WO2006138647A2 - Procede de fabrication d'anticorps humains de recombinaison destines a etre utilises dans la technologie des biocapteurs - Google Patents

Procede de fabrication d'anticorps humains de recombinaison destines a etre utilises dans la technologie des biocapteurs Download PDF

Info

Publication number
WO2006138647A2
WO2006138647A2 PCT/US2006/023622 US2006023622W WO2006138647A2 WO 2006138647 A2 WO2006138647 A2 WO 2006138647A2 US 2006023622 W US2006023622 W US 2006023622W WO 2006138647 A2 WO2006138647 A2 WO 2006138647A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibodies
biosensor
antibody
interest
agents
Prior art date
Application number
PCT/US2006/023622
Other languages
English (en)
Other versions
WO2006138647A3 (fr
Inventor
Gregory Liposky
Byron V. Olsen
Original Assignee
Gtc Biotherapeutics, 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 US11/156,831 external-priority patent/US20060286548A1/en
Application filed by Gtc Biotherapeutics, Inc. filed Critical Gtc Biotherapeutics, Inc.
Publication of WO2006138647A2 publication Critical patent/WO2006138647A2/fr
Publication of WO2006138647A3 publication Critical patent/WO2006138647A3/fr

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6842Proteomic analysis of subsets of protein mixtures with reduced complexity, e.g. membrane proteins, phosphoproteins, organelle proteins
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to the production and use of recombinant human antibodies to detect the presence of certain molecules of interest including those that may case specific disease related pathologies.
  • the current invention provides for the production of human antibodies of interest for use in various biosensor detection apparati.
  • the present invention relates generally to the field of the production of antibodies and the methods of using these antibodies for biosensor applications.
  • those antibodies are produced from the milk of transgenic animals. More particularly, it concerns improved methods for generating large quantities of transgenic antibodies capable of detecting certain pathogenic agents of interest and thereafter reporting such an interaction.
  • Bio pathogens for example derived from anthrax bacteria or the hanta virus, respectively
  • toxins produced by biological organisms for example, botulinus toxin
  • These same biotoxins which can occur naturally, can result in disease, fear, disruption to society, significant economic harm and potentially large-scale loss of life.
  • Individuals can be exposed to agents of this sort from inhalation, skin contact, or by the ingestion of contaminated food or water. After exposure to a pathogen or toxin, physical symptoms can be delayed and prove difficult to distinguish from naturally occurring illnesses as well as shielding the location or time of exposure.
  • the present invention relates generally to the production and purification of antibodies or modified antibodies that make them available for the detection of pathogenic agents of interest and/or chemical agents.
  • Uses for recombinant biosensor antibodies include use for the detection of known Weapons of Mass Destruction ("WMD”) agents in particular biological agents including: anthrax, smallpox, botulism, ebola, hanta virus etc.
  • WMD Weapons of Mass Destruction
  • An objective of the current invention is to provide an antibody array that can be used to detect antibody expression profiles.
  • several antibodies can be simultaneously examined with an array comprising a large number of immobilized antibodies put down in a pre-determined order. This then allows for multiple antibodies to recognize their corresponding antigens independently without cross reactions. When associated with known reporter mechanisms this interaction then allows for output data to indicate the type and breadth of a given biological agent/chemical agent threat.
  • An objective of the current invention is to provide an antibody array of the invention can also be used to detect antibody—protein interactions.
  • We provide here the use of an antibody array in detection and identification of proteins that interact with specific proteins of interest.
  • FIG. 1 Shows a Flowchart of an Embodiment of the Process of Creating Cloned Animals through Nuclear Transfer.
  • FIG. 2 Shows a Simple Schematic Representation of Biosensor Antibody with two identical antibody antigen/analyte binding sites. It is composed of four polypeptide chains— two identical heavy chains and two identical light chains. The two antigen-binding sites are identical, each formed by the N-terminal region of a light chain and the N-terminal region of a heavy chain. Both the tail (Fc) and hinge region are formed by the two heavy chains.
  • FIG. 3 Shows the Construction of a cDNA Vector for Biosensor
  • FIG. 4 Shows a Process for the Purification of Antibodies of Interest from a Milk Feedstream.
  • FIG. 5 Shows Embodiment of potential Biosensor Antibody Array of the Invention.
  • FIG. 6 Shows known and efficient peptide coupling reagents for solid support attachment of biosensor antibodies.
  • PCR Polymerase Chain Reaction
  • MFI Mean Fluorescence Intensity
  • MDL Minimum Detection Limits
  • ABSORPTION The process of an agent being drawn into a media through a surface (clothing, fabric, wood, etc.), much like a sponge and water.
  • ACETYLCHOLINE A chemical compound formed from an acid and an alcohol, which causes muscles to contract and acts as a neurotransmitter. It is found in various organs and tissues of the body. It is rapidly broken down by an enzyme, cholinesterase.
  • ACETYLCHOLINESTERASE An enzyme which inhibits the action of acetylcholine by breaking acetylcholine into its component parts. Nerve agents combine with acetylcholinesterase to prevent it from performing its inactivation of acetylcholine.
  • ADSORPTION The process of an agent sticking to or becoming chemically attached to a surface.
  • AEROSOLS A suspension or dispersion of small particles (solids or liquids) in a gaseous medium.
  • ANALYTE Compound which is to be measured in an assay or biosensor application.
  • ANTIBODY AGENTS Refers to antibodies, recombinant antibodies, calin, synthesized antibody fragments or single chain antibodies immobilized on the solid support of an antibody array.
  • ANTICHOLINERGIC An agent or chemical that blocks or impedes the action of acetylcholine, such as the (also cholinolytic) antidote atropine.
  • ANTI-CHOLINESTERASE A substance that blocks the action of cholinesterase (acetylcholinesterase) such as nerve agents.
  • ANTIDOTE A substance that neutralizes toxic agents or their effects.
  • ATROPINE An anticholinergic used as an antidote for nerve agents to counteract excessive amounts of acetylcholine. It also has other medicinal uses.
  • BIOLOGICAL FLUID An aqueous solution produced by an organism, such as a mammal, bird, amphibian, or reptile, which contains antibodies that are secreted by cells that are bathed in the aqueous solution. Examples include: milk, urine, saliva, seminal fluid, vaginal fluid, synovial fluid, lymph fluid, amniotic fluid, blood, sweat, and tears; as well as an aqueous solution produced by a plant, including, for example, exudates and guttation fluid, xylem, phloem, resin, and nectar.
  • BLISTER AGENT A chemical warfare agent which produces local irritation and damage to the skin (vesicant) and mucous membranes, pain and injury to the eyes, reddening and blistering of the skin, and when inhaled, damage to the respiratory tract.
  • BLOOD AGENT A chemical warfare agent that is inhaled and absorbed into the blood.
  • the blood carries the agent to all body tissues where it interferes with the tissue oxygenation process (ex: potassium cyanide).
  • CHEMICAL AGENT Any chemical substance which is intended for use in military operations to kill, seriously injure, or incapacitate humans because of its physiological effects.
  • CHEMICAL WARFARE AGENT A chemical substance, which, because of its physiological, psychological, or pharmacological effects, is intended for use in military operations to kill, seriously injure, or incapacitate humans (or animals) through its toxicological effects. Excluded are riot control agents, chemical herbicides, and smoke and flame agents.
  • CHOKING AGENTS These agents exert their effects solely on the lungs and result in the irritation of the alveoli of the lungs. Agents cause the alveoli to constantly secrete watery fluid into the air sacs, which is called pulmonary edema. When a lethal amount of a choking agent is received, the air sacs become so flooded that the air cannot enter and the victim dies of anoxia (oxygen deficiency); also known as a dry land drowning.
  • CONCENTRATION The amount of a chemical agent present in a unit volume of air, usually expressed in milligrams per cubic meter (mg/m3).
  • DILUTION FACTOR Dilution of contaminated air with uncontaminated air in a general area, room or building for the purposes of health hazard or nuisance control, and/or for heating and cooling.
  • DOSAGE The concentration of a chemical agent in the atmosphere (C) multiplied by the time (t) the concentration remains, expressed as mg-min/m.
  • the dosage (Ct) received by a person depends upon how long he is exposed to the concentration. That is, the respiratory dosage in mg-min//m 3 is equal to the time in minutes as individual is unmasked in an agent cloud multiplied by the concentration of the cloud.
  • ENZYME CONJUGATE A synthetic compound produced by linking an enzyme to an antibody or part of an antibody which recognizes another molecule (e.g. antibody or antigen), to the animal in which it was raised.
  • ENCODING refers generally to the sequence information being present in a translatable form, usually operably linked to a promoter (e.g., a beta-casein or beta-lacto globulin promoter).
  • a sequence is operably linked to a promoter when the functional promoter enhances transcription or expression of that sequence.
  • An anti-sense strand is considered to also encode the sequence, since the same informational content is present in a readily accessible form, especially when linked to a sequence which promotes expression of the sense strand.
  • the information is convertible using the standard, or a modified, genetic code.
  • EPITOPE A region of an antigenic molecule which acts as a recognition site for an antibody.
  • An epitope consists of a minimum of three amino acids but most stable epitopes are often between 5-7 amino acids.
  • EXPRESSION VECTOR A genetically engineered plasmid or virus, derived from, for example, a bacteriophage, adenovirus, retrovirus, poxvirus, herpesvirus, or artificial chromosome, that is used to transfer transgenic antibody coding sequence, operably linked to a promoter, into a host cell, such that the encoded recombinant transgenic antibody is expressed within the host cell.
  • FUNCTIONAL ANTIBODIES Antibodies which have a biological or other activity or use, similar to that seen when produced endogenously.
  • INCAPACITATING AGENT An agent that produces physiological or mental effects, or both, that may persist for hours or days after exposure, rendering an individual incapable of performing his or her assigned duties.
  • the leader sequence may be the native human leader sequence, an artificially-derived leader, or may obtained from the same gene as the promoter used to direct transcription of the transgene coding sequence, or from another antibody that is normally secreted from a cell.
  • LETHAL CHEMICAL AGENT An agent that may be used effectively in a field concentration to produce death.
  • LIQUID DOSAGE The weight of a liquid agent received by a person on his skin is usually expressed as dosage in milligrams of contaminant per kilogram of body weight (mg/kg). This is equivalent to parts per million (ppm).
  • MILK-PRODUCING CELL A cell (e.g., a mammary epithelial cell) that secretes an antibody into milk.
  • MILK-SPECIFIC PROMOTER A promoter that naturally directs expression of a gene in a cell that secretes a antibody into milk (e.g., a mammary epithelial cell) and includes, for example, the casein promoters, e.g., ⁇ -casein promoter (e.g., alpha
  • ⁇ -casein promoter e.g., the goat beta casein gene promoter (DiTullio, BIOTECHNOLOGY 10:74-77, 1992), ⁇ - casein promoter, and ⁇ -casein promoter; the whey acidic antibody (WAP) promoter (Gorton et al., BIOTECHNOLOGY 5: 1183-1187, 1987); the ⁇ - lactoglobulin promoter (Clark et al., BIOTECHNOLOGY 7: 487-492, 1989); and the ⁇ -lactalbumin promoter (Soulier et al., FEBS LETTS. 297:13, 1992).
  • WAP whey acidic antibody
  • promoters that are specifically activated in mammary tissue and are thus useful in accordance with this invention, for example, the long terminal repeat (LTR) promoter of the mouse mammary tumor virus (MMTV).
  • LTR long terminal repeat
  • NERVE AGENTS Agents that effect the transmission of nerve impulses by reacting with the enzyme cholinesterase, permitting an accumulation of acetylcholine and
  • NUCLEAR TRANSFER This refers to a method of cloning wherein the nucleus from a donor cell is transplanted into an enucleated oocyte.
  • OPERABLY LINKED A gene and one or more regulatory sequences are connected in such a way as to permit gene expression when the appropriate molecules (e.g., transcriptional activator antibodies) are bound to the regulatory sequences.
  • ORGANOPHOSPHATE A compound with a specific phosphate group which inhibits acetylcholinesterase. Organophosphates are used in chemical warfare and as an insecticide.
  • PERSISTENT AGENT An agent that remains in the target area for longer periods of time. Hazards from both vapor and liquid may exist for hours, days, or in exceptional cases, weeks or months after dissemination of the agent. As a general rule, persistent agents duration will be greater than 12 hours.
  • POLYPEPTIDE A polymer whose monomer linkages are usually but not necessarily peptide bonds, and whose side groups are usually but not necessarily selected from the naturally occurring amino acid side groups such as hydrogen (glycine), methyl (alanine), etc.. Numerous unnatural polypeptide derivations, with modifications to both the amine linkages and side groups are known. Relatively small polypeptides are often termed peptides.
  • SOLUBILITY The ability of a material to dissolve in water or another liquid.
  • TEAR AGENTS Compounds that cause a large flow of tears and intense eye pain and irritation of the skin.
  • TOXICITY The property a material possesses which enables it to injure the physiological mechanism of an organism by chemical means, with the maximum effect being incapacitation or death.
  • TRANSGENE Any piece of a nucleic acid molecule that is inserted by artifice into a cell, or an ancestor thereof, and becomes part of the genome of the animal which develops from that cell.
  • a transgene may include a gene which is partly or entirely exogenous (i.e., foreign) to the transgenic animal, or may represent a gene having identity to an endogenous gene of the animal.
  • TRANSGENIC ORGANISM An organism into which genetic material from another organism has been experimentally transferred, so that the host acquires the genetic information of the transferred genes in its chromosomes in addition to that already in its genetic complement.
  • UNGULATE Relating to a hoofed typically herbivorous quadruped mammal, including, without limitation, sheep, swine, goats, cattle and horses.
  • VECTOR Means a plasmid, a phage DNA, or other DNA sequence that (1) is able to replicate in a host cell, (2) is able to transform a host cell, and (3) contains a marker suitable for identifying transformed cells.
  • VOLATILITY With chemical agents, it refers to their ability to change from a liquid state into a gaseous state. That is, the ability of a material to evaporate.
  • VOMITING AGENT Compounds that cause irritation of the upper respiratory tract and involuntary vomiting.
  • the process comprises expressing in the milk of a transgenic non-human placental mammal one or more transgenic antibodies or antibody fragments useful for various biosensor applications.
  • the current invention pertains to the production of antibodies.
  • the antibodies used for biosensor array applications are produced in the milk of a transgenic mammal, though other well-known methods exist in the prior art. These other methods can also be used for the production of antibodies for use in the methods of the current invention.
  • Various aspects of the invention are also known in the prior art in regards to the methods of producing an antibody or fragments thereof. These fragments can also be preferably be produced in the milk of a transgenic mammal.
  • Methods of producing a transgenic mammal whose somatic and germ cells include a modified antibody coding sequence are also known in the prior art. Use of nucleic acid sequences for expression for a variety of modified antibodies are also provided by the prior art.
  • a "class" of antibodies refers to the five major isotypes of antibodies, including IgA, IgD, IgE, IgG 5 and IgM.
  • a "subclass” of antibodies refers to the a subclassification of a given class of antibodies based on amino acid differences among members of the class, e.g., the class of antibodies designated IgG can be divided into the subclasses of, e.g., IgGl, IgG2, IgG3, and IgG4, and the class of antibodies designated as IgA can be divided into the subclasses of IgAl and IgA2.
  • antibody refers to a protein comprising at least one, and preferably two, heavy (H) chain variable regions (abbreviated herein as VH), at least one and preferably two light (L) chain variable regions (abbreviated herein as VL), and at least one, preferably two heavy chain constant regions.
  • VH and VL regions can be further subdivided into regions of hypervariability, termed “complementarity determining regions” (“CDR"), interspersed with regions that are more conserved, termed “framework regions” (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • the antibodies of the invention can further include a light chain constant region, to thereby form a heavy and light immunoglobulin chains.
  • the antibody is a tetramer of two heavy immunoglobulin chains and two light immunoglobulin chains, wherein the heavy and light immunoglobulin chains are interconnected by, e.g., disulfide bonds.
  • the heavy chain constant region is comprised of three domains, CHl, CH2 and CH3.
  • the light chain constant region is comprised of one domain, CL.
  • the variable region of the heavy and light chains contains a binding domain that interacts with an antigen.
  • the constant regions of the antibodies typically mediate the binding of the antibody to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (CIq) of the classical complement system.
  • the antibodies of the invention can further include a hinge region, described in further detail below.
  • an "assembled" antibody is an antibody in which the heavy chains are associated with each other, e.g., interconnected by disulfide bonds.
  • Each heavy chain hinge region includes at least one, and often several, cysteine residues.
  • the cysteine residues in the heavy chains are aligned so that disulfide bonds can be formed between the cysteine residues in the hinge regions covalently bonding the two heavy-light chain heterodimers together.
  • fully assembled antibodies are bivalent in that they have two antigen binding sites.
  • antibody also refers to fragments of a full-length antibody, such as, e.g., a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies.
  • an "antigen-binding fragment" of an antibody refers to one or more portions of an antibody that retain the ability to specifically bind to an antigen.
  • binding fragments encompassed within the term “antigen-binding fragment” of an antibody include one or more complementarities determining region (CDR).
  • a "chimeric antibody heavy chain” refers to those antibody heavy chains having a portion of the antibody heavy chain, e.g., the variable
  • the heavy chain variable region has a sequence substantially identical to the heavy chain variable region of an antibody from one species (e.g., a "donor" antibody, e.g., a rodent antibody), while the constant region is substantially identical to the constant region of another species antibody (e.g., an "acceptor” antibody, e.g., a human antibody).
  • the donor antibody can be an in vitro generated antibody, e.g., an antibody generated by phage display.
  • humanized or “CDR-grafted” light chain variable region refers to an antibody light chain comprising one or more CDR's, or having an amino acid sequence which differs by no more than 1 or 2 amino acid residues to a corresponding one or more CDR's from one species, or antibody class or type, e.g., a "donor” antibody (e.g., a non-human (usually a mouse or rat) immunoglobulin, or an in vitro generated immunoglobulin); and a framework region having an amino acid sequence about 85% or higher, preferably 90%, 95%, 99% or higher identical to a corresponding part of an acceptor antibody framework from a different species, or antibody class or type, e.g., a naturally-occurring immunoglobulin framework (e.g., a human framework) or a consensus framework.
  • a donor antibody e.g., a non-human (usually a mouse or rat) immunoglobulin, or an in vitro generated immunoglobulin
  • the framework region includes at least about 60, and more preferably about 70 amino acid residues identical to those in the acceptor antibody light chain variable region framework, e.g., a naturally-occurring antibody framework (e.g., a human framework) or a consensus framework.
  • acceptor antibody light chain variable region framework e.g., a naturally-occurring antibody framework (e.g., a human framework) or a consensus framework.
  • a “heterologous antibody” or “exogenous antibody” is an antibody that normally is not produced by the mammal, or is not normally produced in the mammary gland (e.g., an antibody only present in serum), or is produced in the mammary gland but the level of expression is augmented or enhanced in its production.
  • any of the antibodies described herein that is, chimeric, humanized or human antibodies, can include further modifications to their sequence. In this sense the sequences can be modified by addition, deletion or substitution, including a conservative substitution.
  • the DNA constructs used to make the nucleic acid sequences of the antibodies of the invention will preferably employ one insulator sequence during their production.
  • the terms "insulator”, “insulator sequence” and “insulator element” are used interchangeably herein.
  • An insulator element is a control element which insulates the transcription of genes placed within its range of action but which does not perturb gene expression, either negatively or positively.
  • an insulator sequence is inserted on either side of the DNA sequence to be transcribed.
  • the insulator can be positioned about 200 bp to about 1 kb, 5' from the promoter, and at least about 1 kb to 5 kb from the promoter, at the 3' end of the gene of interest.
  • the distance of the insulator sequence from the promoter and the 3' end of the gene of interest can be determined by those skilled in the art, depending on the relative sizes of the gene of interest, the promoter and the enhancer used in the construct.
  • more than one insulator sequence can be positioned 5' from the promoter or at the 3' end of the transgene.
  • two or more insulator sequences can be positioned 5' from the promoter.
  • the insulator or insulators at the 3' end of the transgene can be positioned at the 3' end of the gene of interest, or at the 3'end of a 3' regulatory sequence, e.g., a 3' untranslated region (UTR) or a 3' flanking sequence.
  • UTR 3' untranslated region
  • a preferred insulator is a DNA segment which encompasses the 5' end of the chicken ⁇ -globin locus and corresponds to the chicken 5' constitutive hypersensitive site as described in PCT Publication 94/23046, the contents of which is incorporated herein by reference.
  • a preparation refers to two or more antibody molecules.
  • the antibody molecules of interest can be produced by one or more than one transgenic animal.
  • Certain embodiments of the invention can also provide antibodies or kallikrines that vary in glycosylation profile for what is seen in vivo.
  • a “purified preparation”, “substantially pure preparation of antibodies”, or “isolated antibodies as used herein, refers to an antibody that is substantially free of material with which it occurs in the milk of a transgenic mammal.
  • the antibody is also preferably separated from substances, e.g., gel matrix, e.g., polyacrylamide, which is used to purify it.
  • the language “substantially free” includes preparations of an antibody having less than about 30% (by dry weight) of non-antibody
  • Non-antibody 16 material also referred to herein as a "milk impurity" or "milk component", more preferably less than about 20% of non-antibody material, still more preferably less than about 10% of non-antibody material, and most preferably less than about 5% non- antibody material.
  • Non-antibody material includes casein, lipids (e.g., soluble lipids and phospholipids), lactose and other small molecules (e.g., glucose, galactose), small peptides (e.g., microbial peptides and anti-microbial peptides) and other milk proteins (e.g., whey proteins such as ⁇ -lactoglobulin and ⁇ -lactalbumin, lactoferrin, and serum albumin).
  • lipids e.g., soluble lipids and phospholipids
  • lactose and other small molecules e.g., glucose, galactose
  • small peptides e.g., m
  • the antibodies preferably constitute at least 10, 20, 50, 70, 80 or 95% dry weight of the purified preparation.
  • the preparation contains: at least 1, 10, or 100 ⁇ g of the antibodies; at least 1, 10, or 100 mg of the antibodies.
  • the purified preparation preferably contains about 70%, 75%, 80%, 85%, 90%, 95%, 98% assembled antibodies.
  • Antibodies can be isolated from milk using standard protein purification methods known in the art. For example, the methods of Kutzko et al. (U.S. Patent No. 6,268,487) can be utilized to purify antibodies and/or fragments of the present invention.
  • Milk proteins, or according to the current invention - antibodies of interest, must often be purified by a combination of processes.
  • raw milk can first be fractionated to remove fats, for example, by skimming, centrifugation, sedimentation (H. E. Swaisgood, Developments in Dairy Chemistry, in: CHEMISTRY OF MILK PROTEIN, Applied Science Publishers, NY, 1982), acid precipitation (U.S. Pat. No.# 4,644,056) or enzymatic coagulation with rennin or chymotrypsin.
  • the major milk proteins may be fractionated into either a clear solution or a bulk precipitate from which the specific protein of interest may be readily purified.
  • French Patent NoJ 2,487,642 describes the isolation of milk proteins from skim milk or whey by membrane ultrafiltration in combination with exclusion chromatography or ion exchange chromatography. Whey is first produced by removing the casein by coagulation with rennet or lactic acid.
  • U.S. Pat. NoJ 4,485,040 describes the isolation of an alpha-lactoglobulin-enriched product in the retentate from whey by two sequential ultrafiltration steps.
  • 4,644,056 provides a method for purifying immunoglobulin from milk or colostrum by acid precipitation at pH 4.0-5.5, and sequential cross-flow filtration first on a membrane with 0.1-1.2 micrometer pore size to clarify the product pool and then on a membrane with a separation limit of 5-80 kd to
  • U.S. Pat. No.# 4,897,465 teaches the concentration and enrichment of a protein such as immunoglobulin from blood serum, egg yolks or whey by sequential ultrafiltration on metallic oxide membranes with a pH shift. Filtration is carried out first at a pH below the isoelectric point (pi) of the selected protein to remove bulk contaminants from the protein retentate, and next at a pH above the pi of the selected protein to retain impurities and pass the selected protein to the permeate.
  • a different filtration concentration method is taught by European Patent No. EP 467 482 Bl in which defatted skim milk is reduced to pH 3-4, below the pi of the milk proteins, to solubilize both casein and whey proteins. Three successive rounds of ultrafiltration or diafiltration then concentrate the proteins to form a retentate containing 15-20% solids of which 90% is protein.
  • milk containing an antibody of interest can initially be clarified.
  • a typical clarification protocol can include the following steps: (a) diluting milk 2: 1 with 2.0 M Arginine-HCl pH 5.5; (b) spinning diluted sample in centrifuge for approximately 20 minutes at 4-8°C;
  • antibody DNA sequences can be derived in several ways.
  • One method provides for using mRNA derived from antibody- producing cells such as antigen-stimulated B lymphocytes from the spleen or peripheral blood.
  • Hybridomas are also a source of mRNA that predominantly or exclusively encodes a single antibody.
  • mRNAs have a polyadenylate (poly A) sequence at their 3' end, they can be purified from the total RNA population by affinity chromatography on oligodeoxythymidylate-cellulose according to methods known in the prior art.
  • a complementary DNA (cDNA) copy of the mRNA is then made using the enzyme reverse transcriptase.
  • Commercial kits are also available that provide all the
  • More straightforward methods include obtaining the recombinant antibody sequences of interest can be obtained by screening libraries of genomic material or reverse-translated messenger RNA derived from the animal of choice (such as cattle or mice), or through appropriate sequence databases such as NCBI, genbank, etc. These sequences along with the desired polypeptide sequence of the antibody of interest can then be cloned into an appropriate plasmid vector and amplified in a suitable host organism, usually E. coll The DNA sequence encoding the peptide of choice can then be constructed, for example, by polymerase chain reaction amplification of a mixture of overlapping annealed oligonucleotides. The DNA sequence can then be inserted in a production system that would include in vitro cell culture systems or transgenic animal systems.
  • Antibody engineering is the process of altering antibody structure and functional properties by recombinant DNA methods. According to the methods of the current invention once the DNA sequences of the variable regions are known, the amino acid sequence can be deduced. Methods of in vitro mutagenesis can then be applied to insert, delete, or change one or several amino acids, or to exchange entire variable domains as desired. Many laboratories worldwide are now using these techniques to produce antibodies that would be difficult or impossible to obtain from animals.
  • the DNA sequence is determined using chain termination sequencing methods that are now available in commercial kits (Maniatis et al., 1989).
  • the sequencing requires oligonucleotide primers complementary to the 5' ends of the region to be sequenced. As portions of the sequence are determined, additional primers may be needed to extend the sequence. Overlapping regions of sequence determined in separate runs are aligned to obtain the entire sequence. Both strands of the DNA are sequenced in order to verify the results.
  • a frequently updated database of all reported antibody sequences has been compiled (Kabat et al., 1991) and is available. New antibody sequences can be analyzed by first comparing them to the most similar counterparts in known antibody sequence databases.
  • a DNA sequence encoding a fusion protein sequence of the present invention is constructed using known recombinant DNA techniques to assemble separate DNA sequences encoding the first and second polypeptides into an appropriate expression vector.
  • the 3' end of a DNA sequence encoding the first polypeptide is ligated, with or without a peptide linker, to the 5' end of a DNA sequence encoding the second polypeptide so that the reading frames of the sequences are in phase to permit mRNA translation of the two DNA sequences into a single fusion protein that retains the biological activity of both the first and the second polypeptides.
  • the solid support of the invention may be any solid material known to those of ordinary skill in the art to which the antigen may be attached.
  • the solid support may be a test well in a microtiter plate or a nitrocellulose or other membrane.
  • the support may be a bead or disc, such as glass, gold, fiberglass, latex or a plastic material such as polystyrene or polyvinylchloride.
  • the support may also be a magnetic particle or a fiber optic sensor, such as those disclosed, for example, in U.S. Pat. No. 5,359,681.
  • polypeptides may be bound to the solid support using a variety of techniques known to those of ordinary skill in the art, which are amply described in the patent and scientific literature.
  • the term "bound” refers to both noncovalent association, such as adsorption, and covalent attachment (which may be a direct linkage between the antigen and functional groups on the support or may be a linkage by way of a cross-linking agent). Binding by covalent attachment through adding cyclized peptides to target antibodies is preferred.
  • adsorption it may be achieved by contacting the polypeptide, in a suitable buffer, with the solid support for a suitable amount of time.
  • the contact time varies with temperature, but is typically between about 1 hour and 1 day.
  • contacting a well of a plastic microtiter plate (such as polystyrene or polyvinylchloride) with an amount of polypeptide ranging from about 10 ng to about 1 ⁇ g, preferably about 100 ng, is sufficient to bind an adequate amount of antigen.
  • Covalent attachment of polypeptide to a solid support may generally be achieved by first reacting the support with a bifunctional reagent that will react with both the support and a functional group, such as a hydroxyl or amino group, on the polypeptide.
  • a bifunctional reagent that will react with both the support and a functional group, such as a hydroxyl or amino group, on the polypeptide.
  • the polypeptide may be bound to supports having an appropriate polymer coating using benzoquinone or by condensation of an aldehyde group on the support with an amine and an active hydrogen on the polypeptide.
  • Cyclization of peptides reduces the conformational freedom of these flexible, linear molecules, and often results in higher receptor binding affinities by reducing unfavorable entropic effects. Because of the more constrained structural framework, these agents are more selective in their affinity to specific receptor cavities and structurally offer better support. By the same reasoning, structurally constrained cyclic peptides confer greater stability against the action of proteolytic enzymes and can be more densely packed onto the surface of a selected support.
  • Methods for cyclization can be classified into "backbone to backbone” cyclization strategies through the formation of amide bond between the N-terminal and the C-terminal amino acid residues, and cyclizations involving the side chains of individual amino acids.
  • the latter method includes: the formation of disulfide bridges between two w-tliio amino acid residues (cysteine, homocysteine); the formation of
  • lactam bridges between glutamic/aspartic acid and lysine residues the formation of lactone or thiolactone bridges between amino acid residues containing carboxyl, hydroxyl or mercapto functional groups; the formation of thioether or ether bridges between the amino acids containing hydroxyl or mercapto functional groups; and, other special methods.
  • lactone or thiolactone bridges between amino acid residues containing carboxyl, hydroxyl or mercapto functional groups the formation of thioether or ether bridges between the amino acids containing hydroxyl or mercapto functional groups
  • more recently developed general methods to effectively construct some of the aforementioned cyclic peptide derivatives can be used to aid in the anchoring of a biosensor antibody to a solid support.
  • the conformation ally constrained bicyclic antibodies of the invention provide a structurally balanced structure for specific biosensor antibodies.
  • lantibiotics In proteins and in many physiologically important eukaryotic peptides, the presence of intramolecular disulfide bonds provide major conformational controls towards the constraint and stabilization of structures.
  • lantibiotics In a subset of natural product peptides, termed lantibiotics, there are cyclic sub-structures that contain thioether linkages spanning 4 to 7 amino acids. Lantibiotics, such as nisin, epidermin, and duramicycin, are produced by bacterial microorganisms. Many of these possess antimicrobial properties.
  • the common building block in these peptides is lanthionine, an amino acid building block that consists of two alanines with their b-carbons bridged with a single sulfur atom.
  • these thioether linked alanine subunits in lantibiotics are part of the cyclic peptide loops of various sizes in these natural products. According to the current invention these disulfide bonds or cyclized peptides may be added to antibodies or calins of interest to provide additional support, promote optimal orientation and optimal "packing" on a support.
  • the main advantage of thioether-cyclized peptide analogues over disulfide-linked constructs is the stability of the thioether to redox conditions.
  • the creation of redox stable thioether linkages in rationally designed peptidomimetics is an attractive approach to restrict the conformational space and improve the biostability of such experimental agents.
  • the agents immobilized on a solid support can be antibodies, recombinant antibodies, or modified antibodies.
  • Antibodies are raised by immunizing animals (e.g., rabbit, mouse, rat, goat or chicken) with antigens (proteins or peptides). A large number of antibodies (monoclonal and polyclonal) are commercially available. Recombinant and modified antibodies are constructed by using recombinant DNA techniques.
  • the supports are either plates (glass or plastics), membranes made of nitrocellulose, nylon, or polyvinylidene difluoride (PVDF), or gold beads. Membranes are easier to handle and agents can be readily immobilized on them. Glass or plastic plates provide rigid support and are therefore necessary in some special applications.
  • agents are immobilized on a solid support directly or indirectly.
  • Agents can be directly deposited at high density on a support, which can be as small as a microscopic slide. Similar technology was developed for a making high density DNA microarray (Shalon et al, GENOME RESEARCH, 1996 JuI; 6(7): 639-45).
  • Agents can also be immobilized indirectly on the support. For instance, antibody X or Y can be printed on a support. Agents (antibodies) are then immobilized on the support through their interactions with antibody X or Y.
  • variable regions of the antibodies will be fully exposed to interact with antigens.
  • Recombinant antibodies can be immobilized through the interaction between their tags and the ligands printed on the support.
  • One most important characteristic of protein arrays utilizing visual cues or that is read visually is that all agents are immobilized at predetermined positions, so that each agent can be identified by its position or read as appropriate.
  • multiple array sections can be designed to report contact with antigens of interest through known methods (e.g., changes in weight, conductivity, fluorescence, chemical activity, etc., ).
  • the deciphering of the human genome sequence, elucidation of the complete genomes of many pathogens, and the development of new antibodies to those specific pathogenic agents provides new uses for substantial amounts of individual antibodies for various desirable biosensor applications. That is, in a preferred embodiment the current invention utilizes multiple antibodies directed towards a variety of pathogenic organisms or specific sensor molecules built into a biosensor antibody array detection device. In this fashion the current invention will provide unprecedented opportunities for antibody-based biosensor technologies to quickly detect and allow users to counter exposure to weaponized biological agents.
  • Bio agents of interest would include the following organisms, with antibodies of the invention directed against them and/or the biotoxins they produce: Bacillus Anthracis; Yersina Pestis; Yersina enterocolitica; Francisella Tularensis; Vibrio Cholerae;
  • Molecular toxins or chemicals that would be detected by embodiments of the biosensor array of the invention would include: ricin; sarin; soman, tabun; cyanogens; chloride and hydrogen chloride; oleoresin capsicum; arsene; chlorine, diphosgene; phosgene; distilled mustard, ethyldichloroarsine, mustard-lewisite mixture; nitrogen mustard; and, organophosphate pesticides; aflatoxin; Trichothecene mycotoxins, and the Staphylococcus enterotoxins A, B and C.
  • Agricultural bioterrorism causative agents in addition to several of the above, would also include: foot-and-mouth disease, mad cow disease, swine fever and karnal bunt of wheat.
  • the current invention relies on an array of various antibodies comprising a surveillance system for multiple aerosolized pathogenic agents of interest.
  • detection of pathogenic biological agents relies in part on the use of nucleic acid sequence databases for pathogen strain type identification and the use of that information for synthetic sequencing of an appropriate antigen carrying the detection target of interest.
  • the methods of the current invention provide for the selective, as well as, more efficient methods of production of selected antibodies modified to be useful for the detection of certain detrimental pathogens or antigens. This may be accomplished, according to the current invention, through the use of transgenically derived antibodies designed to detect pathogens of interest and then put together in certain combinations reflecting the profile of agents that users were aware of and want to be in a position to detect early.
  • the present invention an antibody array for use in biosensor applications provides a powerful and quantitative tool for detection of a variety of antigens and/or protein sequences of interest.
  • the antibody array of the invention is based on several principles. First, a protein can be recognized and identified unambiguously by specific molecules such as antibodies, recombinant proteins and small chemicals that can specifically interact with it. Second, an antibody or a small chemical can be immobilized on a solid support with the immobilized molecule retaining its ability interact in an antibody— protein binding.
  • Antibodies can be immobilized on solid supports such as glass plates, agarose beads, gold beads or PVDF membranes (LeGendre, 1990, BIOTECHNIQUES, Vol.9, No.6, p. 788- 805).
  • solid supports such as glass plates, agarose beads, gold beads or PVDF membranes (LeGendre, 1990, BIOTECHNIQUES, Vol.9, No.6, p. 788- 805).
  • many different antibody agents can be immobilized at different positions on a solid support without cross interactions among them, with supporting reporting mechanism means remaining intact and specific. This insures that each agent independently interacts with its respective detection target - typically a protein.
  • biosensor antibody arrays can be made for different purposes or perceived threats.
  • a typical WMD array could be made against a variety known biotoxins, viruses or small molecules developed for that purpose or thought to be available for that purpose.
  • a separate array could be made to detect for multiple small molecule agents present in various Chemical Agents or Blistering Agents - such as sarin gas or mustard gas or for application in a specific location such as an urban environment or in a food processing center with the bioarray focusing on bacterial agents that could cause food spoilage or virsues indicating animals or produce affected by specific pathogens.
  • 26 expression pattern in a source e.g. a cell line
  • thousands of different antibodies are immobilized in a single support.
  • the amount of antibodies immobilized can also be different, preferably in the range of nanogram to microgram.
  • the number of different agents immobilized on one solid support varies depending on the particular applications envisioned for the needs of the users.
  • the protein sample can also be labeled by biotinylation in vitro. Biotinylated proteins trapped on the array will then be detected by avidin or streptavidin which strongly binds biotin. If avidin is conjugated with horseradish peroxidase or alkaline phosphatase, the captured protein can be visualized by enhanced chemical luminescence. The amount of proteins bound to each antibody represents the level of the specific protein in the sample. If a specific group of proteins are interested, they can be detected by agents which specifically recognize them. Other methods, like immunochemical staining, surface plasmon resonance, matrix-assisted laser desorption/ionization-time of flight, can also be used to detect the captured proteins and provide means to appropriately indicate the specific agents detected.
  • biotinylated proteins trapped on the array will then be detected by avidin or streptavidin which strongly binds biotin. If avidin is conjugated with horseradish peroxidase or alkaline phosphatase, the captured protein can be visualized by enhanced chemical lumin
  • a further challenge is the need for highly sensitive systems, as some highly infectious pathogens require the inhalation of only 1 to 10 organisms to cause disease. In general, much greater attention is needed to translate basic laboratory research into field applications and clinical validation. Finally, because no test is perfect, it is important to be able to anticipate false-positive test results in a reliable and quantitative fashion.
  • One potential strategy for minimizing the impact of false-positive test results is to create a system of multiple, parallel, independent technical platforms so as to avoid dependence on any one testing procedure. This requires crosscutting, interdisciplinary science (e.g., combining environmental microbiology, cell biology, biophysics, electronics, materials science and microfabrication, microfluidics, and bioinformatics/statistics) .
  • Antibodies of the present invention include naturally purified products, products of chemical synthetic procedures, and products produced by recombinant techniques from a eukaryotic host, including, for example, yeast, higher plant, insect and mammalian cells. Depending upon the host employed in a recombinant production procedure, the antibody of the present invention can be glycosylated or can be non-glycosylated, with glycosylation being preferred, and glycosylation from a transgenic mammal being most preferred.
  • the isolated antibodies and antibodies of the present invention comprise at least one antibody and/or antibody amino acid sequence disclosed or described herein encoded by any suitable polynucleotide, or any at least one isolated or prepared antibody.
  • the at least one antibody has at least one biosensor activity and the at least
  • one antibody binds human biosensor antibody and, thereby partially or substantially modulates at least one structural or biological activity of at least one biosensor antibody.
  • biosensor antibody of the current invention refers to a antibody as described herein that has at least one biosensor-dependent activity, such as 5-10,000%, of the activity of a known or other biosensor antibody or active portion thereof, preferably by at least about 10, 20, 30, 40, 50, 55, 60, 65, 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100% or more, depending on the assay.
  • the capacity of a biosensor antibody of the current invention to have at least one biosensor- dependent activity is preferably assessed by at least one suitable biosensor antibody or receptor assay, as described herein and/or as known in the art.
  • neutralizing antibody refers to an antibody that can inhibit at least one biosensor-dependent activity by about 5-120%, preferably by at least about 10, 20, 30, 40, 50, 55, 60, 65, 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100% or more depending on the assay.
  • the capacity of a biosensor antibody of the current invention to inhibit a biosensor-dependent activity is preferably assessed by at least one suitable biosensor antibody or receptor assay, as described herein and/or as known in the art.
  • An antibody of the invention can be of any class (IgG, IgA, IgM, IgE, IgD, etc.) or isotype and can comprise a kappa or lambda light chain.
  • the human antibody comprises an IgG heavy chain or defined fragment, for example, at least one of isotypes, IgGl, IgG2, IgG3 or IgG4.
  • Antibodies of this type can be prepared by employing a transgenic mouse or other transgenic non-human mammal comprising at least one human light chain (e.g., IgG, IgA and IgM (e.g., ⁇ l, ⁇ 2, ⁇ 3, ⁇ 4) transgenes as described herein and/or as known in the art.
  • a biosensor antibody human antibody comprises an IgGl heavy chain and an IgGl light chain.
  • At least one antibody of the invention binds at least one specified epitope specific to at least one biosensor antibody of the current invention, subunit, fragment, portion or any combination thereof.
  • the at least one epitope can comprise at least one antibody binding region that comprises at least one portion of the antibody, which epitope can optionally comprise at least one portion of at least one extracellular, soluble, hydrophillic, external or cytoplasmic portion of the antibody.
  • the at least one antibody of the present invention can preferably comprise at least one antigen-binding region that comprises at least one human
  • the antibody and antibody can have an antigen-binding region that comprises at least a portion of at least one heavy chain (HC) CDR (i.e., HC CDRl , HC CDR2 and/or HC CDR3) having the amino acid sequence of the corresponding HC CDRs 1, 2 and/or 3.
  • HC heavy chain
  • the antibody or antigen-binding portion or variant can have at least one antigen-binding region that comprises at least a portion of at least one light chain (LC) CDR (i.e., LC CDRl, LC CDR2 and/or LC CDR3).
  • LC light chain
  • the three heavy chain CDRs and the three light chain CDRs of the antibody or antigen-binding fragment have the amino acid sequence of the corresponding CDR of at least one of biosensor antibody monoclonal antibody, as described herein.
  • Such antibodies can be prepared by chemically joining together the various portions (e.g., CDRs, framework) of the antibody using conventional techniques, by preparing and expressing a (i.e., one or more) nucleic acid molecule that encodes the antibody using conventional techniques of recombinant DNA technology or by using any other suitable method.
  • a nucleic acid molecule that encodes the antibody using conventional techniques of recombinant DNA technology or by using any other suitable method.
  • the biosensor antibody of the current invention can comprise at least one of a heavy or light chain variable region having a defined amino acid sequence.
  • antibodies of the current invention comprises at least one of at least one heavy chain variable region; and/or at least one light chain variable region.
  • Antibody Arrays [0078] Several novel truncated antibody constructs according to the current invention are possible, including single chain Fv-Fc dimers, minibodies which lack the antibody CHl and CH2 domains, and maxibodies which lack the CHl domain.
  • An antibody according to the present invention can include any antibody or peptide containing molecule that comprises at least a portion of an immunoglobulin molecule, such as but not limited to at least one complementarity determining region (CDR) (also termed the hypervariable region or HV) of a heavy or light chain variable region, or a ligand binding portion thereof, a heavy chain or light chain variable region, a heavy chain or light chain constant region, a framework region, or any portion thereof,
  • CDR complementarity determining region
  • the present invention provides, in one aspect, isolated nucleic acid molecules comprising, complementary, or hybridizing to, a polynucleotide encoding specific biosensor antibody antibodies, comprising at least one specified sequence, domain, portion or variant thereof.
  • the present invention further provides recombinant vectors comprising at least one biosensor antibody of the current invention or antibody encoding or complementary nucleic acid molecules, host cells containing such nucleic acids and/or recombinant vectors, as well as methods of making and/or using such antibody nucleic acids, vectors and/or host cells.
  • At least one antibody of the invention binds at least one specified epitope specific to at least one biosensor antibody of the current invention, subunit, fragment, portion or any combination thereof.
  • the at least one epitope can comprise at least one antibody binding region that comprises at least one portion of said antibody, which epitope is preferably comprised of at least 1-5 amino acids of at least one portion thereof, such as but not limited to, at least one functional, extracellular, soluble, hydrophillic, external or cytoplasmic domain of said antibody, or any portion thereof.
  • Bispecific, heterospecif ⁇ c, heteroconjugate or similar antibodies can also be used that are monoclonal, preferably human or humanized, antibodies that have binding specificities for at least two different antigens.
  • one of the binding specificities is for at least one biosensor antibody of the current invention, the other one is for any other antigen.
  • Methods for making bispecific antibodies are known in the art. Traditionally, the recombinant production of bispecific antibodies is based on the co-expression of two immunoglobulin heavy chain-light chain pairs, where the two heavy chains have different specificities (Milstein and Cuello, Nature 305:537 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of 10 different antibody molecules, of which only one has the correct bispecific structure. The purification of the correct molecule, which is usually done by affinity chromatography steps, is rather cumbersome, and the product yields are low. Similar procedures are disclosed, e.g., in WO 93/08829, U.S. Pat. Nos, 6,210,668, 6,193,967, 6,132,992, 6,106,833, 6,060,285, 6,037,453,
  • Such antibodies optionally further affect a specific ligand, such as but not limited to where such antibody modulates, decreases, increases, antagonizes, angonizes, mitigates, aleviates, blocks, inhibits, abrogates and/or interferes with at least oneBiosensor antibody activity or binding, or with Biosensor antibody receptor activity or binding, in vitro, in situ and/or in vivo.
  • a suitable Biosensor antibody of the current invention, specified portion or variant of the present invention can bind at least oneBiosensor antibody, or specified portions, variants or domains thereof.
  • a suitable biosensor antibody of the current invention, specified portion, or variant can also optionally affect at least one of antibody activity or function, such as but not limited to, RNA, DNA or antibody synthesis, antibody release, antibody receptor signaling, membrane antibody cleavage, antibody activity, antibody production and/or synthesis.
  • antibody activity or function such as but not limited to, RNA, DNA or antibody synthesis, antibody release, antibody receptor signaling, membrane antibody cleavage, antibody activity, antibody production and/or synthesis.
  • Biosensor antibodies useful in the methods and compositions of the present invention can optionally be characterized by high affinity binding to specific antigens.
  • the isolated nucleic acids of the present invention can be made using (a) recombinant methods, (b) synthetic techniques, (c) purification techniques, or combinations thereof, as well-known in the art.
  • the nucleic acids can conveniently comprise sequences in addition to a polynucleotide of the present invention.
  • a multi-cloning site comprising one or more endonuclease restriction sites can be inserted into the nucleic acid to aid in isolation of the polynucleotide.
  • translatable sequences can be inserted to aid in the isolation of the translated polynucleotide of the present invention.
  • a hexa-histidine marker sequence provides a convenient means to purify the antibodies of the present invention.
  • the nucleic acid of the present invention—excluding the coding sequence— is optionally a vector, adapter, or linker for cloning and/or expression of a polynucleotide of the present invention.
  • Additional sequences can be added to such cloning and/or expression sequences to optimize their function in cloning and/or expression, to aid in isolation of the polynucleotide, or to improve the introduction of the polynucleotide into a cell.
  • Use of cloning vectors, expression vectors, adapters, and linkers is well known in the art. (See, e.g., Ausubel, supra; or Sambrook, supra)
  • RNA, cDNA, genomic DNA, or any combination thereof can be obtained from biological sources using any number of cloning methodologies known to those of skill in the art.
  • oligonucleotide probes that selectively hybridize, under stringent conditions, to the polynucleotides of the present invention are used to identify the desired sequence in a cDNA or genomic DNA library.
  • the isolation of RNA, and construction of cDNA and genomic libraries, is well known to those of ordinary skill in the art. (See, e.g., Ausubel, supra; or Sambrook, supra)
  • a cDNA or genomic library can be screened using a probe based upon the sequence of a polynucleotide of the present invention, such as those disclosed herein. Probes can be used to hybridize with genomic DNA or cDNA sequences to isolate homologous genes in the same or different organisms. Those of skill in the art will appreciate that various degrees of stringency of hybridization can be employed in the assay; and either the hybridization or the wash medium can be stringent. As the conditions for hybridization become more stringent, there must be a greater degree of complementarity between the probe and the target for duplex formation to occur.
  • the degree of stringency can be controlled by one or more of temperature, ionic strength, pH and the presence of a partially denaturing solvent such as formamide.
  • the stringency of hybridization is conveniently varied by changing the polarity of the reactant solution through, for example, manipulation of the concentration of formamide within the range of 0% to 50%.
  • the degree of complementarity (sequence identity) required for detectable binding will vary in accordance with the stringency of the
  • the degree of complementarity will optimally be 100%, or 70-100%, or any range or value therein. However, it should be understood that minor sequence variations in the probes and primers can be compensated for by reducing the stringency of the hybridization and/or wash medium. [0089] Methods of amplification of RNA or DNA are well known in the art and can be used according to the present invention without undue experimentation, based on the teaching and guidance presented herein.
  • immunoassays are based upon the immunological reaction between proteins such as antibodies, antibody fragments, or even artificially generated elements simulating antibody binding sites such as peptides, templated polymers and the like (hereafter referred to as antibody recognition) and the substance for which they are specific, the ligand.
  • Immunological reactions are characterized by their high specificity and accordingly, numerous schemes have been developed in order to take advantage of this characteristic. The goal is to identify a particular state with absolute specificity using as few assays as possible.
  • an antibody is immobilized on a solid phase such as microparticles, microtiter wells, paddles, and the like.
  • the sample is then contacted with the immobilized antibody and the ligand binds if present in the sample.
  • the bound substance is detected and quantitated by an entity associated directly or indirectly therewith.
  • detectable entity include fluorescent molecules, chemiluminescent molecules, enzyme, isotopes, microparticles and the like. Many variants have been developed such as competition, indirect competition, and the like. Various methods are available to those skilled in the art for quantitating the amount of substance bound using these assays.
  • the isolated nucleic acids of the present invention can also be prepared by direct chemical synthesis by known methods (see, e.g., Ausubel, et al, supra). Chemical synthesis generally produces a single-stranded oligonucleotide, which can be converted into double-stranded DNA by hybridization with a complementary sequence, or by polymerization with a DNA polymerase using the single strand as a template.
  • Chemical synthesis of DNA can be limited to sequences of about 100 or more bases, longer sequences can be obtained by the ligation of shorter sequences.
  • the present invention further provides recombinant expression cassettes comprising a nucleic acid of the present invention.
  • a nucleic acid sequence of the present invention for example a cDNA or a genomic sequence encoding an antibody of the present invention, can be used to construct a recombinant expression cassette that can be introduced into at least one desired host cell.
  • a recombinant expression cassette will typically comprise a polynucleotide of the present invention operably linked to transcriptional initiation regulatory sequences that will direct the transcription of the polynucleotide in the intended host cell. Both heterologous and non-heterologous (i.e., endogenous) promoters can be employed to direct expression of the nucleic acids of the present invention.
  • isolated nucleic acids that serve as promoter, enhancer, or other elements can be introduced in the appropriate position (upstream, downstream or in intron) of a non-heterologous form of a polynucleotide of the present
  • endogenous promoters can be altered in vivo or in vitro by mutation, deletion and/or substitution.
  • a biosensor antibody of the current invention of the present invention can include one or more amino acid substitutions, deletions or additions, either from natural mutations or human manipulation, as specified herein.
  • the number of amino acid substitutions a skilled artisan would make depends on many factors, including those described above. Generally speaking, the number of amino acid substitutions, insertions or deletions for any given biosensor antibody of the current invention, fragment or variant will not be more than 40, 30, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, such as 1-30 or any range or value therein, as specified herein.
  • Amino acids in an biosensor antibody of the current invention of the present invention that are essential for function can be identified by methods known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis (e.g., Ausubel, supra, Chapters 8, 15; Cunningham and Wells, SCIENCE 244:1081-1085 (1989)).
  • site-directed mutagenesis or alanine-scanning mutagenesis e.g., Ausubel, supra, Chapters 8, 15; Cunningham and Wells, SCIENCE 244:1081-1085 (1989)
  • the latter procedure introduces single alanine mutations at every residue in the molecule.
  • the resulting mutant molecules are then tested for biological activity, such as, but not limited to at least one biosensor neutralizing activity.
  • Sites that are critical for antibody binding can also be identified by structural analysis such as crystallization, nuclear magnetic resonance or photoaffmity labeling (Smith, et al, J. MOL. BiOL.
  • Non-limiting CDRs or portions of biosensor antibodies or antibodies of the invention that can enhance or maintain at least one of the listed activities include, but are not limited to, any of the above polypeptides, further comprising at least one mutation corresponding to at least one substitution selected from the group consisting of at least one of extracellular, intracellular, soluble, at least 10 contiguous amino acids, and the like, extracellular, intracellular, soluble, at least 10 contiguous amino acids, and the like.
  • Non-limiting variants that can enhance or maintain at least one of the listed activities include, but are not limited to, any of the above polypeptides, further
  • the antibodies and antibodies of the present invention can comprise any number of contiguous amino acid residues from an antibody of the present invention, wherein that number is selected from the group of integers consisting of from 10-100% of the number of contiguous residues in a biosensor antibody of the current invention or antibody.
  • this subsequence of contiguous amino acids is at least about 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250 or more amino acids in length, or any range or value therein.
  • the number of such subsequences can be any integer selected from the group consisting of from 1 to 20, such as at least 2, 3, 4, or 5.
  • the present invention includes at least one biologically active antibody or antibody of the present invention.
  • Biologically active antibodies or antibodies have a specific activity at least 20%, 30%, or 40%, and preferably at least 50%, 60%, or 70%, and most preferably at least 80%, 90%, or 95%- 1000% of that of the native (non-synthetic), endogenous or related and known antibody or antibody.
  • Methods of assaying and quantifying measures of enzymatic activity and substrate specificity are well known to those of skill in the art.
  • Receptors are often labeled with biotin allowing the receptors to be immobilized to an avidin-coated support. Biotin labeling can be performed using the biotinylating enzyme, BirA (see, Schatz, BIOTECHNOLOGY 11:1138-43 (1993)).
  • BirA biotinylating enzyme
  • Antibody chains can then be purified by methods including ammonium sulfate precipitation, tangential flow filtration, high performance tangential flow filtration, affinity chromatography to immobilized target, column chromatography, gel electrophoresis and the like (see Scopes, in PROTEIN PURIFICATION (Springer- Verlag, N. Y., 1982)).
  • Purified antibodies are dialyzed against a minimum of 100 volumes of phosphate buffered saline (PBS), pH 7.4, for at least 4 hours. Antibodies are diluted to a final concentration of 2 mg/ml in PBS.
  • a stock solution containing 40 mM of biotin-XX- NHS ester (Molecular Probes, Eugene, Oreg.) can be prepared in dimethylsulfoxide.
  • the biotin-XX-NHS solution can be added to antibodies at a final concentration of 0.4 mM and reacted for 90 minutes at room temperature.
  • Aminoethanesulfonic acid can be added to a final concentration of 20 mM and incubated for five minutes to quench remaining reactive groups.
  • the biotinylated antibodies are dialyzed extensively to remove small molecules containing biotin from the antibodies.
  • the observed orientation has been essentially accidental, so that not all the adsorbed molecules could be expected to be capable of analyte capture.
  • molecular orientation can some extent be controlled by buffer composition (e.g. relative pH and molecular charge), and the choice between available solid surfaces.
  • buffer composition e.g. relative pH and molecular charge
  • analyte binding sites are usually associated with functional groups, it may often be favorable to aim at hydrophobic adsorption, using a hydrophobic surface.
  • more precise orientation control may be obtainable by covalent coupling or the addition of cyclized peptides. In general, if an available surface is not immediately suitable, it may be made so by coating with an appropriate precapture reagent.
  • the current invention it is desirable to immobilize antibodies in a single orientation, with the antigen-binding site positioned away from the surface and facing the capture solution, which will optimize the activities of immobilized biosensor antibodies and the performance of arrays.
  • One available strategy involves the a site-specific introduction of biotin, which in turn provide a uniform orientation of the immobilized antibody. This overcomes the prior art limitation of the chemical biotinylation of antibodies, followed by their subsequent immobilization onto avidin- or streptavidin-coated surfaces. While these methods would likely allow biotinylated antibodies to retain their native conformation when immobilized, the antibodies are also often linked in multiple orientations relative to the surface.
  • cyanosilane is an available coupling agent for direct binding of antibodies to silica supports. Also according to the current invention precise placement of the antibodies on a given support may be critically important for reporting
  • microarray substrates have been described in the prior art including: nylon membranes, plastic microwells, planar glass slides, gel-based arrays and beads in suspension arrays. Much effort has been expended in optimizing antibody attachment to the microarray substrate.
  • various signal generation and signal enhancement strategies have been employed in antibody arrays, including colorimetry, radioactivity, fluorescence, chemiluminescence, quantum dots and other nanoparticles, enzyme-linked assays, resonance light scattering, tyramide signal amplification and rolling circle amplification.
  • Each of these formats and procedures has distinct advantages and disadvantages, relating broadly to sensitivity, specificity, dynamic range, multiplexing capability, precision, throughput, and ease of use.
  • multiplexed microarray immunoassays are ambient analyte assays.
  • the anchored component may be immobilized by non-covalent or covalent attachments.
  • Non-covalent attachment may be accomplished by simply coating the solid surface with a solution of the protein and drying.
  • an immobilized antibody preferably a monoclonal antibody, specific for the protein to be immobilized may be used to anchor the protein to the solid surface.
  • the surfaces may be prepared in advance and stored.
  • the nonimmobilized component is added to the coated surface containing the anchored component. After the reaction is complete, unreacted components are removed (e.g., by washing) under conditions such that any complexes formed will remain immobilized on the solid surface.
  • the detection of complexes anchored on the solid surface can be accomplished in a number of ways.
  • the detection of label immobilized on the surface indicates that complexes are formed.
  • an indirect label can be used to detect complexes anchored on the surface; e.g., using a labeled antibody specific for the previously nonimmobilized component (the antibody, in turn, may be directly labeled or indirectly labeled with a labeled anti-Ig antibody).
  • a reaction can be conducted in a liquid.
  • Immobilization can be based on covalent or noncovalent interaction of the molecule with the surface, Noncovalent interactions include hydrophobic interactions, hydrogen bonding, van der Waals forces, electrostatic forces, or physical adsorption. These include the use of surfaces such as nitrocellulose or silane-coated glass slides and require no modifications to the molecule before attachment. However, since these interactions are weak, the molecules can get denatured or dislodged, thus causing loss of signal.
  • the covalent attachment onto surfaces leads to molecules being arranged in a definite, orderly fashion and uses spacers and linkers to help minimize steric hindrances.
  • the modified human antibodies and antibodies can be prepared using suitable methods, such as by reaction with one or more modifying agents.
  • An "activating group” is a chemical moiety or functional group that can, under appropriate conditions, react with a second chemical group thereby forming a covalent bond between the modifying agent and the second chemical group.
  • amine-reactive activating groups include electrophilic groups such as tosylate, mesylate, halo (chloro, bromo, fluoro, iodo), N-hydroxysuccinimidyl esters (NHS), and the like.
  • Activating groups that can react with thiols include, for example, maleimide, iodoacetyl, acrylolyl, pyridyl disulfides, 5-thiol-2-nitrobenzoic acid thiol (TNB-thiol), and the like.
  • An aldehyde functional group can be coupled to amine- or hydrazide-containing molecules, and an azide group can react with a trivalent phosphorous group to form phosphoramidate or phosphorimide linkages.
  • Suitable methods to introduce activating groups into molecules are known in the art (see for example, Hermanson, G. T., BIOCONJUGATE TECHNIQUES, in Academic Press: San Diego, Calif. (1996)).
  • An activating group can be bonded directly to the organic group (e.g., hydrophilic polymer, fatty acid, fatty acid ester), or through a linker moiety, for example a divalent C 1 -C 12 group wherein one or more carbon atoms can be replaced by a heteroatom such as oxygen, nitrogen or sulfur.
  • Suitable linker moieties include, for example, tetraethylene glycol, -(CH 2 ) 3 ⁇ , -NH-(CH 2 ) 6 -NH-, --(CH 2 ) 2 ⁇ NH ⁇ ; and, -CH 2 -O-CH 2 -CH 2 -
  • Modifying agents that comprise a linker moiety can be produced, for example, by reacting a mono-Boc-alkyldiamine (e.g., mono-Boc- ethylenediamine, mono-Boc-diaminohexane)("BOC”) with a fatty acid in the presence of l-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) to form an amide bond between the free amine and the fatty acid carboxylate.
  • a mono-Boc-alkyldiamine e.g., mono-Boc- ethylenediamine, mono-Boc-diaminohexane
  • EDC l-ethyl-3-(3-dimethylaminopropyl) carbodiimide
  • the BOC protecting group can be removed from the product by treatment with trifluoroacetic acid (TFA) to expose a primary amine that can be coupled to another carboxylate as described, or can be reacted with maleic anhydride and the resulting product cyclized to produce an activated maleimido derivative of the fatty acid.
  • TFA trifluoroacetic acid
  • the present invention also provides at least one biosensor antibody of the current invention or antibody composition comprising at least one, at least two, at least three, at least four, at least five, at least six or more biosensor antibodies as described herein and/or as known in the art that are provided in a non-naturally occurring composition, mixture or form.
  • biosensor antibody used in accordance with the present invention can be produced by recombinant means, including from mammalian cell or transgenic preparations, or can be purified from other biological sources, as described herein or as known in the art.
  • the range of at least oneBiosensor antibody of the current invention in at least one product of the present invention includes amounts yielding upon reconstitution, if in a wet/dry system, concentrations from about 1.0 ng/ml to about 1000 mg/ml, although lower and higher concentrations are operable and are dependent on the intended delivery vehicle, e.g., solution formulations will differ from transdermal patch, pulmonary, transmucosal, or osmotic or micro pump methods.
  • the range of at least one biosensor antibody of the current invention in at least one product of the present invention includes amounts yielding upon reconstitution, if in a wet/dry system, concentrations from about 1.0 ⁇ g/ml to about 1000 mg/ml, although lower and higher concentrations are operable and are dependent on the intended delivery vehicle, e.g., solution formulations will differ from transdermal patch, pulmonary, transmucosal, or osmotic or micro pump methods.
  • electrostatic space chargers (negative air ionizers) have been found to reduce dust levels in poultry hatchers and housing facilities, thereby also diminishing airborne bacterial levels and the experimental transmission of Salmonella to chicks. Negative air ionization can also reduce S, enteritidis numbers in aerosols and on exposed surfaces.
  • electrostatic space chargers When applied to the current invention the ability of electrostatic charging to attract and hold bacteria or viruses borne on airborne particulate matter is critical to the success of this technology as a control strategy for monitoring the environmental presence or absence of pathogenic agents or chemical agents.
  • All electronic components except for the plug-in batteries are housed in a completely waterproof enclosure to allow disinfection by spraying after use.
  • the device operates by applying a strong electrostatic field to draw charged airborne dust particles and aerosols onto the surface of antibody covered supports. As charged particles are drawn to the media, adjacent air is pulled in behind them — similar to the effect of air being pulled down with rain as it falls from a storm cloud — and the charged particles and associated airborne microorganisms are tightly bound to the media by electrostatic attraction.
  • the electrostatic sampling device (with two plates each time) can be used to collect air samples for 20 min, 1 hr, or 3 hr. Once internalized ionized and purified water is added to provide an aqueous environment for testing with biosensor antibodies.
  • Detection/Reporting Within 15-20 minutes analytes detected, identified and reported.
  • the current invention contemplates the use of a Biosensor array containing a variety of antibodies against identified pathogenic organisms or molecules. These antibodies are fixed to the surface of a biosensor antibody array and are exposed to a possible source of ligand. Typically two steps are involved in the process: first, the sample and reagent must be mixed manually, and, second, the sample must be applied to the presence or absence of the target antigen is detected by a small red dot that the user compares to a color chart.
  • Antibody-conjugated horseradish peroxidase can be used as the enzymatic label and hydrogen peroxide and iodide ions used as substrates.
  • Horseradish peroxidase catalyzes the oxidation of the iodide ions by hydrogen peroxide to givelodine.
  • This iodine is then electrochemically reduced at the working electrode surface with an uptake of two electrons.
  • the flux of electrons out of the working electrode is measured in the form of a current signal.
  • the magnitude of the current signal is proportional to the number of biological agents that have been captured and labeled.
  • the protein of interest is then selected by its unique property, i.e., interaction with an antibody.
  • agents e.g., antibodies, ligands
  • solid supports e.g., agarose beads
  • proteins retain their abilities of interacting with other proteins or ligands after immobilization.
  • Antibody array which has many antibodies directed against specific pathogenic organisms immobilized on a solid support. Agents are immobilized in a predetermined order, i.e., each agent is immobilized at a specific position so that it can be identified by its unique position on the support.
  • the device can capture and identify the specific proteins from a mixture (e.g., cell lysate). After capture and separation, the proteins can be further characterized. Therefore, the antibody array of the current invention makes it possible to study a wide variety of proteins in a single experiment by a large number of antibodies and/or recombinant proteins immobilized on a support.
  • Antibody array and the methods presented here have several significant advantages over the current methods.
  • antibody array allows rapid detection of many proteins and thus makes it possible to compare protein expression profiles from different sources or those from the same source but under different conditions. Information on protein expression profile is very useful in identifying diagnostic and Biosensor targets.
  • antibody array makes it possible to detect posttranslational modifications of numerous proteins and provide a valuable tool to investigate protein and cellular regulations. Third, it can screen a large number of potential interactions directly; and it can detect interactions that take place only under certain conditions, e.g. phosphorylation. Antibody array is therefore useful for a variety of applications, particularly for revealing disease mechanisms, searching for diagnostic indicators and for identifying Biosensor targets. In addition, antibody array allows an individual user to access a large number of immobilized antibodies or recombinant proteins.
  • Biopolymers such as DNA, RNA, proteins or polypeptides, and polysaccharides can be directly activated using similar bi-functional silane compounds or other crosslinking reagents resulting in an immobilized biopolymer to a solid surface.
  • This invention demonstrates that the target molecules to be arrayed are first modified so that they have binding affinity for solid surfaces without losing their probing abilities. Because the modification is a separate process, virtually any biological molecule can be modified and arrayed. Thus, a skilled artisan realizes that this invention is not limited to nucleic acids, but can be used for a spectrum of biological molecules.
  • Caprine Fetal Somatic Cell Lines Primary caprine fetal fibroblast cell lines to be used as karyoplast donors are derived from 35 - and 40-day fetuses. Fetuses are surgically removed and placed in equilibrated phosphate-buffered saline (PBS, Ca /Mg -free). Single cell suspensions are prepared by mincing fetal tissue exposed to 0.025 % trypsin, 0.5 mM EDTA at 38°C for 10 minutes.
  • PBS equilibrated phosphate-buffered saline
  • fetal cell medium fetal cell medium [equilibrated Medium- 199 (M 199, Gibco) with 10% fetal bovine serum (FB S) supplemented with nucleosides, 0.1 mM 2-mercaptoethanol, 2 mM L-glutamine and 1%
  • Transfected fetal somatic cells are seeded in 4-well plates with fetal cell medium and maintained in culture (5% CO 2 , 39°C). After 48 hours, the medium can be replaced with fresh low serum (0.5 % FBS) fetal cell medium. The culture medium can be replaced with low serum fetal cell medium every 48 to 72 hours over the next 2 - 7 days following low serum medium, somatic cells (to be used as karyoplast donors) are harvested by trypsinization. The cells are re-suspended in equilibrated M 199 with 10% FBS supplemented with 2 mM L-glutamine, 1% penicillin/streptomycin (10,000 I. U. each/mL) for at least 6 hours.
  • the current experiments for the generation of desirable transgenic animals are preferably carried out with goat cells or mouse cells for the generation or goats or mice respectively but, according to the current invention, could be carried out with any mammalian cell line desired.
  • Oocyte donor does are synchronized and super ovulated as previously described (Ongeri, et al., 2001), and are mated over a 48-hour interval to fertile males for microinjection procedures and to vasectomized males for nuclear transfer procedures. After collection, fertilized embryos or unfertilized oocytes are cultured in equilibrated Ml 99 with 10% FBS supplemented with 2 mM L-glutamine and 1% penicillin/streptomycin (10,000 LU. each/mL).
  • oocytes are treated with cytochalasin-B (Sigma, 5 ⁇ g/mL in SOF with 10% FBS) 15 to 30 minutes prior to enucleation.
  • Metaphase-II stage oocytes are enucleated with a 25 to 30 ⁇ m glass pipette by aspirating the first polar body and adjacent cytoplasm surrounding the polar body ( ⁇ 30 % of the cytoplasm) to remove the metaphase plate. After enucleation, all oocytes are immediately reconstructed.
  • Donor cell injection can be conducted in the same medium used for oocyte enucleation.
  • One donor cell can be placed between the zona pellucida and the ooplasmic membrane using a glass pipet.
  • the cell-oocyte couplets are incubated in SOF for 30 to 60 minutes before electrotransgenic and activation procedures.
  • Reconstructed oocytes are equilibrated in transgenic buffer (300 mM mannitol, 0.05 mM CaCl 2 , 0.1 mM MgSO 4 , 1 mM K 2 HPO 4 , 0.1 mM glutathione, 0.1 mg/ml BSA) for 2 minutes.
  • Electro-fusion and activation are conducted at room temperature, in a transgenic chamber with 2 stainless steel electrodes fashioned into a "transgenic slide" (500 ⁇ m gap; BTX- Genetronics, San Diego, CA) filled with transgenic medium.
  • Transgenic fusion can be performed using a transgenic slide.
  • the transgenic slide can be placed inside a transgenic dish, and the dish can be flooded with a sufficient amount of transgenic buffer to cover the electrodes of the transgenic slide. Couplets are removed from the culture incubator and washed through transgenic buffer. Using a stereomicroscope, couplets are placed equidistant between the electrodes, with the karyoplast/cytoplast junction parallel to the electrodes. It should be noted that the voltage range applied to the couplets to promote activation and transgenic fusion can be
  • the initial single simultaneous transgenic and activation electrical pulse has a voltage range of 2.0 to 3.0 kV/cm, most preferably at 2.5 kV/cm, preferably for at least 20 ⁇ sec duration.
  • This is applied to the cell couplet using a BTX ECM 2001ELECTROCELL MANIPULATOR®.
  • the duration of the micropulse can vary from 10 to 80 ⁇ sec.
  • the treated couplet is typically transferred to a drop of fresh transgenic buffer. Transgenic treated couplets are washed through equilibrated SOF/FBS, then transferred to equilibrated SOF/ FBS with or without cytochalasin-B.
  • cytocholasin-B its concentration can vary from 1 to 15 ⁇ g/mL, most preferably at 5 ⁇ g/mL.
  • the couplets are incubated at 37-39 0 C in a humidified gas chamber containing approximately 5% CO 2 in air.
  • mannitol may be used in the place of cytocholasin-B throughout any of the protocols provided in the current disclosure (HEPES-buffered mannitol (0.3 mm) based medium with Ca +2 and BSA).
  • All nuclear transfer embryos of the current invention are cultured in 50 ⁇ L droplets of SOF with 10% FBS overlaid with mineral oil. Embryo cultures are maintained in a humidified 39°C incubator with 5% CO 2 for 48 hours before transfer of the embryos to recipient does. Recipient embryo transfer can be performed as previously described (Baguisi et al., 1999).
  • known microinjection protocols can be utilized to produce a transgenic animal contemplated by the invention and capable of producing a desired antibody.
  • pregnancy can be determined by ultrasonography starting on day 25 after the first day of standing estrus. Does are evaluated weekly until day 75 of gestation, and once a month thereafter to assess fetal viability.
  • parturition can be induced with 5 mg of PGF2 ⁇ (Lutalyse, Upjohn). Parturition occurred within 24 hours after treatment. kids are removed from the dam immediately after birth, and received heat-treated colostrum within 1 hour after delivery. Time frames appropriate for other ungulates with regard to pregnancy and perinatal care (e.g., bovines) are known in the art.
  • the present invention also includes a method of cloning a genetically engineered or transgenic mammal, by which a desired gene is inserted, removed or modified in the differentiated mammalian cell or cell nucleus prior to insertion of the differentiated mammalian cell or cell nucleus into the enucleated oocyte.
  • mammals obtained according to the above method and the offspring of those mammals.
  • the present invention is preferably used for cloning caprines or bovines but could be used with any mammalian species.
  • the present invention further provides for the use of nuclear transfer fetuses and nuclear transfer and chimeric offspring in the area of cell, tissue and organ transplantation.
  • Suitable mammalian sources for oocytes include goats, sheep, cows, pigs, rabbits, guinea pigs, mice, hamsters, rats, primates, etc.
  • the oocytes will be obtained from ungulates, and most preferably goats or cattle. Methods for isolation of oocytes are well known in the art. Essentially, this will comprise isolating oocytes from the ovaries or reproductive tract of a mammal, e.g., a goat.
  • a readily available source of ungulate oocytes is from hormonally induced female animals.
  • oocytes may preferably be matured in vivo before these cells may be used as recipient cells for nuclear transfer, and before they can be fertilized by the sperm cell to develop into an embryo.
  • Metaphase II stage oocytes which have been matured in vivo, have been successfully used in nuclear transfer techniques. Essentially, mature metaphase II oocytes are collected surgically from either non-super
  • hCG human chorionic gonadotropin
  • transgenic production of human recombinant pharmaceuticals in the milk of transgenic farm animals solves many of the problems associated with microbial bioreactors (e.g., lack of post-translational modifications, improper protein folding, high purification costs) or animal cell bioreactors (e.g., high capital costs, expensive culture media, low yields).
  • the current invention enables the use of transgenic production of biopharmaceuticals, transgenic proteins, plasma proteins, and other molecules of interest in the milk or other bodily fluid (i.e., urine or blood) of transgenic animals hemizygous for a desired gene.
  • chromosomes it is not necessary to remove the assembled construct from the vector; in such cases the amplified vector may be used directly to make transgenic animals.
  • this case refers to the presence of a first polypeptide encoded by enough of a protein nucleic acid sequence to retain its biological activity, this first polypeptide is then joined to a the coding sequence for a second polypeptide also containing enough of a polypeptide sequence of a protein to retain its physiological activity.
  • the coding sequence being operatively linked to a control sequence which enables the coding sequence to be expressed in the milk of a transgenic non-human placental mammal.
  • a DNA sequence which is suitable for directing production to the milk of transgenic animals carries a 5 '-promoter region derived from a naturally-derived milk protein and is consequently under the control of hormonal and tissue-specific factors. Such a promoter should therefore be most active in lactating mammary tissue. According to the current invention the promoter so utilized can be followed by a DNA sequence directing the production of a protein leader sequence which would direct the secretion of the transgenic protein across the mammary epithelium into the milk.
  • a suitable 3 '-sequence preferably also derived from a naturally secreted milk protein, may be added to improve stability of mRNA.
  • control sequences for the production of proteins in the milk of transgenic animals are those from the caprine beta casein promoter.
  • the production of transgenic animals can now be performed using a variety of methods.
  • the methods preferred by the current invention is microinjection or nuclear transfer.
  • the transcriptional promoters useful in practicing the present invention are those promoters that are preferentially activated in mammary epithelial cells, including promoters that control the genes encoding milk proteins such as caseins, beta- lacto globulin (Clark et al., (1989) BIO/TECHNOLOGY 7: 487-492), whey acid protein (Gorton et al. (1987) BIO/TECHNOLOGY 5: 1183-1187), and lactalbumin (Soulier et al., (1992) FEBS LETTS. 297: 13).
  • Casein promoters may be derived from the alpha, beta, gamma or kappa casein genes of any mammalian species; a preferred promoter is derived from the goat beta casein gene (DiTullio, (1992) BIO/TECHNOLOGY 10:74-77).
  • 50 mammary tissue may be derived from either cDNA or genomic sequences. Preferably, they are genomic in origin.
  • DNA sequence information is available for all of the mammary gland specific genes listed above, in at least one, and often several organisms. See, e.g., Richards et al., J. BiOL. CHEM. 256, 526-532 (1981) ( ⁇ -lactalbumin rat); Campbell et al, NUCLEIC ACIDS RES. 12, 8685-8697 (1984) (rat WAP); Jones et al., J. BIOL. CHEM. 260, 7042-7050 (1985) (rat ⁇ -casein); Yu-Lee & Rosen, J. BiOL. CHEM. 258, 10794-10804 (1983) (rat ⁇ -casein); Hall, BIOCHEM. J.
  • BIOCHEM. 178, 395-401 (1988) (bovine K casein); Brignon et al., FEBS LETT. 188, 48-55 (1977) (bovine ⁇ S2 casein); Jamieson et al., GENE 61, 85-90 (1987), Ivanov et al., BIOL. CHEM. Hoppe-Seyler 369, 425-429 (1988), Alexander et al., NUCLEIC ACIDS RES. 17, 6739 (1989) (bovine ⁇ lactoglobulin); Vilotte et al., BIOCHIMIE 69, 609-620 (1987) (bovine ⁇ -lactalbumin).
  • the signal sequences that are useful in accordance with this invention are milk-specific signal sequences or other signal sequences which result in the secretion of eukaryotic or prokaryotic proteins.
  • the signal sequence is selected from milk-specific signal sequences, i.e., it is from a gene which encodes a product secreted into milk.
  • the milk-specific signal sequence is related to the milk-specific promoter used in the expression system of this invention.
  • the size of the signal sequence is not critical for this invention. All that is required is that the sequence be of a sufficient size to effect secretion of the desired recombinant protein,
  • signal sequences from genes coding for caseins e.g., alpha, beta, gamma or kappa caseins, beta lactoglobulin, whey acid protein, and lactalbumin are useful in the present invention.
  • the preferred signal sequence is the goat ⁇ -casein signal sequence.
  • Signal sequences from other secreted proteins e.g., proteins secreted by liver cells, kidney cell, or pancreatic cells can also be used.
  • the efficacy with which a non-secreted protein is secreted can be enhanced by inclusion in the protein to be secreted all or part of the coding sequence of a protein which is normally secreted.
  • the entire sequence of the protein which is normally secreted is not included in the sequence of the protein but rather only a portion of the amino terminal end of the protein which is normally secreted.
  • a protein which is not normally secreted is fused (usually at its amino terminal end) to an amino terminal portion of a protein which is normally secreted.
  • the protein which is normally secreted is a protein which is normally secreted in milk.
  • proteins include proteins secreted by mammary epithelial cells, milk proteins such as caseins, beta lacto globulin, whey acid protein, and lactalbumin.
  • Casein proteins include alpha, beta, gamma or kappa casein genes of any mammalian species.
  • a preferred protein is beta casein, e.g., goat beta casein.
  • the sequences which encode the secreted protein can be derived from either cDNA or genomic sequences. Preferably, they are genomic in origin, and include one or more introns.
  • the expression system or construct, described herein, can also include a 3' untranslated region downstream of the DNA sequence coding for the non-secreted protein. This region apparently stabilizes the RNA transcript of the expression system and thus increases the yield of desired protein from the expression system.
  • 3' untranslated regions useful in the constructs of this invention are sequences that provide a poly A signal. Such sequences may be derived, e.g., from the SV40 small t antigen, the casein 3' untranslated region or other 3' untranslated sequences well known in the art.
  • the 3' untranslated region is derived from a milk specific protein.
  • the expression system or construct includes a 5' untranslated region between the promoter and the DNA sequence encoding the signal sequence.
  • Such untranslated regions can be from the same control region from which promoter is taken or can be from a different gene, e.g., they may be derived from other synthetic, semisynthetic or natural sources. Again their specific length is not critical, however, they appear to be useful in improving the level of expression.
  • the construct can also include about 10%, 20%, 30%, or more of the N- terminal coding region of the gene preferentially expressed in mammary epithelial cells.
  • the N-terminal coding region can correspond to the promoter used, e.g., a goat ⁇ -casein N-terminal coding region.
  • the above-described expression systems may be prepared using methods well known in the art. For example, various ligation techniques employing conventional linkers, restriction sites etc. may be used to good effect.
  • the expression systems of this invention are prepared as part of larger plasmids. Such preparation allows the cloning and selection of the correct constructions in an efficient manner as is well known in the art.
  • the expression systems of this invention are located between convenient restriction sites on the plasmid so that they can be easily isolated from the remaining plasmid sequences for incorporation into the desired mammal.
  • Prior art methods often include making a construct and testing it for the ability to produce a product in cultured cells prior to placing the construct in a transgenic animal. Surprisingly, the inventors have found that such a protocol may not be of predictive value in determining if a normally non-secreted protein can be secreted, e.g., in the milk of a transgenic animal. Therefore, it may be desirable to test constructs directly in transgenic animals, e.g., transgenic mice, as some constructs which fail to be secreted in CHO cells are secreted into the milk of transgenic animals.
  • the DNA constructs of the invention are introduced into the germ-line of a mammal.
  • one or several copies of the construct may be incorporated into the genome of a mammalian embryo by standard transgenic techniques known in the art.
  • Any non-human mammal can be usefully employed in this invention.
  • Mammals are defined herein as all animals, excluding humans, which have mammary glands and produce milk. Preferably, mammals that produce large volumes of milk and have long lactating periods are preferred. Preferred mammals are cows, sheep, goats, mice, oxen, camels and pigs. Of course, each of these mammals may not be as effective as the others with respect to any given expression sequence of this invention. For example, a particular milk-specific promoter or signal sequence may be more effective in one mammal than in others. However, one of skill in the art may easily make such choices by following the teachings of this invention.
  • a transgenic non- human animal is produced by introducing a transgene into the germline of the non- human animal.
  • Transgenes can be introduced into embryonal target cells at various developmental stages. Different methods are used depending on the stage of development of the embryonal target cell.
  • the specific line(s) of any animal used should, if possible, be selected for general good health, good embryo yields, good pronuclear visibility in the embryo, and good reproductive fitness.
  • the litters of transgenic mammals may be assayed after birth for the incorporation of the construct into the genome of the offspring.
  • this assay is accomplished by hybridizing a probe corresponding to the DNA sequence coding for the desired recombinant protein product or a segment thereof onto chromosomal material from the progeny.
  • Those mammalian progeny found to contain at least one copy of the construct in their genome are grown to maturity.
  • the female species of these progeny will produce the desired protein in or along with their milk.
  • the transgenic mammals may be bred to produce other transgenic progeny useful in producing the desired proteins in their milk.
  • transgenic primary cell line from either caprine, bovine, ovine, porcine or any other non-human vertebrate origin
  • transfection of the transgenic protein nucleic acid construct of interest for example, a
  • the transgene construct can either contain a selection marker (such as neomycin, kanamycin, tetracycline, puromycin, zeocin, hygromycin or any other selectable marker) or be co-transfected with a cassette able to express the selection marker in cell culture.
  • a selection marker such as neomycin, kanamycin, tetracycline, puromycin, zeocin, hygromycin or any other selectable marker
  • Transgenic females may be tested for protein secretion into milk, using any of the assay techniques that are standard in the art (e.g., Western blots or enzymatic assays).
  • the invention provides expression vectors containing a nucleic acid sequence described herein, operably linked to at least one regulatory sequence.
  • Many such vectors are commercially available, and other suitable vectors can be readily prepared by the skilled artisan.
  • "Operably linked” or “operatively linked” is intended to mean that the nucleic acid molecule is linked to a regulatory sequence in a manner which allows expression of the nucleic acid sequence by a host organism. Regulatory sequences are art recognized and are selected to produce the encoded polypeptide or protein.
  • regulatory sequence includes promoters, enhancers, and other expression control elements which are described in Goeddel, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, (Academic Press, San Diego, Calif. (1990)).
  • native regulatory sequences or regulatory sequences native to the transformed host cell can be employed.
  • the design of the expression vector may depend on such factors as the choice of the host cell to be transformed and/or the type of protein desired to be expressed.
  • the polypeptides of the present invention can be produced by ligating the cloned gene, or a portion thereof, into a vector suitable for expression in either prokaryotic cells, eukaryotic cells or both. (A LABORATORY MANUAL, 2nd Ed., ed. Sambrook et al. (Cold Spring Harbor Laboratory Press, 1989) Chapters 16 and 17)).
  • 55 integration site (although the same technique could be used with multiple integration sites) can then be used as karyoplast donors in a somatic cell nuclear transfer protocol known in the art. Following nuclear transfer, and embryo transfer to a recipient animal, and gestation, live transgenic offspring are obtained. [00164] Typically this transgenic offspring carries only one transgene integration on a specific chromosome, the other homologous chromosome not carrying an integration in the same site. Hence the transgenic offspring is hemizygous for the transgene, maintaining the current need for at least two successive breeding cycles to generate a homozygous transgenic animal.
  • Useful promoters for the expression of a target protein the mammary tissue include promoters that naturally drive the expression of mammary-specific polypeptides, such as milk proteins. These include, e.g., promoters that naturally direct expression of whey acidic protein (WAP), alpha S 1 -casein, alpha S2-casein, beta-casein, kappa-casein, beta-lactoglobulin, alpha-lactalbumin (see, e.g., Drohan et al., U.S. Patent No. 5,589,604; Meade et al., U.S. Patent No. 4, 873,316; and Karatzas et al., U.S. Patent No.
  • WAP whey acidic protein
  • Whey acidic protein (WAP; Genbank Accession No. XOl 153), the major whey protein in rodents, is expressed at high levels exclusively in the mammary gland during late pregnancy and lactation (Hobbs et al., J. BIOL. CHEM. 257:3598-3605, 1982).
  • desired mammary gland-specific promoters see, e.g., Richards et al., J. BIOL. CHEM. 256:526-532, 1981 ( ⁇ -lactalbumin rat); Campbell et al., NUCLEIC ACIDS RES.
  • inducible promoters include heat shock protein, metallothionien, and MMTV-LTR, while inducible enhancer elements include those for ecdysone, muristerone A, and tetracycline/ doxycycline.
  • mammalian cell culture systems can also be employed to express recombinant proteins.
  • mammalian expression systems include selected mouse L cells, such as thymidine kinase-negative (TK) and adenine phosphoribosul transferase-negative (APRT) cells.
  • Other examples include the COS-7 lines of monkey kidney fibroblasts, described by Gluzman, CELL 23:175 (1981), and other cell lines capable of expressing a compatible vector, for example, the C127, 3T3, CHO, HeLa and BHK cell lines.
  • yeasts there may be mentioned yeasts of the genus Saccharomyces, Kluyveromyces, Pichia, Sckwanniomyces, or
  • Hansenula Among the fungi capable of being used in the present invention, there may be mentioned more particularly Aspergillus ssp, or Trichoderma ssp.
  • Mammalian expression vectors will comprise an origin of replication, a suitable promoter and enhancer, and also any necessary ribosome binding sites, polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences, and 5' flanking non-transcribed sequences.
  • DNA sequences derived from the S V40 viral genome for example, S V40 origin, early promoter, enhancer, splice, and polyadenylation sites may be used to provide the required non-transcribed genetic elements.
  • Mammalian promoters include beta-casein, beta-lactoglobulin, whey acid promoter others include: HSV thymidine kinase, early and late SV40, LTRs from retrovirus, and mouse metallothionein-1.
  • Exemplary mammalian vectors include pWLneo, pSV2cat, pOG44, pXTl, pSG (Stratagene) pSVK3, pBPV, pMSG, and pSVL
  • the mammalian expression vector is pUCIG- MET.
  • Selectable markers include CAT (chloramphenicol transferase).
  • the methods of the invention provide a convenient, economic vehicle to produce antibodies, recombinant antibodies and other precious reagents for use in biosensor applications.
  • the methods employing the device are extremely powerful in detecting pathologic organisms and screening for specific aerosolized chemical agents.
  • the antibody array of the invention can also be used to screen samples from aqueous systems such as water supplies or in agricultural applications. Accordingly, the scope of the invention should be determined not by the embodiment(s) illustrated but by the following claims and their legal equivalents.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Pathology (AREA)
  • Medicinal Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Selon cette invention, on utilise un appareil des procédés pour produire des anticorps modifiés et calins destiné à être utilisé dans des applications de biocapteurs.
PCT/US2006/023622 2005-06-16 2006-06-16 Procede de fabrication d'anticorps humains de recombinaison destines a etre utilises dans la technologie des biocapteurs WO2006138647A2 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US11/156,831 US20060286548A1 (en) 2005-06-16 2005-06-16 Method of making recombinant human antibodies for use in biosensor technology
US11/156,831 2005-06-16
USPCT/US2005/021476 2005-06-17
PCT/US2005/021476 WO2007001264A2 (fr) 2005-06-16 2005-06-17 Procede pour produire des anticorps humain de recombinaison utilises dans le cadre d'une technologie de biocapteur

Publications (2)

Publication Number Publication Date
WO2006138647A2 true WO2006138647A2 (fr) 2006-12-28
WO2006138647A3 WO2006138647A3 (fr) 2007-05-03

Family

ID=37571259

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/023622 WO2006138647A2 (fr) 2005-06-16 2006-06-16 Procede de fabrication d'anticorps humains de recombinaison destines a etre utilises dans la technologie des biocapteurs

Country Status (1)

Country Link
WO (1) WO2006138647A2 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0063810A1 (fr) * 1981-04-29 1982-11-03 Ciba-Geigy Ag Dispositif et trousses pour les analyses immunologiques
WO1997042835A1 (fr) * 1996-05-13 1997-11-20 Genzyme Transgenics Corporation Purification de peptides de lait, actifs du point de vue biologique
US6197599B1 (en) * 1998-07-30 2001-03-06 Guorong Chin Method to detect proteins
US20020055125A1 (en) * 2000-06-05 2002-05-09 Chiron Corporation Microarrays for performing proteomic analyses

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0063810A1 (fr) * 1981-04-29 1982-11-03 Ciba-Geigy Ag Dispositif et trousses pour les analyses immunologiques
WO1997042835A1 (fr) * 1996-05-13 1997-11-20 Genzyme Transgenics Corporation Purification de peptides de lait, actifs du point de vue biologique
US6197599B1 (en) * 1998-07-30 2001-03-06 Guorong Chin Method to detect proteins
US20020055125A1 (en) * 2000-06-05 2002-05-09 Chiron Corporation Microarrays for performing proteomic analyses

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CHANG T-W: "BINDING OF CELLS TO MATRICES OF DISTINCT ANTIBODIES COATED ON SOLID SURFACE" JOURNAL OF IMMUNOLOGICAL METHODS, ELSEVIER, AMSTERDAM, NL, vol. 65, no. 1-2, 1983, pages 217-223, XP009018805 ISSN: 0022-1759 *
GOLDMAN M: "Processing Challenges for Transgenic Milk Products" BIOPROCESS INTERNATIONAL, INFORMA LIFE SCIENCES GROUP, WESTBOROUGH, MA, US, vol. 1, October 2003 (2003-10), pages 60-63, XP009074885 ISSN: 1542-6319 *
SASAKURA Y ET AL: "PROTEIN MICROARRAY SYSTEM FOR DETECTING PROTEIN-PROTEIN INTERACTIONS USING AN ANTI-HIS-TAG ANTIBODY AND FLUORESCENCE SCANNING: EFFECTS OF THE HEME REDOX STATE ON PROTEIN-PROTEIN INTERACTIONS OF HEME-REGULATED PHOSPHODIESTERASE FROM ESCHERICHIA COLI" ANALYTICAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. COLUMBUS, US, vol. 76, no. 22, 15 November 2004 (2004-11-15), pages 6521-6527, XP001225801 ISSN: 0003-2700 *

Also Published As

Publication number Publication date
WO2006138647A3 (fr) 2007-05-03

Similar Documents

Publication Publication Date Title
US20060286548A1 (en) Method of making recombinant human antibodies for use in biosensor technology
CN110249226B (zh) 评估患者样品中gfap状态的改进方法
DE69724292T2 (de) Reagenzien zum gebrauch als kalibratoren und kontrollen
JP4421900B2 (ja) モチーフ・グラフトされたハイブリッドポリペプチドおよびその使用
KR20220140568A (ko) 중증 급성 호흡기 증후군 코로나바이러스 2(SARS-CoV-2)에 대한 인간 단일클론 항체
US20110065177A1 (en) Novel Collectin
JP6824892B2 (ja) ユニバーサル抗体媒介バイオセンサー
Hoodbhoy et al. ZP2 and ZP3 traffic independently within oocytes prior to assembly into the extracellular zona pellucida
US20200115439A1 (en) HUMAN IgE ANTIBODIES BINDING TO ASPERGILLUS ALLERGENS
WO2006138647A2 (fr) Procede de fabrication d'anticorps humains de recombinaison destines a etre utilises dans la technologie des biocapteurs
WO2019210144A1 (fr) Anticorps largement neutralisants dirigés contre le virus de l'hépatite c
WO2005110433A1 (fr) Animal non humain dont l’expression de bach2 est artificiellement inhibée, et utilisation de celui-ci
JP2811089B2 (ja) イヌ×マウスヘテロハイブリドーマおよびイヌ免疫グロブリンλ鎖の定常領域をコードする遺伝子断片
WO2021096829A1 (fr) Anticorps monoclonaux humains dirigés contre le hantavirus et leurs procédés d'utilisation
JP2811086B2 (ja) ネコ×マウスヘテロハイブリドーマおよびネコ免疫グロブリンλ鎖の定常領域をコードする遺伝子断片
US20240026035A1 (en) Human ige monoclonal antibodies to antibodies to alpha-gal (galactose-a-1,3-galactose) and uses therefor
US20100329987A1 (en) Uses of Sperm Membrane Protein OBF
US20230072640A1 (en) Human monoclonal antibodies against yellow fever virus and uses therefor
WO2017047672A1 (fr) Procédé d'identification de protéine bioactive et protéine bioactive obtenue par ce procédé
EP4087590A1 (fr) Anticorps humains dirigés contre le virus de la fièvre hémorragique de crimée-congo
WO2008059991A1 (fr) Diagnostic et traitement de l'autisme en utilisant cd38
US20210041463A1 (en) Antibody screens using transgenic antigen(s)
WO2021207531A2 (fr) Anticorps monoclonaux et leurs utilisations
Hui A molecular and in silico analysis of the t complex sperm-oolemma penetration trait
Pellucida ZP2 and ZP3 Traffic Independently within

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 06785041

Country of ref document: EP

Kind code of ref document: A2