay
    MAb
    Description (μg/ml) % killed
    PMNs alone 0
    C′ alone 0
    PMNs + C′ 0
    M120 alone 200 17
    M120 + PMNs + C′ 200 95
    100 97
    30 75
    10 87
    3.3 55
  • A similar assay was used to determine the opsonic activity of MAb-391.4 against S. epidermidis strain Hay (Table 7). Thus, MAb-391.4, which was raised against UV-killed S. aureus, has strong opsonic activity against S. epidermidis strain Hay.
  • TABLE 7
    Opsonic activity of MAb-391.4 against S. epidermidis strain Hay
    MAb
    Description (pg/ml) % killed
    PMNs alone 10.9
    C′ alone 0
    PMNs + C′ 0
    M120 alone 120 24.7
    M120 + PMNS + C′ 120 81.9
    50 57.9
  • Example 3 Cloning of the M120 Variable Regions
  • Total RNA was isolated from 4×106 frozen 00-107 GG12 ID12 hybridoma cells using the Midi RNA Isolation kit (Qiagen) following the manufacturer's procedure. The RNA was dissolved in 10 mM Tris, 0.1 mM EDTA (pH 8.4) containing 0.03 U/(μg Prime RNase Inhibitor (Sigma) to a final concentration of 025 μg/μl.
  • FIG. 3 shows the strategy for cloning the variable region genes. The total RNA (2 μg) was converted to cDNA by using Superscript II-MMLV Reverse Transcriptase (Life Technologies) and mouse Kappa chain-specific primer (JSBX-18; SEQ ID NO: 5) and a mouse heavy chain-specific primer (JSBX-25A; SEQ ID NO: 6) according to the manufacturer's procedures (see FIG. 4 for primer sequences). The first strand cDNA synthesis products were then purified using a Centricon-30 concentrator device (Amicon). Of the 40 μl of cDNA recovered, 5 μl was used as template DNA for PCR. Typical PCR amplification reactions (50 μl) contained template DNA, 30 pmoles of the appropriate primers (JSBX-9A, 11 A, and 18 for light chains, SEQ ID NOs: 3-5; JSBX-1, 4 and JSBX-25A for heavy chains, SEQ ID NOs: 1, 2, and 6), 2.5 units of ExTaq polymerase (PanVera), 1×ExTaq reaction buffer, 200 μM dNTP, 2 mM MgC12. The template was denatured by an initial incubation at 96° C. for 3 min. The products were amplified by 30 thermal cycles of 96° C. for 1 min., 60° C. for 30 sec, 72° C. for 30 seconds. The PCR products from the successful reactions were purified using the Nucleospin PCR Purification system (Clontech) as per manufacturer's procedure.
  • The PCR products (approximately 400 base pairs each) were then cloned into a bacterial vector, pGEM T (Promega) for DNA sequence determination. PCR fragments were ligated into pGEM T, a T/A style cloning vector, following the manufacturer's procedures using a 3:1 insert to vector molar ratio. One half (5 μl) of the ligation reactions were used to transform Ultracompetent XL1 Blue cells (Stratagene) as per the manufacturer's procedure. Bacterial clones containing plasmids with DNA inserts were identified using diagnostic restriction enzyme digestions with Dralll and BsiWI (for heavy chain clones) or Dralll and EcoRV (for light chain clones) (New England Biolabs). The DNA sequences of plasmids containing inserts of the appropriate size (˜400 bp) were then determined. The plasmid containing the A120 heavy chain sequence was designated pJSB16-6 and the plasmid containing the A120 light chain variable region was designated pJSB17-23. The final consensus DNA sequences of the light chain and heavy chain variable regions are shown in FIG. 5 and FIG. 6, respectively.
  • Having sequenced the variable regions of both M110 and M120, we compared them. The homology was striking at both the DNA and amino acid levels. As set forth in FIG. 9 there is a 96% homology at the DNA level, with 662 out of 687 bases the same Further, at the amino acid level, there is a 94% homology, with 216 amino acids out of 225 the same, as set forth in FIG. 10. As noted above, M120 was raised agains S. aureus LTA, while M110 was raised against S. epidermis strain Hay. Both antibodies exhibit opsonic activity against both S. epidermis and S. aureus. The high level of homology between the M110 and M120 variable regions may suggest a common structural motif that contributes to the opsonic capability of the antibodies.
  • Example 4 Production of Recombinant Chimeric Mouse/Human Antibody Molecules
  • The heavy and light chain variable regions were then subcloned into a mammalian expression plasmid vector for production of recombinant chimeric mouse/human antibody molecules. The human/mouse chimera of the M120 antibody is designated A120, and the human/mouse chimera of the M110 antibody is designated A110 (see U.S. Pat. No. 6,610,293, filed Jun. 15, 1998).
  • As set forth below, vectors were designed that express recombinant antibody molecules under the control of CMV transcriptional promoters. The chimeric heavy chains are expressed as a fusion of a heavy chain variable region and a human IgG1 constant domain. The chimeric light chains are expressed as a fusion of a light chain variable region and a human kappa chain constant region. The chimeric light chain cDNA contains a mouse kappa intron between the variable region and the human kappa constant region. After splicing, the variable region becomes fused to a human Kappa constant region exon. The selectable marker for the vector in mammalian cells is Neomycin resistance (resistance to G418).
  • The variable region gene fragments of M120 were re-amplified by PCR using primers that adapted the fragments for cloning into the expression vector {see FIG. 4, JSBX-46 through JSBX-49, SEQ ID NOs: 7-9 and 18). The heavy chain front primer (JSBX-46; SEQ ID NO: 7) includes a 5′ tail that encodes the C-terminus of the heavy chain leader and a Ss/WI restriction site for cloning, while the heavy chain reverse primer (JSBX-47; SEQ ID NO: 8) adds a 3′ EcoRI restriction site for cloning. This results in the addition of two amino acids, glutamine (E) and phenylalanine (F) between the heavy chain variable region and the human lgG1 constant region. The light chain front primer (JSBX-48; SEQ ID NO: 9) introduces a 5′ tail that encodes the two C-terminal amino acids of the light chain leader and an Age/restriction site for cloning purposes. The light chain reverse primer (JSBX-49; SEQ ID NO: 18) adds a 3′ DNA sequence for the joining region-Kappa exon splice junction followed by a BstBI restriction site for cloning. The variable regions were re-amplified from the plasmid DNA using vector pJSB16-6 for the heavy chain variable region and vector pJSB17-23 for the light chain variable region. PCR reactions were performed as described above. Following a 3 minute incubation at 96° C., the PCR parameters were 30 thermal cycles of 58° C. for 30 seconds, 70° C. for 30 seconds, and 96° C. for 1 minute.
  • The heavy chain variable region PCR product was digested with BsiWI and EcoRI (New England Biolabs), purified using a Nucleospin PCR Purification column (Clontech), as described by the manufacturer, and ligated into BsiWI/EcoRI/PfIMI-digested and gel-purified pJRS383 vector using the Takara Ligation Kit (Panvera) following the manufacturer's procedure. The ligation mix was then transformed into XL1Blue cells (Stratagene), resulting in plasmid mammalian expression vector pJSB23-1 (FIG. 7). The light chain variable region PCR product (approximately 350 bp) was digested with AgeI and BstBI (New England Biolabs), and purified using a Nucleospin PCR Purification column (Clontech) as described by the manufacturer. The light chain variable region fragment was ligated into pJRS384 that had been AgeI/BstBI/Xcml-digested and gel-purified using the Takara Ligation Kit (Panvera) following the manufacturer's procedure. The ligation mix was transformed into XL1Blue cells (Stratagene), resulting in mammalian expression plasmid pJSB24 (FIG. 8).
  • Because of the similarity between the A110 and the A120 antibody sequences, we decided to construct mammalian cell expression plasmids that contained the A120 heavy and light chain variable regions in a bi-cistronic plasmid, as well as plasmids that combined the A110 heavy chain variable region with the A120 light chain variable region, and the A110 light chain variable region with the A120 heavy chain variable region, in order to investigate the binding and opsonic properties of the different antibodies. Construction of the bi-cistronic vectors was done in a step-wise fashion, in which the heavy and light chain variable regions of A120 were cloned into a bi-cistonic expression plasmid already containing the A110 light and heavy chain variable regions (pJRS354, FIG. 11), replacing the A110 light chain variable region, heavy chain variable region, or both. The plasmid pJRS354 was digested with ClaI and XhoI (New England Biolabs), the digestion products were separated on an agarose gel and the backbone fragment was cut out and gel purified using a Nucleospin Gel Fragment DNA Purification column (Clontech), as described by the manufacturer. The plasmid pJSB24 was digested with ClaI and XhoI (New England Biolabs), the digestion products were separated on an agarose gel and the light chain variable region fragment was cut out and gel purified using a Nucleospin Gel Fragment DNA Purification column (Clontech), as described by the manufacturer. These fragments were then ligated together using the Takara Ligation Kit (Panvera) following the manufacturer's procedure. The resulting bi-cistronic expression vector, pJSB25-3 (FIG. 12), which contained the A120 antibody light chain variable region and the A110 antibody heavy chain variable region, was then used for antibody production in transfected mammalian cells after sequence confirmation of the variable regions.
  • The two other bi-cistronic plasmids were constructed in a similar manner. The plasmid pJSB25-3 was digested with BspEI and NotI (New England Biolabs), the digestion products were separated on an agarose gel and the backbone fragment was cut out and gel purified using a Nucleospin Gel Fragment DNA Purification column (Clontech), as described by the manufacturer. The plasmid pJSB23-1 was digested with BspEl and NotI (New England Biolabs), the digestion products were separated on an agarose gel and the heavy chain variable region fragment was cut out and gel purified using a Nucleospin Gel Fragment DNA Purification column (Clontech), as described by the manufacturer. These fragments were ligated together using the Takara Ligation Kit (Panvera) following the manufacturer's procedure. The resulting bi-cistronic expression vector, pJSB26 (FIG. 13), which contained the light and heavy chain variable regions of the A120 antibody, was then used for antibody production in transfected mammalian cells.
  • The plasmid pJRS354 was digested with BspEl and NotI (New England Biolabs), the digestion products were separated on an agarose gel and the backbone fragment was cut out and gel purified using a Nucleospin Gel Fragment DNA Purification column (Clontech), as described by the manufacturer. The plasmid pJSB23-1 was digested with BspEl and NotI (New England Biolabs), the digestion products were separated on an agarose gel and the heavy chain variable region fragment was cut out and gel purified using a Nucleospin Gel Fragment DNA Purification column (Clontech), as described by the manufacturer. These fragments were ligated together using the Takara Ligation Kit (Panvera) following the manufacturer's procedure and transformed into XUBlue cells (Stratagene). The resulting bi-cistronic expression vector, pJSB27 (FIG. 14), which contained the heavy chain variable region of the A120 antibody and the light chain variable region of the A110 antibody, was then used for antibody production in transfected mammalian cells.
  • Example 5 Comparison of the A120 and A110 Anti-LTA Human/Mouse Chimeric Antibodies
  • Anti-LTA human/mouse chimeric antibody A110 was previously described in U.S. patent application Ser. No. 09/097,055, now U.S. Pat. No. 6,610,293, which is herein incorporated by reference. The binding activities of anti-LTA human/mouse chimeric antibodies A110 and A120 were compared in ELISA assays against LTA.
  • Dilutions of A120 supernatant were compared to dilutions of purified A110 antibody in an immunoassay as described above under the heading “Binding Assays”, subheading “Immunoassay on LTA”. Briefly, the wells of a 96-well plate were coated with 1 μg/ml of S. aureus LTA for three hours at room temperature. After washing, dilutions of purified A110 antibody or A120 supernatant in PBS-T were added to quadruplicate wells and incubated for 30 to 60 minutes at room temperature. After washing, HRP-conjugated gamma-specific goat anti-human IgG, diluted 1:5000, was added to each well and incubated for 30 to 60 minutes at room temperature. After removing the secondary antibody and washing, 100 μl TMB substrate was added to each well and incubated for 15 minutes at room temperature. One hundred microliters of TMB stop reagent was then added to each well to stop the reaction, and the absorbance of each well at 450 nm was determined. The results of the anti-LTA ELISA assay are shown in Table 8.
  • TABLE 8
    Binding of A110 and A120 to LTA-coated plates
    A120
    A110 supernatant
    (ng/ml) A450 dilution A450
    40 2.691 10 4.000
    20 1.741 20 3.967
    10 0.952 40 3.927
    5 0.555 80 3.327
    2.5 0.322 160 2.824
    1.25 0.180 320 1.907
    0.625 0.115 640 1.148
    PBS-T 0.050 PBS-T 0.052

    This assay shows that monoclonal antibody A120, like A110, binds to LTA of S. aureus. In order to compare the binding affinity of the two antibodies, A120 was purified using Protein G Ultralink (Pierce) per the manufacturer's procedure, and the two antibodies were tested for binding to LTA in a second ELISA assay.
  • Dilutions of purified A110 and A120 antibodies were compared for binding to LTA in an ELISA assay, using substantially the same protocol as above. The data for the anti-LTA ELISA using dilutions of purified antibodies are shown in Table 9.
  • TABLE 9
    Binding of purified A110 and A120 to LTA-coated plates
    antibody
    concentration A450 A450
    (ng/ml) A110 A120
    8000 3.921 3.566
    2000 3.922 3.078
    500 3.960 1.445
    125 3.838 0.422
    31.25 2.398 0.131
    7.8125 0.903 0.068
    1.953 0.276 0.054
    PBS-T 0.047 0.048

    These data demonstrate that A110 has a greater affinity for S. aureus LTA than does A120. This difference is particularly striking at an antibody concentration of 125 ng/ml, where A120 gives an ELISA signal of 0.422, and A110 gives a signal that is nearly ten times stronger.
  • Example 6 Comparison of the Opsonic Activity of Anti-LTA Antibodies A110 and A120
  • Purified human/mouse chimeric A110 and A120, and mouse M120 MAbs were assayed for their opsonic activity as described above, under the heading “Neutrophil-Mediated Opsonophagocytic Bactericidal Assay.” Briefly, dilutions of purified A110, A120, or M120 antibodies were combined with neutrophils (PMNs) in the wells of a microtiter plate. Mid-log phase bacteria were added to each well, followed by immunoglobulin-depleted human serum, which serves as a source of complement (C). Samples were incubated for 2 hours at 37° C., and were then plated on blood agar and incubated overnight to determine the number of live bacteria remaining. The opsonic activity is expressed as“% killed”, which is determined according to the following formula: % killed=100%-N2hr/N0hr, where N2hr is the number of colonies formed after a 2 hour incubation with antibody, PMNs, and C\ and N0hr is the number of colonies formed after a 0 hour incubation. Control reactions lacked one of the above components. Table 10 shows the results of the opsonic activity assay for antibodies A110, A120, and M120.
  • TABLE 10
    Opsonic activity of A110, A120, and M120
    against S. epidermidis strain Hay
    MAb cone,
    Antibody Description (μg/ml) % killed
    A110 purified human/mouse 100 99
    chimeric
    anti-LTA MAb 50 96
    10 96
    A120 purified human/mouse 100 100
    chimeric
    anti-LTA MAb 50 99
    10 94
    M120 purified mouse 100 99
    anti-LTA MAb
    50 97
    purified mouse 10 94
    anti-LTA MAb
    PMNs alone purified mouse N/A 0
    anti-LTA MAb
    C′ alone purified mouse N/A 0
    anti-LTA MAb
    PMNs + C′ (no MAb) purified mouse N/A 14
    anti-LTA MAb
    A110 alone purified mouse 100 0
    anti-LTA MAb
    A120 alone purified mouse 100 0
    anti-LTA MAb
    M120 alone purified mouse 100 8
    anti-LTA MAb
  • These data demonstrate that MAbs A110, A120, and M120 are equally active in the opsonic activity assay described herein. Thus, the chimerization of M120 to make A120 has little or no effect on the opsonic activity of the antibody, and the two different anti-LTA chimeric antibodies, A110 and A120, are of comparable activity.
  • Example 7 Transient Production of Recombinant Chimeric Mouse/Human A120 Antibodies
  • The plasmids pJSB25, pJSB26 and pJSB27 were transected into COS cells grown in IMDM plus 10% fetal bovine serum, using Superfect (Qiagen) in 6 well tissue culture wells as described by the manufacturer. After two days the supernatant was assayed for the production of chimeric antibody and for the capability for the expressed antibody to bind to S. aureus LTA antigen.
  • Antibody production assays were preformed in 8-well strips from 96-well microtiter plates (Maxisorp F8; Nunc, Inc.) coated at a 1:500 dilution with a goat antihuman Fc (Pierce). The plates are covered with pressure sensitive film and incubated overnight at 4° C. Plates were then washed once with Wash solution (Imidazole/NaCl/0.4% Tween-20). One hundred microliters of culture supernatant dilutions were then applied to duplicate wells and allowed to incubate for 60 minutes on a plate rotator at room temperature. The plates were washed seven times with Wash solution. A Goat anti-Human IgG H+L-HRP (Zymed) conjugate was diluted 1:4000 in the sample/conjugate diluent. One hundred microliters was added to the samples, and then incubated on a plate rotator for 60 minutes at room temperature. The samples were washed as above and then incubated with 100 (xL/well of TMB developing substrate (BioFx) for 1 minute at room temperature. The binding reaction was stopped with 100 μUwell of Quench buffer (BioFx) and the absorbance value at 450 nm was determined using an automated microtiter plate ELISA reader. This assay (see FIG. 15) demonstrates that the transfection of cells with this plasmid construct results in the cells producing a molecule containing both human IgG and Kappa domains.
  • The supernatants were then assayed for the ability of the expressed antibodies to bind to lipoteichoic acid. The activity assays were preformed in 8-well strips from 96-well microtiter plates (Maxisorp F8; Nunc, Inc.) coated at 1 μg/mL with S. aureus LTA (Sigma) using PBS. The plates were covered and incubated overnight at 4° C. Plates were then washed once with PBS. One hundred microliters of culture supernatant dilutions were then applied to duplicate wells and allowed to incubate for 60 minutes on a plate rotator at room temperature. The plates were washed seven times with Wash solution. Goat anti-Human IgG H+L-HRP (Zymed) was diluted 1:4000 in the sample/conjugate diluent, and 100 ul were added to the samples, and then incubated on a plate rotator for 60 minutes at room temperature. The samples were washed as above and then incubated with 100 μL/well of TMB developing substrate (BioFx) for 10-15 minutes on a plate rotator at room temperature. The binding reaction was stopped with 100 μL/well of Quench buffer (BioFx) and the absorbance value at 450 nm was determined using an automated microtiter plate ELISA reader. As a positive control, the original human/mouse chimeric antibody A110 (produced by plasmid pJRS354) was used. This assay (FIG. 16) demonstrates that the transfection of cells with these plasmid constructs results in the cells producing a molecule that binds to the S. aureus LTA antigen.
  • These data demonstrate that the chimeric human antibody directed against LTA is opsonic and enhances survival against staphylococci. In addition, the antibody promotes clearance of the staphylococci from the blood. Thus antibody to LTA provides prophylactic and therapeutic capabilities against staphylococcal infections and vaccines using LTA or peptide mimeotopes of LTA that induce anti-LTA antibodies would also have prophylactic capabilities.
  • Example 8 Human Antibodies that Bind LTA
  • Rather than humanizing a mouse antibody to minimize the HAMA response during treatment as described above, a skilled artisan can isolate a protective anti-LTA antibody that is fully human. There are a number of well-known alternative strategies one of ordinary skill in the art may use to produce completely human recombinant antibodies. One is the generation of antibodies using phage display technologies (59, 63). Specifically, human RNA is used to produce a cDNA library of antibody heavy and light chain fragments expressed on the surface of bacteriophage. These libraries can be used to probe against the antigen of interest (i.e., LTA) and the phage that bind, because of the antibody expressed on the surface, are then isolated. The DNA encoding the variable regions is sequenced and cloned for antibody expression.
  • Another method of producing human antibodies employs “humanized” mice. These transgenic mice have had their own antibody genes replaced with a portion of the human antibody gene complex so that upon inoculation with antigen, they produce human antibodies (57, 59, 60, 61, 63). The antibody producing cells that result can then be incorporated into the standard hybridoma technology for the establishment of specific monoclonal antibody producing cell lines.
  • Recombinant human antibodies are also produced by isolating antibody-producing B cells from human volunteers that have a robust anti-LTA response. Using fluorescence activated cell sorting (FACS) and fluorescently labeled LTA, cells producing the anti-LTA antibodies can be separated from the other cells. The RNA can then be extracted and the sequence of the reactive antibody variable regions determined (58, 62). The DNA sequence of the functional variable regions can be synthesized or cloned into mammalian expression vectors for large-scale human recombinant antibody production.
  • CONCLUSION
  • Monoclonal antibodies were raised in mice against S. aureus LTA. One hybridoma that produced antibodies that bound strongly to LTA in an ELISA assay was subcloned further. Hybridoma subclone 00-107GG12 ID12 produced an IgG2a monoclonal antibody with a kappa light chain that bound strongly to LTA. The antibody produced by this hybridoma was designated M120 (Example 1).
  • M120 was tested in an opsonophagocytic bacteriocidal assay for opsonic activity against S. aureus type 5 and S. epidermidis strain Hay. The antibody was mixed with PMNs and complement, which was derived from human serum that had been depleted of anti-S. aureus and anti-S. epidermidis antibodies, and then tested for activity against the bacteria. M120 showed opsonic activity against both S. aureus and S. epidermidis, killing 95% of S. epidermidis and an average of 82% of S. aureus at 200 μg/ml (Example 2, Tables 5 and 6). MAb-391.4, which was raised to UV-killed S. aureus, was tested for opsonic activity against S. epidermidis strain Hay in a similar assay, and showed 81.9% killing (Table 7).
  • The M120 variable regions were then cloned and sequenced, and the sequence compared to another anti-LTA antibody, M110. Surprisingly, M110 and M120 were found to share about 94% sequence identity at the amino acid level, and about 96% sequence identity at the nucleotide level. A third anti-LTA antibody, MAb-391.4, was also sequenced compared to the other two. The three antibodies share 88% sequence identity at the amino acid level. This high level of sequence identity may suggest that the antibodies bind to a common epitope on LTA (Example 3, FIGS. 9 and 10). Human/mouse chimeric antibodies were then made, fusing the heavy chain variable region of either M120 or M110 to a human IgG1 constant region, and the light chain variable region of either M120 or M110 to a human kappa light chain constant region. The human/mouse chimera of M120 is referred to as A120 and the human/mouse chimera of M110 is referred to as A110. Because of the similarity between the two antibodies, an antibody that contained the heavy chain of A110 and the light chain of A120, designated A120a, was made. Similarly, an antibody that contained the light chain of A110 and the heavy chain of A120, designated A120b, was also made (Example 4).
  • The human/mouse chimeric antibodies A120 and A110 were tested for their ability to bind to LTA in an ELISA assay. Both chimeric antibodies bound strongly to LTA, indicating that replacing the mouse constant regions with human constant regions had little effect on the binding properties of the antibodies (Example 5, Tables 8 and 9). Next, the opsonic activity of chimeric antibodies A110 and A120, and of M120, were compared in an opsonic assay against S. epidermidis strain Hay. All three antibodies showed at least 94% killing of S. epidermidis. These results show that the chimeric antibodies are strongly opsonic against S. epidermidis, and because they have a reduced HAMA response in humans, they should be suitable therapeutic molecules for fighting Gram-positive bacterial infections (Example 6, Table 10).
  • Finally, three of the chimeric antibodies, A120, A120a, and A120b were produced in COS cells and tested for the ability to bind to S. aureus LTA. All three chimeric antibodies bound to LTA in the ELISA assay, with A120 and A120a showing the strongest binding. These results suggest that M110 and M120 do bind to a similar or overlapping epitope on LTA, because antibodies that have variable regions from both retain the ability to bind to the antigen. These results may indicate that a particular epitope on LTA is able to elicit antibodies that are opsonic against S. aureus and S. epidermidis. This epitope may be more accessible than others, or may be positioned such that antibodies that are bound are ideally situated to attract the factors required for opsonization of the bacterium.
  • Previously, it was unclear whether a monoclonal antibody could enhance phagocytosis, because the polyclonal sera that were used contained many different antibodies that bound to many different epitopes on the surface of the bacteria, and the sum of this collective binding and activities may have accounted for the overall activity of the serum. Here, we demonstrate that monoclonal antibodies, which bind to a single epitope on the surface of bacteria, can be opsonic against that bacteria. We have also demonstrated that monoclonal antibodies raised against LTA can have that activity, and that those antibodies may be opsonic for a number of different types of Gram-positive bacteria.
  • Furthermore, we have shown that three different monoclonal antibodies, one of which was raised to whole S. epidermidis, one to purified and conjugated LTA from S. aureus, and one to whole UV-killed S. aureus, share a striking degree of homology. This Jevel of homology between monoclonal antibodies that were raised to similar antigens in different mice has previously not been shown. In fact, it has long been believed that antibodies have evolved the ability to bind identical antigens using very dissimilar determinants to provide the body with a very broad antibody repertoire. The level of homology between the M110, M120, and MAb-391.4 variable regions may indicate that opsonic antibodies to LTA recognize a nearly identical epitope using nearly identical modes of binding, and that this mode of binding is important to their functional activity. Furthermore, the epitope to which the antibodies bind appears to be highly conserved between S. epidermidis and S. aureus, and may be common to most, if not all, Gram-positive bacteria. Monoclonal antibodies to this epitope may, therefore, be broadly opsonic against a wide range of bacteria, allowing researchers to develop a few antibodies that will have broad opsonic and protective activity against many Gram-positive bacteria.
  • The following literature references are herein specifically incorporated by reference:
    • 1. Ames, B. N. 1966. Assay of inorganic phosphate, total phosphate and phosphatase, Methods in Enzymology 8: 115-118.
    • 2. Ellman, G. L. 1959. Tissue Sulfhydryl Groups, Arch. Biochem. & Biophys. 82: 70
    • 3. Endl, J.; Seidl, H P.; Fiedler, F.; and Schleifer, K. H. 1983. Chemical composition and structure of cell wall teichoic acid of staphylococci, Arch Microbiol, 135: 215-223.
    • 4. Espersen, F.; Hertz, J. B.; and Hoiby, N. 1981. Cross-reactions between Staphylococcus epidermis and 23 other bacterial species, Ada Path. Microbial. Scand, Sect. B. 89: 253-260.
    • 5. Exley A. R.; Cohen J.; Buurman W.; Owen R.; Hanson G.; Lumley J.; Aulakh J. M.; Bodmer M.; Riddell A.; Stephens S.; et al. 1990. Monoclonal antibody to TNF in severe septic shock, Lancet 335: 1275-1277.
    • 6. Fattom A.; Shepherd S.; Karakawa W. 1992. Capsular polysaccharide serotyping scheme for Staphylococcus epidermidis, J. Clin. Micro. 30: 3270-3273.
    • 7. Fischer, Gerald W. Broadly reactive opsonic antibodies that react with common staphylococcal antigens, U.S. Pat. No. 5,571,511, issued Nov. 5, 1996.
    • 8. Fischer, Gerald W. Directed human immune globulin for the prevention and treatment of staphylococcal infections, U.S. Pat. No. 5,955,074, issued Sep. 21, 1999.
    • 9. Fischer W.; Koch H. U.; Haas R. 1983. Improved preparation of lipoteichoic acids, Eur. J. Biochem. 133: 523-530.
    • 10. Fleer, A.; Senders R. C.; Visser M. R.; Bijlmer R. P.; Gerards L. J.; Kraaijeveld C. A.; Verhoef J. 1983. Septicemia due to coagulase-negative staphylococci in a neonatal intensive care unit: clinical and bacteriological features and contaminated parenteral fluids as a source of sepsis, Pediatr. Infect Dis. 2:426-431.
    • 11. Fournier, Jean-Michel. 1991. Staphylococcus Aureus, Vaccines and Immunotherapy, Ch. 13, pp. 166-171.
    • 12. Garrett, Laurie. 1994. The Revenge of the Germs or Just Keep Inventing New Drugs, The Coming Plague, Ch. 13, Farrar, Straus and Giroux, N.Y., (ed.), pp. 411-456.
    • 13. Genarro, A. (ed.) 1990. Remington's Pharmaceutical Sciences, 1Sf h Edition, Mack Publishing, Easton, Pa.
    • 14. Hancock, I. C. 1997. Bacterial cell surface carbohydrates: Structure and assembly, Biochem. Soc. Trans. 25: 183-187.
    • 15. Jendeberg, Lena; Nilsson, Peter; Larsson, Antonella; Denker, Per; Uhlen, Mathias; Nilsson, Bjorn; Nygren, Per-Ake. 1997. Engineering of Fc1 and Fc3 from human immunoglobulin G to analyse subclass specificity for Staphylococcal Protein A, J. Immunol. Methods 201: 25-34.
    • 16. Kojima Y.; Tojo M.; Goldmann D. A.; Tosteson T. D.; Pier G. B. 1990. Antibody to the capsular polysaccharide/adhesin protects rabbits against catheter-related bacteremia due to coagulase-negative staphylococci, J. Infect Dis. 162: 435-441.
    • 17. Krieger, Monty; Joiner, Keith A. Method for treating Gram-positive septicemia, U.S. Pat. No. 5,624,904, issued Apr. 29, 1997.
    • 18. Lee, J. C. 1996. The prospects for developing a vaccine against Staphylococcus aureus, Trends in Micro. 4: 162-66.
    • 19. LoBuglio A. F.; Wheeler R. H.; Trang J.; Haynes A.; Rogers K.; Harvey E. B.; Sun L; Ghrayeb J.; Khazaeli M. B. 1989. Mouse/human chimeric monoclonal antibody in man: kinetics and immune response, P.N.A.S. 86: 4220-4224.
    • 20. Nakamura, K. et al. 1999. Uptake and release of budesonide from mucoadhesive, pH-sensitive copolymers and their application to nasal delivery. J. Control. Release 61:329-335.
    • 21. Natsume, H., S. Iwata, K. Ohtak, M. Miyamoto, M. Yamaguchi, K. Hosoya, and D. Kobayashi. 1999. Screening of cationic compounds as an absorption enhancer for nasal drug delivery. Int. J. Pharma. 185:1-12.
    • 22. Naumova, I. B.; Kuznetsov, V. D.; Kudrina, K. S.; and Bezzubenkova, A. P. 1980. The Occurrence of Teichoic Acids in Streptomycetes, Arch. Microbiol. 126:71-75.
    • 23. Navarre, William Wiley and Schneewind, Olaf. 1999. Surface proteins of Gram-positive bacteria and mechanisms of their targeting to the cell wall envelope, Microbiology and Molecular Biology Reviews 63:174-229.
    • 24. Osland, Arve; Grov, Arne; and Oeding, Per. 1980. Immunochemical analysis of the teichoic acid from Staphylococcus simulans, Acta Path. Microbiol. Scand, Sect. B., 88:121-123.
    • 25. Patrick, C. C. 1990. Coagulase-negative staphylococci: Pathogens with increasing clinical Significance, J. Pediatr. 116:497-507.
    • 26. Peterson, Phillip K.; Verhoef, Jan; Sabath, L. D.; and Quie, Paul G. 1977. Effect of Protein A on staphylococcal opsonization, Infection and Immunity 15: 760-764.
    • 27. Peterson, Phillip K.; Wilkinson, Brian J.; Kim, Youngki; Schmeling, David; and Quie, Paul G. 1978. Influence of Encapsulation on Staphylococcal Opsonization and Phagocytosis by Human Polymorphonuclear Leukocytes, Infection and Immunity 19: 943-949.
    • 28. Quie, Paul G.; Hill, Harry R.; and Davis, Todd A. 1974. Defective phagocytosis of Staphylococci, Annals New York Academy of Sciences, pp. 233-243.
    • 29. Ramkissoon-Ganorkar, C. et al. 1999. Modulating insulin-release profile from pH/thermosensivite polymeric beads through polymer molecular weight. J. Contr. Release 59:287-298.
    • 30. Raynor, Robert H.; Scott, David F.; and Best, Gary K. 1981. Lipoteichoic acid inhibition of phagocytosis of Staphylococcus aureus by Human Polymorphonuclear Leukocytes, Clinical Immunology and Immunopathology 19:181-189.
    • 31. Romero-Vivas J.; Rubio M.; Fernandez C; Picazo J. J. 1995. Mortality associated with nosocomial bacteremia due to methicillin-resistant Staphylococcus aureus, Clin. Infect. Dis. 21: 1417-23.
    • 32. Salton, M. R. J. 1994. The Bacterial Cell Envelope—A Historical Perspective, in J.-M. Ghuyson and R. Hakenbeck (ed.), Bacterial Cell Wall, Elsevier Science BV, Amsterdam, pp. 1-22.
    • 33. Schwab, U. E., A. E. Wold, J. L. Carson, M. W. Leigh, P.-W. Cheng, P. H. Gilligan and T. F. Boat. 1993. Increased adherence of Staphylococcus aureus from cystic fibrosis lungs to airway epithelial cells. Am. Rev. Respir. Dis. 148:365-369.
    • 34. Shulman, M.; Wilde, C D.; Kohler, G. 1978. A Better Cell Line for Making Hybridomas Secreting Specific Antibodies, Nature 276: 269-270.
    • 35. Soto, N., A. Vaghjimal, A. Stahl-Avicolli, J. Protic, L. Lutwick and E. Chapnick. 1999. Bacitracin versus mupirocin for Staphylococcus aureus nasal colonization. Infect. Cont. Hosp. Epidem. 20:351-353.
    • 36. Suzuki, Y. and Y. Makino. 1999. Mucosal drug delivery using cellulose derivative as a functional polymer. J. Control. Release. 62:101-107.
    • 37. Takada H.; Kawabata Y.; Arakaki R.; Kusumoto S.; Fukase K.; Suda Y.; Yoshimura T.; Kokeguchi S.; Kato K.; Komuro T.; et al. 1995. Molecular and structural requirements of a lipoteichoic acid from Enterococcus hirae ATCC 9790 for cytokine-inducing, antitumor, and antigenic activities, Infection and Immunity 63: 57-65.
    • 38. Takeda S.; Pier G. B.; Kojima Y.; Tojo M.; Muller E.; Tosteson T.; Goldmann D. A. 1991. Protection against endocarditis due to Staphylococcus epidermidis by immunization with capsular polysaccharide/adhesin, Circulation 86: 2539-2546.
    • 39. Timmerman C. P.; Besnier J. M.; De Graaf L.; Torensma R.; Verkley A. J.; Fleer A.; Verhoef J. 1991. Characterisation and functional aspects of monoclonal antibodies specific for surface proteins of coagulase-negative staphylococci, J. Med. Micro. 35: 65-71.
    • 40. Tomasz, Alexander. 2000. The Staphylococcal Cell Wall, in V. A. Fischetti et al. (ed.) Gram-Positive Pathogens, Ch. 36, pp. 351-355.
    • 41. Waldvogel, Francis A. 1990. Staphylococcus Aureus (Including Toxic Shock Syndrome), in Mandell, G. L. et al. (ed.) Principles and Practices of Infectious Diseases, Third Edition, Churchill Livingstone, New York, Ch. 173, pp. 1489-1510.
    • 42. West, Timothy E.; Cantey, J. R.; Apicella, Michael A.; and Burdash, N. M. 1983. Detection of anti-teichoic acid immunoglobulin G antibodies in experimental Staphylococcus epidermidis endocarditis, Infection and Immunity 42: 1020-1026.
    • 43. Wortham, Charles; Grinberg, Luba; Kaslow, David C; Briles, David E.; McDaniel, Larry S.; Lees, Andrew; Flora, Michael; Snapper, Clifford M.; and Mond, James J. 1998. Enhanced protective antibody response to PspA after intranasal or subcutaneous injections of PspA genetically fused to Granulocyte-Macrophage Colony-Stimulating Factor or Interleukin-2, Infection and Immunity 66:1513-1520.
    • 44. Sambrook, Joseph; Russell, David W. 1989. Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Press, Cold Spring Harbor, N.Y.
    • 45. Ausubel et al. (ed.) 1989. Current Protocols in Molecular Biology, John Wiley & Sons.
    • 46. Merkus, F. W., J. C. Verhoef, N. G. Schipper, and E. Marttin. 1999. Cyclodextrins in nasal drug delivery. Advan. Drug Deliv. Rev. 36:41-57.
    • 47. Kiser, Kevin B.; Cantey-Kiser, Jean M.; Lee, Jean C. 1999. Development and characterization of a Staphylococcus aureus nasal colonization model in mice. Infection and Immunity 67: 5001-5006.
    • 48. Bartal, Arie H.; Hirshaut, Yashar. 1987. Current Methods in Hybridoma Formation Bartal, A. H. et al. (ed.) Methods of Hybridoma Formation, Humana Press, Clifton, N.J.
    • 49. De Kimpe, S. J., M. Kengatharan, C. Thiemermann, J. R. Vane. 1995. The cell wall components peptidoglycan and lipoteichoic acid from S. aureus act in synergy to cause shock and multiple organ failure. Proc. Nat. Acad. Sci. (USA) 92:10359-10363.
    • 50. Carruthers, M. M., W. J. Kabat. 1983. Mediation of staphylococcal adherence to mucosal cells by lipoteichoic acid. Infect Immun. 40: 444-6.
    • 51. Chugh T D, Burns G J, Shuhaiber H J, Bahr G M. 1990. Adherence of Staphylococcus epidermidis to fibrin-platelet clots in vitro mediated by lipoteichoic acid. Infect Immun. 58: 315-9.
    • 52. Granato D, Perotti F, Masserey I, Rouvet M, Golliard M, Servin A, Brassart D. 1999. Cell surface-associated lipoteichoic acid acts as an adhesion factor for attachment of Lactobacillus johnsonii La1 to human enterocyte-like Caco-2 cells. Appl Environ Microbiol. 65:1071-7.
    • 53. Nealon T J, Mattingly S J. 1984. Role of cellular lipoteichoic acids in mediating adherence of serotype III strains of group B streptococci to human embryonic, fetal, and adult epithelial cells. Infect Immun. 43: 523-30.
    • 54. Teti G, Tomasello F, Chiofalo M S, Orefici G, Mastroeni P. 1987. Adherence of group B streptococci to adult and neonatal epithelial cells mediated by lipoteichoic acid. Infect Immun. 55: 3057-64.
    • 55. Nickerson, K. G.; Tao, M.-H.; Chen, H.-T.; Larrick, J.; Kabat, E. A. 1995. Human and mouse monoclonal antibodies to blood group A substance, which are nearly identical immunochemically, use radically different primary sequences. J. Biol. Chem. 270: 12457-12465.
    • 56. Fleury, D.; Daniels, R. S.; Skehel, J. J.; Knossow, M.; Bizebard, T. 2000. Structural evidence for recognition of a single epitope by two distinct antibodies. Proteins 40: 572-578.
    • 57. Green, L. L, M. C. Hardy, et al. (1994). “Antigen-specific human monoclonal antibodies from mice engineered with human Ig heavy and light chain YACs.” Nat Genet 7(1): 13-21.
    • 58. Kantor, A. B., C. E. Merrill, et al. (1995). “Development of the antibody repertoire as revealed by single-cell PCR of FACS-sorted B-cell subsets.” Ann NY Acad Sci 764: 224-7.
    • 59. Low, N. M., P. H. Holliger, et al. (1996). “Mimicking somatic hypermutation: affinity maturation of antibodies displayed on bacteriophage using a bacterial mutator strain.” J Mol Biol 260(3): 359-68.
    • 60. Wagner, S. D., A. V. Popov, et al. (1994). “The diversity of antigen-specific monoclonal antibodies from transgenic mice bearing human immunoglobulin gene miniloci.” Eur J Immunol 24(11): 2672-81.
    • 61. Wagner, S. D., G. T. Williams, et al. (1994). “Antibodies generated from human immunoglobulin miniloci in transgenic mice.” Nucleic Acids Res 22(8): 1389-93.
    • 62. Wang, X. and B. D. Stollar (2000). “Human immunoglobulin variable region gene analysis by single cell RT-PCR.” J Immunol Methods 244(1-2): 217-25.
    • 63. Winter, G., A. D. Griffiths, et al. (1994). “Making antibodies by phage display technology.” Annu Rev Immunol 12: 433-55.
    • 64. Borrebaeck, Carl A. K. 1995. Antibody Engineering, 2.sup.nd Ed., Oxford University Press, NY.
    • 65. Harlow, Ed; Lane, David. 1988. Antibodies: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, N.Y.
  • Having now fully described the invention, it will be appreciated by those skilled in the art that the invention can be performed within a range of equivalents and conditions without departing from the spirit and scope of the invention and without undue experimentation. In addition, while the invention has been described in light of certain embodiments and examples, the inventors believe that it is capable of further modifications. This application is intended to cover any variations, uses, or adaptations of the invention which follow the general principles set forth above.
  • Other embodiments of the invention will be apparent to those skilled in the art from consideration of the specification and practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with a true scope and spirit of the invention being indicated by the following claims.

Claims (27)

1. An isolated monoclonal antibody, or antigen-binding fragment thereof, that binds to lipoteichoic acid (LTA) of Gram positive bacteria, wherein at least a portion of the antibody variable region is obtained from a phage display library comprising human antibody sequences or is obtained from a cell comprising a human antibody sequence.
2. The isolated monoclonal antibody of claim 1 wherein at least a portion of the antibody variable region has been identified as binding to a peptide mimetic of an LTA epitope, Staphylococcus, or a Staphylococcus antigen.
3. The isolated monoclonal antibody of claim 1 wherein the phage display library comprising human antibody sequences is derived from sequences from a Staphylococcus seropositive human or human immunized with a Staphylococcus antigen or mimetic thereof.
4. (canceled)
5. The isolated monoclonal antibody of claim 1, wherein the cell is a human B-cell or a transgenic B-cell comprising a human antibody sequence.
6. The isolated monoclonal antibody of claim 5 wherein the transgenic B-cell is obtained from a transgenic animal comprising a human antibody sequence.
7. The isolated monoclonal antibody of claim 6 wherein the transgenic animal is a mouse that has been immunized with Gram positive bacteria, an antigen thereof, or mimetic thereof.
8. The isolated monoclonal antibody of claim 1, wherein the antibody is a fully human antibody.
9. An isolated human monoclonal antibody or fragment thereof that specifically binds to lipoteichoic acid (LTA) of Gram positive bacteria.
10. (canceled)
11. The isolated monoclonal antibody of claim 1, wherein the antibody specifically binds to LTA of Gram positive bacteria that are coagulase positive, coagulase negative, or both coagulase positive and coagulase negative Gram positive bacteria.
12. The isolated monoclonal antibody of claim 1, wherein the antibody specifically binds to LTA of Gram positive bacteria that are Staphylococcus epidermidis.
13. The isolated monoclonal antibody of claim 1, wherein the antibody specifically binds to LTA of Gram positive bacteria that are Staphylococcus epidermidis, Staphylococcus aureus, or both Staphylococcus epidermidis and Staphylococcus aureus.
14. The isolated monoclonal antibody of claim 1, wherein the antibody specifically binds to LTA of the Gram positive bacteria Staphylococcus epidermidis and Staphylococcus aureus.
15. The isolated monoclonal antibody of claim 1, wherein the antibody specifically binds to LTA of Gram positive bacteria that are multiple serotypes of Staphylococcus epidermidis, Staphylococcus aureus, or both Staphylococcus epidermidis and Staphylococcus aureus.
16. The isolated monoclonal antibody of claim 15, wherein the multiple serotypes of Staphylococcus aureus are serotype 5, serotype 8, or both serotype 5 and serotype 8.
17. The isolated monoclonal antibody of claim 1, wherein the antibody specifically binds to LTA of Gram positive bacteria that are Staphylococcus epidermidis and one or more Gram positive bacteria selected from the group consisting of Staphylococcus aureus, Streptococcus mutans, Streptococcus faecalis, and Streptococcus pyogenes.
18. The isolated monoclonal antibody of claim 1, wherein the antibody recognizes the peptide sequence WRMYFSHRHAHLRSP (SEQ ID NO:23), the peptide sequence WHWRHRIPLQLAAGR (SEQ ID NO:24), a peptide sequence having substantial identity to the peptide sequence WRMYFSHRHAHLRSP (SEQ ID NO:23), a peptide sequence having substantial identity to the peptide sequence WHWRHRIPLQLAAGR (SEQ ID NO:24), or combination thereof.
19. The isolated monoclonal antibody of claim 1, wherein the antibody recognizes LTA of Gram positive bacteria which are coagulase positive, or coagulase negative, or recognizes both coagulase positive and coagulase negative Gram positive bacteria.
20. The isolated monoclonal antibody of claim 1, wherein the antibody recognizes Gram positive bacteria which are Staphylococcus epidermis, Staphylococcus aureus, Streptococcus mutans, Streptococcus faecalis, Streptococcus pyogenes, or a combination thereof.
21.-28. (canceled)
29. The isolated antigen-binding antibody fragment of claim 1, wherein the fragment is a Fab, Fab′, F(ab′)2, SFv, light chain, heavy chain, or combination thereof.
30. A polynucleotide encoding an antibody, or fragment thereof, of claim 1.
31. A vector comprising the polynucleotide of claim 30.
32. A cell comprising the vector of claim 31.
33. An antibody, or antigen-binding fragment thereof, produced by the cell of claim 32.
34. A composition comprising an isolated monoclonal antibody or antigen-binding fragment thereof, and a pharmaceutically acceptable carrier, wherein the antibody specifically binds to lipoteichoic acid (LTA) of Gram positive bacteria and comprises a portion of a human antibody sequence.
US12/572,410 1998-06-15 2009-10-02 Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria Abandoned US20100247546A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/572,410 US20100247546A1 (en) 1998-06-15 2009-10-02 Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
US15/081,297 USRE48475E1 (en) 2004-07-29 2016-03-25 Amplification relay device of electromagnetic wave and a radio electric power conversion apparatus using the device

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US09/097,055 US6610293B1 (en) 1997-06-16 1998-06-15 Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
US34350301P 2001-12-21 2001-12-21
US10/323,927 US7250494B2 (en) 1998-06-15 2002-12-20 Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram positive bacteria
US11/724,040 US7777017B2 (en) 1998-06-15 2007-03-13 Nucleic acids encoding opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
US12/572,410 US20100247546A1 (en) 1998-06-15 2009-10-02 Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/724,040 Continuation US7777017B2 (en) 1998-06-15 2007-03-13 Nucleic acids encoding opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria

Publications (1)

Publication Number Publication Date
US20100247546A1 true US20100247546A1 (en) 2010-09-30

Family

ID=46281760

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/323,927 Expired - Fee Related US7250494B2 (en) 1998-06-15 2002-12-20 Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram positive bacteria
US11/724,040 Expired - Fee Related US7777017B2 (en) 1998-06-15 2007-03-13 Nucleic acids encoding opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
US12/572,410 Abandoned US20100247546A1 (en) 1998-06-15 2009-10-02 Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/323,927 Expired - Fee Related US7250494B2 (en) 1998-06-15 2002-12-20 Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram positive bacteria
US11/724,040 Expired - Fee Related US7777017B2 (en) 1998-06-15 2007-03-13 Nucleic acids encoding opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria

Country Status (1)

Country Link
US (3) US7250494B2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10364298B2 (en) 2011-11-18 2019-07-30 National Research Council Of Canada Clostridium difficile lipoteichoic acid and uses thereof
WO2022087122A1 (en) * 2020-10-20 2022-04-28 Longhorn Vaccines And Diagnostics, Llc Immunogenic antigens

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080139789A1 (en) * 1990-10-22 2008-06-12 Henry M. Jackson Foundation For The Advancement Of Military Medicine Isolated Broadly Reactive Opsonic Immunoglobulin for Treating a Pathogenic Coagulase-Negative Staphylococcus Infection
US6610293B1 (en) * 1997-06-16 2003-08-26 The Henry M. Jackson Foundation For The Advancement Of Military Medicine Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
US7250494B2 (en) * 1998-06-15 2007-07-31 Biosynexus Incorporated Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram positive bacteria
US20040052779A1 (en) * 2001-12-21 2004-03-18 Stinson Jeffrey R. Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram positive bacteria
AU2003298770A1 (en) * 2002-12-02 2004-06-23 Biosynexus Inc Wall teichoic acid as a target for anti-staphylococcal therapies and vaccines
WO2005072778A2 (en) * 2004-01-29 2005-08-11 Biosynexus, Inc. Use of amino-oxy functional groups in the preparation of vaccines conjugates
CA2687681A1 (en) 2007-05-31 2009-03-05 Merck & Co., Inc. Antigen-binding proteins targeting s. aureus orf0657n
CA2688490C (en) 2007-06-01 2022-06-21 Scott E. Strome Immunoglobulin constant region fc receptor binding agents
US20100260752A1 (en) 2009-01-23 2010-10-14 Biosynexus Incorporated Opsonic and protective antibodies specific for lipoteichoic acid of gram positive bacteria
US8647621B2 (en) 2009-07-27 2014-02-11 Fina Biosolutions, Llc Method of producing protein-carbohydrate vaccines reduced in free carbohydrate
US20120269814A1 (en) * 2009-11-10 2012-10-25 Amgen Inc. Anti-c mpl antibodies
DK2513056T3 (en) 2009-12-17 2022-05-23 Serum Inst Of India Pvt Ltd CHEMICAL REAGENTS FOR THE ACTIVATION OF POLYSACCHARIDES IN THE MANUFACTURE OF CONJUGATE VACCINES
US20110236399A1 (en) * 2010-03-29 2011-09-29 Biosynexus Incorporated Compositions and methods for prophylactic and therapeutic treatment of infection
US8383783B2 (en) 2010-04-23 2013-02-26 Serum Institute Of India, Ltd. Simple method for simultaneous removal of multiple impurities from culture supernatants to ultralow levels
CN105820256A (en) 2010-07-28 2016-08-03 格利克尼克股份有限公司 Fusion proteins of natural human protein fragments to create orderly multimerized immunoglobulin FC compositions
US9988439B2 (en) 2011-12-23 2018-06-05 Nicholas B. Lydon Immunoglobulins and variants directed against pathogenic microbes
WO2013096948A1 (en) 2011-12-23 2013-06-27 Lydon Nicholas B Immunoglobulins and variants directed against pathogenic microbes
EP2872535A1 (en) 2012-07-16 2015-05-20 Pfizer Inc. Saccharides and uses thereof
WO2015051159A1 (en) * 2013-10-02 2015-04-09 The Rockefeller University Amyloid protofibril antibodies and methods of use thereof
CA2939200A1 (en) * 2014-02-19 2015-08-27 Jody Berry Ebola monoclonal antibodies
US20180282424A1 (en) * 2014-11-06 2018-10-04 Gliknik Inc. Molecules with bimodal activity depleting target at low dose and increasing immunosuppression at higher dose
AU2017279538A1 (en) 2016-06-07 2019-01-03 Gliknik Inc. Cysteine-optimized stradomers
EP3551647A4 (en) 2016-12-09 2021-01-13 Gliknik Inc. Manufacturing optimization of gl-2045, a multimerizing stradomer
CA3053305A1 (en) 2017-02-17 2018-08-23 Mapp Biopharmaceutical, Inc. Monoclonal antibodies and cocktails for treatment of ebola infections

Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4196192A (en) * 1977-10-28 1980-04-01 American Cyanamid Company Combined Haemophilus influenzae type b and pertussis vaccine
US4451570A (en) * 1981-03-26 1984-05-29 The Regents Of The University Of California Immunoglobulin-secreting human hybridomas from a cultured human lymphoblastoid cell line
US4596769A (en) * 1984-03-05 1986-06-24 Temple University Monoclonal antibodies to peptidoglycan and methods of preparing same
US4744982A (en) * 1982-08-24 1988-05-17 Hunter Kenneth W Human monoclonal antibody reactive with polyribosylribitol phosphate
US4744892A (en) * 1985-02-27 1988-05-17 Henkel Kommanditgesellschaft Auf Aktien Process for dressing kaolinite by flotation
US4761283A (en) * 1983-07-05 1988-08-02 The University Of Rochester Immunogenic conjugates
US4954449A (en) * 1982-08-24 1990-09-04 The United States Of America As Represented By The Secretary Of The Army Human monoclonal antibody reactive with polyribosylribitol phosphate
US5043267A (en) * 1984-05-18 1991-08-27 E. I. Du Pont De Nemours And Company Method for rapid detection of bacterial and fungal infection
US5139933A (en) * 1989-09-26 1992-08-18 Vicam, L.P. Assay method for detecting listeria
US5475100A (en) * 1990-07-13 1995-12-12 Fujita Health University Artificial antibody
US5571511A (en) * 1990-10-22 1996-11-05 The U.S. Government Broadly reactive opsonic antibodies that react with common staphylococcal antigens
US5776456A (en) * 1992-11-13 1998-07-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5804370A (en) * 1994-06-08 1998-09-08 Critichem Medical Products Limited Early diagnosis of sepsis utilizing antigen-antibody interactions amplified by whole blood chemiluminescence
US5807688A (en) * 1995-05-27 1998-09-15 Zeneca Limited Catalytic antibodies for carbamate activation by a non-spontaneous reaction mechanism
US5866140A (en) * 1991-11-22 1999-02-02 Nabi Type I surface antigen associated with staphylococcus epidermidis
US5871951A (en) * 1996-09-23 1999-02-16 The Children's Hospital Of Philadelphia Compositions and methods for treatment of infection caused by Haemophilus influenzae and Streptococcus pneumoniae
US5994090A (en) * 1994-06-03 1999-11-30 Seikagaku Corporation Glycoglycerophospholipid, antibody thereagainst, and method for detecting mycoplasma
US6111079A (en) * 1995-06-05 2000-08-29 Bionebraska, Inc. Lead binding polypeptides and nucleotides coding therefore
US6610293B1 (en) * 1997-06-16 2003-08-26 The Henry M. Jackson Foundation For The Advancement Of Military Medicine Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
US20030190705A1 (en) * 2001-10-29 2003-10-09 Sunol Molecular Corporation Method of humanizing immune system molecules
US6790661B1 (en) * 1999-07-16 2004-09-14 Verax Biomedical, Inc. System for detecting bacteria in blood, blood products, and fluids of tissues
US7250494B2 (en) * 1998-06-15 2007-07-31 Biosynexus Incorporated Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram positive bacteria
US20070286866A1 (en) * 2003-11-10 2007-12-13 The Uab Research Foundation Compositions For Reducing Bacterial Carriage And Cnc Invasion And Methods Of Using Same
US20110104168A1 (en) * 2007-05-23 2011-05-05 The Uab Research Foundation Detoxified pneumococcal neuraminidase and uses thereof

Family Cites Families (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1499035A (en) * 1975-04-10 1978-01-25 Ts Nii Gematologii I Perelivan Antistaphylococcus human immune globulin and method of preparing same
JPS5452794A (en) * 1977-09-30 1979-04-25 Kousaku Yoshida Extracting of polysacchride from capusle containing epidermis staphylococus
US4732757A (en) * 1978-02-06 1988-03-22 Stolle Research And Development Corporation Prevention and treatment of rheumatoid arthritis
FR2475900A1 (en) * 1980-02-20 1981-08-21 Fabre Sa Pierre VACCINE COMPLEX CONTAINING A SPECIFIC ANTIGEN AND VACCINE CONTAINING SAME
US4425330A (en) * 1981-05-20 1984-01-10 Cornell Research Foundation, Inc. Bovine mastitis vaccine and method for detecting efficacy thereof
US4482483A (en) * 1983-04-06 1984-11-13 Armour Pharmceutical Company Composition of intravenous immune globulin
US4719290A (en) * 1983-09-02 1988-01-12 Armour Pharmaceutical Corporation Composition of intravenous immune globulin
NZ214503A (en) * 1984-12-20 1990-02-26 Merck & Co Inc Covalently-modified neutral bacterial polysaccharides, stable covalent conjugates of such polysaccharides and immunogenic proteins, and methods of preparing such polysaccharides and conjugates
FR2581877B1 (en) * 1985-05-14 1987-12-18 Louvain Universite Catholique CONJUGATE CONSISTING OF A WALL ADHESIN OF S. MUTANS, OF PROTEIN NATURE AND OF A POLYSACCHARIDE OF S. MUTANS, ITS PREPARATION AND ITS USE IN PARTICULAR IN CARIES VACCINES
CA1341235C (en) 1987-07-24 2001-05-22 Randy R. Robinson Modular assembly of antibody genes, antibodies prepared thereby and use
FR2619122B1 (en) * 1987-08-03 1990-03-09 Pasteur Institut PROCESS FOR OBTAINING CAPSULAR POLYOSIDES OF STAPHYLOCOCCS, POLYOSIDES OBTAINED, APPLICATIONS OF SUCH POLYOSIDES AND STRAINS FOR IMPLEMENTING THE PROCESS
US5034515A (en) * 1987-09-22 1991-07-23 Wisconsin Alumni Research Foundation Staphylococcal fibronectin receptor, monoclonal antibodies thereto and methods of use
US5055455A (en) * 1988-09-28 1991-10-08 Brigham And Women's Hospital Capsular polysaccharide adhesin antigen, preparation, purification and use
SE8901687D0 (en) * 1989-05-11 1989-05-11 Alfa Laval Agri Int FIBRONECTIN BINDING PROTEIN AS WELL AS IT'S PREPARATION
US5440014A (en) * 1990-08-10 1995-08-08 H+E,Uml/Oo/ K; Magnus Fibronectin binding peptide
US5153312A (en) * 1990-09-28 1992-10-06 American Cyanamid Company Oligosaccharide conjugate vaccines
US6632432B1 (en) * 1990-10-22 2003-10-14 Henry M. Jackson Foundation For The Advancement Of Military Medicine Directed human immune globulin for the prevention and treatment of staphylococcal infections
WO1993017044A1 (en) 1992-02-25 1993-09-02 U.S. Government, As Represented By The Secretary Of The Army Directed human immune globulin for the prevention and treatment of staphylococcal infections
ATE197506T1 (en) 1992-03-19 2000-11-11 Jackson H M Found Military Med BROADLY REACTIVE OPSONIC ANTIBODIES THAT REACT WITH COMMON ANTIGENS OF STAPHYLOCOCCUS
US5545721A (en) * 1992-12-21 1996-08-13 Ophidian Pharmaceuticals, Inc. Conjugates for the prevention and treatment of sepsis
JPH08508240A (en) * 1993-01-12 1996-09-03 ジョージ グリスティーナ,アンソニー Methods and compositions for direct transfer of passive immunity
US5955078A (en) * 1993-02-05 1999-09-21 Smithkline Beecham P.L.C. Fibronectin binding protein polypeptides
US5354654A (en) * 1993-07-16 1994-10-11 The United States Of America As Represented By The Secretary Of The Navy Lyophilized ligand-receptor complexes for assays and sensors
US5624904A (en) * 1993-11-17 1997-04-29 Massachusetts Institute Of Technology Method for treating gram positive septicemia
US5686600A (en) * 1994-06-28 1997-11-11 Novartis Finance Corporation Antibodies which bind to insect gut proteins and their use
US5538733A (en) * 1994-07-07 1996-07-23 Willmar Poultry Company, Inc. Method of priming an immune response in a one-day old animal
CA2200691C (en) 1994-09-21 2011-02-15 Henry M. Jackson Foundation For The Advancement Of Military Medicine Broadly reactive opsonic antibodies reactive with common staphylococcal antigens
FR2729960A1 (en) 1995-01-30 1996-08-02 Bio Merieux TOXOPLASMA GONDII MIMOTOPE POLYPEPTIDES AND APPLICATIONS
WO1996039518A1 (en) 1995-06-05 1996-12-12 Bionebraska, Inc. Lead binding polypeptides and nucleotides coding therefor
MA24512A1 (en) * 1996-01-17 1998-12-31 Univ Vermont And State Agrienl PROCESS FOR THE PREPARATION OF ANTICOAGULATING AGENTS USEFUL IN THE TREATMENT OF THROMBOSIS
US5770208A (en) * 1996-09-11 1998-06-23 Nabi Staphylococcus aureus B-linked hexosamine antigen
CN1189483C (en) 2001-11-23 2005-02-16 上海中信国健药业有限公司 Humanized CD3-resisting monoclonal antibody
US20040052779A1 (en) 2001-12-21 2004-03-18 Stinson Jeffrey R. Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram positive bacteria

Patent Citations (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4196192A (en) * 1977-10-28 1980-04-01 American Cyanamid Company Combined Haemophilus influenzae type b and pertussis vaccine
US4451570A (en) * 1981-03-26 1984-05-29 The Regents Of The University Of California Immunoglobulin-secreting human hybridomas from a cultured human lymphoblastoid cell line
US4744982A (en) * 1982-08-24 1988-05-17 Hunter Kenneth W Human monoclonal antibody reactive with polyribosylribitol phosphate
US4954449A (en) * 1982-08-24 1990-09-04 The United States Of America As Represented By The Secretary Of The Army Human monoclonal antibody reactive with polyribosylribitol phosphate
US4761283A (en) * 1983-07-05 1988-08-02 The University Of Rochester Immunogenic conjugates
US4596769A (en) * 1984-03-05 1986-06-24 Temple University Monoclonal antibodies to peptidoglycan and methods of preparing same
US5043267A (en) * 1984-05-18 1991-08-27 E. I. Du Pont De Nemours And Company Method for rapid detection of bacterial and fungal infection
US4744892A (en) * 1985-02-27 1988-05-17 Henkel Kommanditgesellschaft Auf Aktien Process for dressing kaolinite by flotation
US5139933A (en) * 1989-09-26 1992-08-18 Vicam, L.P. Assay method for detecting listeria
US5475100A (en) * 1990-07-13 1995-12-12 Fujita Health University Artificial antibody
US5571511A (en) * 1990-10-22 1996-11-05 The U.S. Government Broadly reactive opsonic antibodies that react with common staphylococcal antigens
US5866140A (en) * 1991-11-22 1999-02-02 Nabi Type I surface antigen associated with staphylococcus epidermidis
US5961975A (en) * 1991-11-22 1999-10-05 Nabi Type I surface antigen associated with staphylococcus epidermidis
US5776456A (en) * 1992-11-13 1998-07-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5994090A (en) * 1994-06-03 1999-11-30 Seikagaku Corporation Glycoglycerophospholipid, antibody thereagainst, and method for detecting mycoplasma
US5804370A (en) * 1994-06-08 1998-09-08 Critichem Medical Products Limited Early diagnosis of sepsis utilizing antigen-antibody interactions amplified by whole blood chemiluminescence
US5807688A (en) * 1995-05-27 1998-09-15 Zeneca Limited Catalytic antibodies for carbamate activation by a non-spontaneous reaction mechanism
US6111079A (en) * 1995-06-05 2000-08-29 Bionebraska, Inc. Lead binding polypeptides and nucleotides coding therefore
US5871951A (en) * 1996-09-23 1999-02-16 The Children's Hospital Of Philadelphia Compositions and methods for treatment of infection caused by Haemophilus influenzae and Streptococcus pneumoniae
US6610293B1 (en) * 1997-06-16 2003-08-26 The Henry M. Jackson Foundation For The Advancement Of Military Medicine Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
US7511122B2 (en) * 1997-06-16 2009-03-31 Henry M. Jackson Foundation For The Advancement Of Military Medicine Humanized and chimeric antibodies specific for lipoteichoic acid of GRAM positive bacteria
US6939543B2 (en) * 1997-06-16 2005-09-06 Henry M. Jackson Foundation For The Advancement Of Military Medicine Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
US8372958B2 (en) * 1997-06-16 2013-02-12 Biosynexus Incorporated; Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram posiive bacteria
US7250494B2 (en) * 1998-06-15 2007-07-31 Biosynexus Incorporated Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram positive bacteria
US6790661B1 (en) * 1999-07-16 2004-09-14 Verax Biomedical, Inc. System for detecting bacteria in blood, blood products, and fluids of tissues
US20030190705A1 (en) * 2001-10-29 2003-10-09 Sunol Molecular Corporation Method of humanizing immune system molecules
US20070286866A1 (en) * 2003-11-10 2007-12-13 The Uab Research Foundation Compositions For Reducing Bacterial Carriage And Cnc Invasion And Methods Of Using Same
US20110104168A1 (en) * 2007-05-23 2011-05-05 The Uab Research Foundation Detoxified pneumococcal neuraminidase and uses thereof

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
Argaman, M et al, The Journal of Immunology, vol. 112(2), Feb. 1974, pages 649-655. *
Briles, DE et al, Infection and Immunity, May 1989, vol. 57(5), pgaes 1457-1464, Antipneumococcal effects of C-reactive protein and monoclonal antibodies to pneumococcal cell wall and capsular antigens. *
Briles, EB et al, The Journal of Biological Chemistry, vol. 248(18), September 25, 1973, pages 6394-6397, Pneumococcal Forssman Antigen. *
Crisel, RM et al, The Journal of Biological Chemistry, Vol. 250(13) July 10, 1975, pages 4926-4930. *
Fischer, W et al, European Journal of Biochemistry, vol. 215, pages 851-857, 1993, Teichoic acid and lipoteichoic acid of Streptococcus pneumoniae posses identical chain structures; A reinvestigation of teichoic acid (C polysaccharide). *
Hamada, H et al, Cancer Research, 1990, vol. 50, pages 3167-3171, Mouse-human chimeric antibody against the multidrug transporter P-glycoprotein, see page 3167 col. 2, p. 2, "anti-phophorylcholine mouse-human chimeric antibody" *
Hamada, Shigeyuki et al, Microbiological Immunology, vol. 28(9), pages 1009-1021, 1984, Characterization of a monoclonal antibody specific for lipoteichoic acid from various Gram Positive bacteria. *
Heidrun, I et al, Journal of Clincial Microbiology, vol. 27(6), pages 1286-1291, June 1989, Antibodies to Staphylococcus petidoglycan and Its peptide epitopes, teichoic acid and lipoteichoic acid in sera from blood donors and patients with Staphylococcal Infections. *
Kameyama, K et al, Feb Letters, vol. 244, (2), pages 301-306, February 1989, Covenient plasmid vectos for construction of chimeric mouse/human antibodies, specific for hapten phosphorylcholine (PC), page 301, col. 2, p. 1. *
Kelly-Quintos, Casie et al, Infection and Immunity, May 2006, vol. 74(5), pages 2742-2750, Characterization of the opsonic and protective activity against Staphylococcus aureus of Fully Human Monoclonal Antibodies specific for the bacterial surface polysaccharide Poly-N-Acetylglucosamine. *
Nowinski, R et al, , Science, vol. 210, October 31, 1080, pages 537-539, Human monoclonal antibody against Forssman Antigen. *
Wicken, AJ et al, Infection and Immunity, 1972, vol. 5(6), pages 982-984, Anti-teichoic acid antibodies and non-trponemal serological tests for syphilis. *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10364298B2 (en) 2011-11-18 2019-07-30 National Research Council Of Canada Clostridium difficile lipoteichoic acid and uses thereof
WO2022087122A1 (en) * 2020-10-20 2022-04-28 Longhorn Vaccines And Diagnostics, Llc Immunogenic antigens
GB2615470A (en) * 2020-10-20 2023-08-09 Longhorn Vaccines & Diagnostics Llc Immunogenic antigens
IE20210235A1 (en) * 2020-10-20 2023-08-16 Longhorn Vaccines & Diagnostics Llc Immunogenic antigens

Also Published As

Publication number Publication date
US20080019976A1 (en) 2008-01-24
US7777017B2 (en) 2010-08-17
US20030235578A1 (en) 2003-12-25
US7250494B2 (en) 2007-07-31

Similar Documents

Publication Publication Date Title
US7777017B2 (en) Nucleic acids encoding opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
AU2009202762B2 (en) Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram positive bacteria
US7511122B2 (en) Humanized and chimeric antibodies specific for lipoteichoic acid of GRAM positive bacteria
US20080014202A1 (en) Multifunctional monoclonal antibodies directed to peptidoglycan of gram-positive bacteria
AU2009210372B2 (en) Multifunctional monoclonal antibodies directed to peptidoglycan of gram-positive bacteria
JP6371758B2 (en) Anti-staphylococcal antibody, method for producing the same and use thereof
JP2005514053A6 (en) Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of Gram-positive bacteria
AU2012254925B2 (en) Opsonic monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
US10100103B2 (en) Opsonic and protective monoclonal antibodies against gram-positive pathogens
AU2012201369A1 (en) Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria
AU2006203322A1 (en) Opsonic and protective monoclonal and chimeric antibodies specific for lipoteichoic acid of gram positive bacteria

Legal Events

Date Code Title Description
AS Assignment

Owner name: ALTOR BIOSCIENCE CORPORATION, FLORIDA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WONG, HING;STINSON, JEFFREY R.;SIGNING DATES FROM 20100503 TO 20100508;REEL/FRAME:024435/0023

Owner name: THE HENRY M. JACKSON FOUNDATION FOR THE ADVANCEMEN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FISCHER, GERALD;SCHUMAN, RICHARD;SIGNING DATES FROM 20100409 TO 20100412;REEL/FRAME:024435/0107

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE