Antibodies, often monoclonal, in which the two antigen-binding sites are specific for separate ANTIGENIC DETERMINANTS. They are artificial antibodies produced by chemical crosslinking, fusion of HYBRIDOMA cells, or by molecular genetic techniques. They function as the main mediators of targeted cellular cytotoxicity and have been shown to be efficient in the targeting of drugs, toxins, radiolabeled haptens, and effector cells to diseased tissue, primarily tumors.
Immunoglobulin molecules having a specific amino acid sequence by virtue of which they interact only with the ANTIGEN (or a very similar shape) that induced their synthesis in cells of the lymphoid series (especially PLASMA CELLS).
The property of antibodies which enables them to react with some ANTIGENIC DETERMINANTS and not with others. Specificity is dependent on chemical composition, physical forces, and molecular structure at the binding site.
Antibodies produced by a single clone of cells.
Immunoglobulins produced in response to VIRAL ANTIGENS.
Immunoglobulins produced in a response to BACTERIAL ANTIGENS.
A form of antibodies consisting only of the variable regions of the heavy and light chains (FV FRAGMENTS), connected by a small linker peptide. They are less immunogenic than complete immunoglobulin and thus have potential therapeutic use.
A measure of the binding strength between antibody and a simple hapten or antigen determinant. It depends on the closeness of stereochemical fit between antibody combining sites and antigen determinants, on the size of the area of contact between them, and on the distribution of charged and hydrophobic groups. It includes the concept of "avidity," which refers to the strength of the antigen-antibody bond after formation of reversible complexes.
The production of ANTIBODIES by proliferating and differentiated B-LYMPHOCYTES under stimulation by ANTIGENS.
Antibodies that reduce or abolish some biological activity of a soluble antigen or infectious agent, usually a virus.
Local surface sites on antibodies which react with antigen determinant sites on antigens (EPITOPES.) They are formed from parts of the variable regions of FAB FRAGMENTS.
Antibodies which react with the individual structural determinants (idiotopes) on the variable region of other antibodies.
Complex of at least five membrane-bound polypeptides in mature T-lymphocytes that are non-covalently associated with one another and with the T-cell receptor (RECEPTORS, ANTIGEN, T-CELL). The CD3 complex includes the gamma, delta, epsilon, zeta, and eta chains (subunits). When antigen binds to the T-cell receptor, the CD3 complex transduces the activating signals to the cytoplasm of the T-cell. The CD3 gamma and delta chains (subunits) are separate from and not related to the gamma/delta chains of the T-cell receptor (RECEPTORS, ANTIGEN, T-CELL, GAMMA-DELTA).
Partial immunoglobulin molecules resulting from selective cleavage by proteolytic enzymes or generated through PROTEIN ENGINEERING techniques.
Test for tissue antigen using either a direct method, by conjugation of antibody with fluorescent dye (FLUORESCENT ANTIBODY TECHNIQUE, DIRECT) or an indirect method, by formation of antigen-antibody complex which is then labeled with fluorescein-conjugated anti-immunoglobulin antibody (FLUORESCENT ANTIBODY TECHNIQUE, INDIRECT). The tissue is then examined by fluorescence microscopy.
Immunoglobulins induced by antigens specific for tumors other than the normally occurring HISTOCOMPATIBILITY ANTIGENS.
Small antigenic determinants capable of eliciting an immune response only when coupled to a carrier. Haptens bind to antibodies but by themselves cannot elicit an antibody response.
Univalent antigen-binding fragments composed of one entire IMMUNOGLOBULIN LIGHT CHAIN and the amino terminal end of one of the IMMUNOGLOBULIN HEAVY CHAINS from the hinge region, linked to each other by disulfide bonds. Fab contains the IMMUNOGLOBULIN VARIABLE REGIONS, which are part of the antigen-binding site, and the first IMMUNOGLOBULIN CONSTANT REGIONS. This fragment can be obtained by digestion of immunoglobulins with the proteolytic enzyme PAPAIN.
Sites on an antigen that interact with specific antibodies.
Antibodies reactive with HIV ANTIGENS.
Immunoglobulins produced in a response to PROTOZOAN ANTIGENS.
The phenomenon of antibody-mediated target cell destruction by non-sensitized effector cells. The identity of the target cell varies, but it must possess surface IMMUNOGLOBULIN G whose Fc portion is intact. The effector cell is a "killer" cell possessing Fc receptors. It may be a lymphocyte lacking conventional B- or T-cell markers, or a monocyte, macrophage, or polynuclear leukocyte, depending on the identity of the target cell. The reaction is complement-independent.
Autoantibodies directed against various nuclear antigens including DNA, RNA, histones, acidic nuclear proteins, or complexes of these molecular elements. Antinuclear antibodies are found in systemic autoimmune diseases including systemic lupus erythematosus, Sjogren's syndrome, scleroderma, polymyositis, and mixed connective tissue disease.
Serological reactions in which an antiserum against one antigen reacts with a non-identical but closely related antigen.
Radiotherapy where cytotoxic radionuclides are linked to antibodies in order to deliver toxins directly to tumor targets. Therapy with targeted radiation rather than antibody-targeted toxins (IMMUNOTOXINS) has the advantage that adjacent tumor cells, which lack the appropriate antigenic determinants, can be destroyed by radiation cross-fire. Radioimmunotherapy is sometimes called targeted radiotherapy, but this latter term can also refer to radionuclides linked to non-immune molecules (see RADIOTHERAPY).
The major immunoglobulin isotype class in normal human serum. There are several isotype subclasses of IgG, for example, IgG1, IgG2A, and IgG2B.
Cells artificially created by fusion of activated lymphocytes with neoplastic cells. The resulting hybrid cells are cloned and produce pure MONOCLONAL ANTIBODIES or T-cell products, identical to those produced by the immunologically competent parent cell.
Use of radiolabeled antibodies for diagnostic imaging of neoplasms. Antitumor antibodies are labeled with diverse radionuclides including iodine-131, iodine-123, indium-111, or technetium-99m and injected into the patient. Images are obtained by a scintillation camera.
Manipulation of the host's immune system in treatment of disease. It includes both active and passive immunization as well as immunosuppressive therapy to prevent graft rejection.
Descriptions of specific amino acid, carbohydrate, or nucleotide sequences which have appeared in the published literature and/or are deposited in and maintained by databanks such as GENBANK, European Molecular Biology Laboratory (EMBL), National Biomedical Research Foundation (NBRF), or other sequence repositories.
Semisynthetic conjugates of various toxic molecules, including RADIOACTIVE ISOTOPES and bacterial or plant toxins, with specific immune substances such as IMMUNOGLOBULINS; MONOCLONAL ANTIBODIES; and ANTIGENS. The antitumor or antiviral immune substance carries the toxin to the tumor or infected cell where the toxin exerts its poisonous effect.
Specific molecular sites on the surface of various cells, including B-lymphocytes and macrophages, that combine with IMMUNOGLOBULIN Gs. Three subclasses exist: Fc gamma RI (the CD64 antigen, a low affinity receptor), Fc gamma RII (the CD32 antigen, a high affinity receptor), and Fc gamma RIII (the CD16 antigen, a low affinity receptor).
Inbred BALB/c mice are a strain of laboratory mice that have been selectively bred to be genetically identical to each other, making them useful for scientific research and experiments due to their consistent genetic background and predictable responses to various stimuli or treatments.
The processes triggered by interactions of ANTIBODIES with their ANTIGENS.
An iron chelating agent with properties like EDETIC ACID. DTPA has also been used as a chelator for other metals, such as plutonium.
Differentiation antigens expressed on B-lymphocytes and B-cell precursors. They are involved in regulation of B-cell proliferation.
The order of amino acids as they occur in a polypeptide chain. This is referred to as the primary structure of proteins. It is of fundamental importance in determining PROTEIN CONFORMATION.
Procedures by which protein structure and function are changed or created in vitro by altering existing or synthesizing new structural genes that direct the synthesis of proteins with sought-after properties. Such procedures may include the design of MOLECULAR MODELS of proteins using COMPUTER GRAPHICS or other molecular modeling techniques; site-specific mutagenesis (MUTAGENESIS, SITE-SPECIFIC) of existing genes; and DIRECTED MOLECULAR EVOLUTION techniques to create new genes.
A lectin and cell adhesion molecule found in B-LYMPHOCYTES. It interacts with SIALIC ACIDS and mediates signaling from B-CELL ANTIGEN RECEPTORS.
A class of immunoglobulin bearing mu chains (IMMUNOGLOBULIN MU-CHAINS). IgM can fix COMPLEMENT. The name comes from its high molecular weight and originally being called a macroglobulin.
Proteins, glycoprotein, or lipoprotein moieties on surfaces of tumor cells that are usually identified by monoclonal antibodies. Many of these are of either embryonic or viral origin.
Antibodies that react with self-antigens (AUTOANTIGENS) of the organism that produced them.
Immunoglobulins produced in a response to FUNGAL ANTIGENS.
A glycoprotein that is secreted into the luminal surface of the epithelia in the gastrointestinal tract. It is found in the feces and pancreaticobiliary secretions and is used to monitor the response to colon cancer treatment.
An immunoglobulin fragment composed of one variable domain from an IMMUNOGLOBULIN HEAVY CHAIN or IMMUNOGLOBULIN LIGHT CHAIN.
The complex formed by the binding of antigen and antibody molecules. The deposition of large antigen-antibody complexes leading to tissue damage causes IMMUNE COMPLEX DISEASES.
That region of the immunoglobulin molecule that varies in its amino acid sequence and composition, and comprises the binding site for a specific antigen. It is located at the N-terminus of the Fab fragment of the immunoglobulin. It includes hypervariable regions (COMPLEMENTARITY DETERMINING REGIONS) and framework regions.
The measurement of infection-blocking titer of ANTISERA by testing a series of dilutions for a given virus-antiserum interaction end-point, which is generally the dilution at which tissue cultures inoculated with the serum-virus mixtures demonstrate cytopathology (CPE) or the dilution at which 50% of test animals injected with serum-virus mixtures show infectivity (ID50) or die (LD50).
Lymphocytes responsible for cell-mediated immunity. Two types have been identified - cytotoxic (T-LYMPHOCYTES, CYTOTOXIC) and helper T-lymphocytes (T-LYMPHOCYTES, HELPER-INDUCER). They are formed when lymphocytes circulate through the THYMUS GLAND and differentiate to thymocytes. When exposed to an antigen, they divide rapidly and produce large numbers of new T cells sensitized to that antigen.
The phenomenon of target cell destruction by immunologically active effector cells. It may be brought about directly by sensitized T-lymphocytes or by lymphoid or myeloid "killer" cells, or it may be mediated by cytotoxic antibody, cytotoxic factor released by lymphoid cells, or complement.
Unglycosylated phosphoproteins expressed only on B-cells. They are regulators of transmembrane Ca2+ conductance and thought to play a role in B-cell activation and proliferation.
Unstable isotopes of indium that decay or disintegrate emitting radiation. In atoms with atomic weights 106-112, 113m, 114, and 116-124 are radioactive indium isotopes.
Antibodies from non-human species whose protein sequences have been modified to make them nearly identical with human antibodies. If the constant region and part of the variable region are replaced, they are called humanized. If only the constant region is modified they are called chimeric. INN names for humanized antibodies end in -zumab.
An immunoassay utilizing an antibody labeled with an enzyme marker such as horseradish peroxidase. While either the enzyme or the antibody is bound to an immunosorbent substrate, they both retain their biologic activity; the change in enzyme activity as a result of the enzyme-antibody-antigen reaction is proportional to the concentration of the antigen and can be measured spectrophotometrically or with the naked eye. Many variations of the method have been developed.
Proteins prepared by recombinant DNA technology.
Cells grown in vitro from neoplastic tissue. If they can be established as a TUMOR CELL LINE, they can be propagated in cell culture indefinitely.
Antibodies that inhibit the reaction between ANTIGEN and other antibodies or sensitized T-LYMPHOCYTES (e.g., antibodies of the IMMUNOGLOBULIN G class that compete with IGE antibodies for antigen, thereby blocking an allergic response). Blocking antibodies that bind tumors and prevent destruction of tumor cells by CYTOTOXIC T-LYMPHOCYTES have also been called enhancing antibodies. (Rosen et al., Dictionary of Immunology, 1989)
Recombinant proteins produced by the GENETIC TRANSLATION of fused genes formed by the combination of NUCLEIC ACID REGULATORY SEQUENCES of one or more genes with the protein coding sequences of one or more genes.
Transfer of immunity from immunized to non-immune host by administration of serum antibodies, or transplantation of lymphocytes (ADOPTIVE TRANSFER).
Established cell cultures that have the potential to propagate indefinitely.
Mutant mice homozygous for the recessive gene "nude" which fail to develop a thymus. They are useful in tumor studies and studies on immune responses.
Antibodies elicited in a different species from which the antigen originated. These antibodies are directed against a wide variety of interspecies-specific antigens, the best known of which are Forssman, Hanganutziu-Deicher (H-D), and Paul-Bunnell (P-B). Incidence of antibodies to these antigens--i.e., the phenomenon of heterophile antibody response--is useful in the serodiagnosis, pathogenesis, and prognosis of infection and latent infectious states as well as in cancer classification.
Antibodies that can catalyze a wide variety of chemical reactions. They are characterized by high substrate specificity and share many mechanistic features with enzymes.
The species Oryctolagus cuniculus, in the family Leporidae, order LAGOMORPHA. Rabbits are born in burrows, furless, and with eyes and ears closed. In contrast with HARES, rabbits have 22 chromosome pairs.
A cell line derived from cultured tumor cells.
Morphologic alteration of small B LYMPHOCYTES or T LYMPHOCYTES in culture into large blast-like cells able to synthesize DNA and RNA and to divide mitotically. It is induced by INTERLEUKINS; MITOGENS such as PHYTOHEMAGGLUTININS, and by specific ANTIGENS. It may also occur in vivo as in GRAFT REJECTION.
Crystallizable fragments composed of the carboxy-terminal halves of both IMMUNOGLOBULIN HEAVY CHAINS linked to each other by disulfide bonds. Fc fragments contain the carboxy-terminal parts of the heavy chain constant regions that are responsible for the effector functions of an immunoglobulin (COMPLEMENT fixation, binding to the cell membrane via FC RECEPTORS, and placental transport). This fragment can be obtained by digestion of immunoglobulins with the proteolytic enzyme PAPAIN.
A malignant disease of the B-LYMPHOCYTES in the bone marrow and/or blood.
Accumulation of a drug or chemical substance in various organs (including those not relevant to its pharmacologic or therapeutic action). This distribution depends on the blood flow or perfusion rate of the organ, the ability of the drug to penetrate organ membranes, tissue specificity, protein binding. The distribution is usually expressed as tissue to plasma ratios.
Experimental transplantation of neoplasms in laboratory animals for research purposes.
Unstable isotopes of iodine that decay or disintegrate emitting radiation. I atoms with atomic weights 117-139, except I 127, are radioactive iodine isotopes.
Mice homozygous for the mutant autosomal recessive gene "scid" which is located on the centromeric end of chromosome 16. These mice lack mature, functional lymphocytes and are thus highly susceptible to lethal opportunistic infections if not chronically treated with antibiotics. The lack of B- and T-cell immunity resembles severe combined immunodeficiency (SCID) syndrome in human infants. SCID mice are useful as animal models since they are receptive to implantation of a human immune system producing SCID-human (SCID-hu) hematochimeric mice.
Deliberate stimulation of the host's immune response. ACTIVE IMMUNIZATION involves administration of ANTIGENS or IMMUNOLOGIC ADJUVANTS. PASSIVE IMMUNIZATION involves administration of IMMUNE SERA or LYMPHOCYTES or their extracts (e.g., transfer factor, immune RNA) or transplantation of immunocompetent cell producing tissue (thymus or bone marrow).
A group of heterogeneous lymphoid tumors generally expressing one or more B-cell antigens or representing malignant transformations of B-lymphocytes.
Substances that are recognized by the immune system and induce an immune reaction.
Differentiation antigens residing on mammalian leukocytes. CD stands for cluster of differentiation, which refers to groups of monoclonal antibodies that show similar reactivity with certain subpopulations of antigens of a particular lineage or differentiation stage. The subpopulations of antigens are also known by the same CD designation.
Technique using an instrument system for making, processing, and displaying one or more measurements on individual cells obtained from a cell suspension. Cells are usually stained with one or more fluorescent dyes specific to cell components of interest, e.g., DNA, and fluorescence of each cell is measured as it rapidly transverses the excitation beam (laser or mercury arc lamp). Fluorescence provides a quantitative measure of various biochemical and biophysical properties of the cell, as well as a basis for cell sorting. Other measurable optical parameters include light absorption and light scattering, the latter being applicable to the measurement of cell size, shape, density, granularity, and stain uptake.
A class of organic compounds containing a ring structure made up of more than one kind of atom, usually carbon plus another atom. The ring structure can be aromatic or nonaromatic.
Represents 15-20% of the human serum immunoglobulins, mostly as the 4-chain polymer in humans or dimer in other mammals. Secretory IgA (IMMUNOGLOBULIN A, SECRETORY) is the main immunoglobulin in secretions.
A form of fluorescent antibody technique commonly used to detect serum antibodies and immune complexes in tissues and microorganisms in specimens from patients with infectious diseases. The technique involves formation of an antigen-antibody complex which is labeled with fluorescein-conjugated anti-immunoglobulin antibody. (From Bennington, Saunders Dictionary & Encyclopedia of Laboratory Medicine and Technology, 1984)
Serum that contains antibodies. It is obtained from an animal that has been immunized either by ANTIGEN injection or infection with microorganisms containing the antigen.
Identification of proteins or peptides that have been electrophoretically separated by blot transferring from the electrophoresis gel to strips of nitrocellulose paper, followed by labeling with antibody probes.
A collection of cloned peptides, or chemically synthesized peptides, frequently consisting of all possible combinations of amino acids making up an n-amino acid peptide.
Methods used for studying the interactions of antibodies with specific regions of protein antigens. Important applications of epitope mapping are found within the area of immunochemistry.
Autoantibodies directed against phospholipids. These antibodies are characteristically found in patients with systemic lupus erythematosus (LUPUS ERYTHEMATOSUS, SYSTEMIC;), ANTIPHOSPHOLIPID SYNDROME; related autoimmune diseases, some non-autoimmune diseases, and also in healthy individuals.
Cells propagated in vitro in special media conducive to their growth. Cultured cells are used to study developmental, morphologic, metabolic, physiologic, and genetic processes, among others.
In vivo methods of screening investigative anticancer drugs, biologic response modifiers or radiotherapies. Human tumor tissue or cells are transplanted into mice or rats followed by tumor treatment regimens. A variety of outcomes are monitored to assess antitumor effectiveness.
Electrophoresis in which a polyacrylamide gel is used as the diffusion medium.
Histochemical localization of immunoreactive substances using labeled antibodies as reagents.
New abnormal growth of tissue. Malignant neoplasms show a greater degree of anaplasia and have the properties of invasion and metastasis, compared to benign neoplasms.
Lymphoid cells concerned with humoral immunity. They are short-lived cells resembling bursa-derived lymphocytes of birds in their production of immunoglobulin upon appropriate stimulation.
Molecules found on the surface of some, but not all, B-lymphocytes, T-lymphocytes, and macrophages, which recognize and combine with the Fc (crystallizable) portion of immunoglobulin molecules.
Immunologic techniques based on the use of: (1) enzyme-antibody conjugates; (2) enzyme-antigen conjugates; (3) antienzyme antibody followed by its homologous enzyme; or (4) enzyme-antienzyme complexes. These are used histologically for visualizing or labeling tissue specimens.
The largest of polypeptide chains comprising immunoglobulins. They contain 450 to 600 amino acid residues per chain, and have molecular weights of 51-72 kDa.
Transplantation between animals of different species.
Substances elaborated by bacteria that have antigenic activity.
Antigens on surfaces of cells, including infectious or foreign cells or viruses. They are usually protein-containing groups on cell membranes or walls and may be isolated.
The sequence of PURINES and PYRIMIDINES in nucleic acids and polynucleotides. It is also called nucleotide sequence.
Polypeptide chains, consisting of 211 to 217 amino acid residues and having a molecular weight of approximately 22 kDa. There are two major types of light chains, kappa and lambda. Two Ig light chains and two Ig heavy chains (IMMUNOGLOBULIN HEAVY CHAINS) make one immunoglobulin molecule.
The process in which substances, either endogenous or exogenous, bind to proteins, peptides, enzymes, protein precursors, or allied compounds. Specific protein-binding measures are often used as assays in diagnostic assessments.
A technique using antibodies for identifying or quantifying a substance. Usually the substance being studied serves as antigen both in antibody production and in measurement of antibody by the test substance.
Elements of limited time intervals, contributing to particular results or situations.
The interaction of two or more substrates or ligands with the same binding site. The displacement of one by the other is used in quantitative and selective affinity measurements.
Anti-CD3 monoclonal antibody that exerts immunosuppressive effects by inducing peripheral T-cell depletion and modulation of the T-cell receptor complex (CD3/Ti).
Antigens expressed on the cell membrane of T-lymphocytes during differentiation, activation, and normal and neoplastic transformation. Their phenotypic characterization is important in differential diagnosis and studies of thymic ontogeny and T-cell function.
The sum of the weight of all the atoms in a molecule.
Antibodies obtained from a single clone of cells grown in mice or rats.
Multi-subunit proteins which function in IMMUNITY. They are produced by B LYMPHOCYTES from the IMMUNOGLOBULIN GENES. They are comprised of two heavy (IMMUNOGLOBULIN HEAVY CHAINS) and two light chains (IMMUNOGLOBULIN LIGHT CHAINS) with additional ancillary polypeptide chains depending on their isoforms. The variety of isoforms include monomeric or polymeric forms, and transmembrane forms (B-CELL ANTIGEN RECEPTORS) or secreted forms (ANTIBODIES). They are divided by the amino acid sequence of their heavy chains into five classes (IMMUNOGLOBULIN A; IMMUNOGLOBULIN D; IMMUNOGLOBULIN E; IMMUNOGLOBULIN G; IMMUNOGLOBULIN M) and various subclasses.
A specific immune response elicited by a specific dose of an immunologically active substance or cell in an organism, tissue, or cell.
Substances elaborated by viruses that have antigenic activity.
Immunologic method used for detecting or quantifying immunoreactive substances. The substance is identified by first immobilizing it by blotting onto a membrane and then tagging it with labeled antibodies.
Classic quantitative assay for detection of antigen-antibody reactions using a radioactively labeled substance (radioligand) either directly or indirectly to measure the binding of the unlabeled substance to a specific antibody or other receptor system. Non-immunogenic substances (e.g., haptens) can be measured if coupled to larger carrier proteins (e.g., bovine gamma-globulin or human serum albumin) capable of inducing antibody formation.
Surface ligands, usually glycoproteins, that mediate cell-to-cell adhesion. Their functions include the assembly and interconnection of various vertebrate systems, as well as maintenance of tissue integration, wound healing, morphogenic movements, cellular migrations, and metastasis.
Serum glycoproteins participating in the host defense mechanism of COMPLEMENT ACTIVATION that creates the COMPLEMENT MEMBRANE ATTACK COMPLEX. Included are glycoproteins in the various pathways of complement activation (CLASSICAL COMPLEMENT PATHWAY; ALTERNATIVE COMPLEMENT PATHWAY; and LECTIN COMPLEMENT PATHWAY).
The insertion of recombinant DNA molecules from prokaryotic and/or eukaryotic sources into a replicating vehicle, such as a plasmid or virus vector, and the introduction of the resultant hybrid molecules into recipient cells without altering the viability of those cells.
Substances that inhibit or prevent the proliferation of NEOPLASMS.
Combinations of diagnostic or therapeutic substances linked with specific immune substances such as IMMUNOGLOBULINS; MONOCLONAL ANTIBODIES; or ANTIGENS. Often the diagnostic or therapeutic substance is a radionuclide. These conjugates are useful tools for specific targeting of DRUGS and RADIOISOTOPES in the CHEMOTHERAPY and RADIOIMMUNOTHERAPY of certain cancers.
Binary classification measures to assess test results. Sensitivity or recall rate is the proportion of true positives. Specificity is the probability of correctly determining the absence of a condition. (From Last, Dictionary of Epidemiology, 2d ed)
Serologic tests based on inactivation of complement by the antigen-antibody complex (stage 1). Binding of free complement can be visualized by addition of a second antigen-antibody system such as red cells and appropriate red cell antibody (hemolysin) requiring complement for its completion (stage 2). Failure of the red cells to lyse indicates that a specific antigen-antibody reaction has taken place in stage 1. If red cells lyse, free complement is present indicating no antigen-antibody reaction occurred in stage 1.
A cell surface protein-tyrosine kinase receptor that is overexpressed in a variety of ADENOCARCINOMAS. It has extensive homology to and heterodimerizes with the EGF RECEPTOR, the ERBB-3 RECEPTOR, and the ERBB-4 RECEPTOR. Activation of the erbB-2 receptor occurs through heterodimer formation with a ligand-bound erbB receptor family member.
Carbohydrate antigens expressed by malignant tissue. They are useful as tumor markers and are measured in the serum by means of a radioimmunoassay employing monoclonal antibodies.
Partial proteins formed by partial hydrolysis of complete proteins or generated through PROTEIN ENGINEERING techniques.
Sensitive tests to measure certain antigens, antibodies, or viruses, using their ability to agglutinate certain erythrocytes. (From Stedman, 26th ed)
Serologic tests in which a known quantity of antigen is added to the serum prior to the addition of a red cell suspension. Reaction result is expressed as the smallest amount of antigen which causes complete inhibition of hemagglutination.
DNA molecules capable of autonomous replication within a host cell and into which other DNA sequences can be inserted and thus amplified. Many are derived from PLASMIDS; BACTERIOPHAGES; or VIRUSES. They are used for transporting foreign genes into recipient cells. Genetic vectors possess a functional replicator site and contain GENETIC MARKERS to facilitate their selective recognition.
Autoantibodies directed against cytoplasmic constituents of POLYMORPHONUCLEAR LEUKOCYTES and/or MONOCYTES. They are used as specific markers for GRANULOMATOSIS WITH POLYANGIITIS and other diseases, though their pathophysiological role is not clear. ANCA are routinely detected by indirect immunofluorescence with three different patterns: c-ANCA (cytoplasmic), p-ANCA (perinuclear), and atypical ANCA.
Members of the class of compounds composed of AMINO ACIDS joined together by peptide bonds between adjacent amino acids into linear, branched or cyclical structures. OLIGOPEPTIDES are composed of approximately 2-12 amino acids. Polypeptides are composed of approximately 13 or more amino acids. PROTEINS are linear polypeptides that are normally synthesized on RIBOSOMES.
EPIDEMIOLOGIC STUDIES based on the detection through serological testing of characteristic change in the serum level of specific ANTIBODIES. Latent subclinical infections and carrier states can thus be detected in addition to clinically overt cases.
Inbred C57BL mice are a strain of laboratory mice that have been produced by many generations of brother-sister matings, resulting in a high degree of genetic uniformity and homozygosity, making them widely used for biomedical research, including studies on genetics, immunology, cancer, and neuroscience.
A cell surface receptor involved in regulation of cell growth and differentiation. It is specific for EPIDERMAL GROWTH FACTOR and EGF-related peptides including TRANSFORMING GROWTH FACTOR ALPHA; AMPHIREGULIN; and HEPARIN-BINDING EGF-LIKE GROWTH FACTOR. The binding of ligand to the receptor causes activation of its intrinsic tyrosine kinase activity and rapid internalization of the receptor-ligand complex into the cell.
Unique genetically-controlled determinants present on ANTIBODIES whose specificity is limited to a single group of proteins (e.g., another antibody molecule or an individual myeloma protein). The idiotype appears to represent the antigenicity of the antigen-binding site of the antibody and to be genetically codetermined with it. The idiotypic determinants have been precisely located to the IMMUNOGLOBULIN VARIABLE REGION of both immunoglobin polypeptide chains.
Techniques used to demonstrate or measure an immune response, and to identify or measure antigens using antibodies.
A chromatographic technique that utilizes the ability of biological molecules to bind to certain ligands specifically and reversibly. It is used in protein biochemistry. (McGraw-Hill Dictionary of Scientific and Technical Terms, 4th ed)
Membrane antigens associated with maturation stages of B-lymphocytes, often expressed in tumors of B-cell origin.
Ribosome inactivating proteins consisting of only the toxic A subunit, which is a polypeptide of around 30 kDa.
A species of gram-negative, facultatively anaerobic, rod-shaped bacteria (GRAM-NEGATIVE FACULTATIVELY ANAEROBIC RODS) commonly found in the lower part of the intestine of warm-blooded animals. It is usually nonpathogenic, but some strains are known to produce DIARRHEA and pyogenic infections. Pathogenic strains (virotypes) are classified by their specific pathogenic mechanisms such as toxins (ENTEROTOXIGENIC ESCHERICHIA COLI), etc.
Techniques for removal by adsorption and subsequent elution of a specific antibody or antigen using an immunosorbent containing the homologous antigen or antibody.
Cytolytic lymphocytes with the unique capacity of killing natural killer (NK)-resistant fresh tumor cells. They are INTERLEUKIN-2-activated NK cells that have no MAJOR HISTOCOMPATIBILITY COMPLEX restriction or need for antigen stimulation. LAK cells are used for ADOPTIVE IMMUNOTHERAPY in cancer patients.
Hoofed mammals with four legs, a big-lipped snout, and a humped back belonging to the family Camelidae.
An encapsulated lymphatic organ through which venous blood filters.
The rate dynamics in chemical or physical systems.

Immunotherapy of human tumors with T-cell-activating bispecific antibodies: stimulation of cytotoxic pathways in vivo. (1/429)

Bispecific monoclonal antibodies (Bi-mAbs) specific for a tumor-associated antigen and the CD3 or CD28 antigen on T lymphocytes represent one of the most successful experimental strategies for the immunotherapy of cancer. We report that the in vivo administration of both alpha-CD3/CD30 and alpha-CD28/CD30 Bi-mAbs results in the specific activation of xenotransplanted, resting human T cells infiltrating the CD30-positive Hodgkin's tumor. Bi-mAb treatment resulted in enhanced expression of cytokines such as interleukin 1beta, interleukin 2, tumor necrosis factor type alpha, and activation markers including Ki-67, CD25, and CD45RO in tumor-infiltrating lymphocytes. This antigen-dependent, local T-cell stimulation led to the activation of the cytolytic machinery in T lymphocytes, determined by the up-regulation of mRNA-encoding perforin and the cytotoxic serine-esterases granzymes A and B. The Bi-mAb-induced generation of CTLs depended on the presence of the CD30 antigen and the combined application of both Bi-mAbs. Our findings suggest that the combined application of T-cell-activating Bi-mAbs is able to achieve a tumor site-specific activation of the T-cell cytolytic machinery in vivo. The fact that these cytotoxic cells do not home in tumor-associated antigen-negative tissue and do not enter circulation might explain our previous observations (C. Renner et al., Blood, 87: 2930-2937, 1996) of a high cure rate in preclinical models even at an advanced stage of disease.  (+info)

Dual specificity antibodies using a double-stranded oligonucleotide bridge. (2/429)

The covalent conjugation of oligonucleotides to antibody Fab' fragments was optimized by using oligonucleotides modified with a hexaethylene linker arm bearing three amino groups. One oligonucleotide was coupled to antibody of one specificity and a complementary oligonucleotide to antibody of a second specificity. The antibodies were then allowed to hybridize by base pairing of the complementary nucleotide sequences and the generation of bispecific antibody was analyzed on SDS-PAGE and confirmed using BIAcore analysis. The strategy of complementary oligonucleotide-linked bispecific molecules is not limited to antibodies but is applicable to linking any two molecules of different characteristics.  (+info)

An effective strategy of human tumor vaccine modification by coupling bispecific costimulatory molecules. (3/429)

A new, generally applicable procedure is described for the introduction of defined costimulatory molecules into human cancer cells to increase their T-cell stimulatory capacity. The procedure involves infection with Newcastle disease virus to mediate the cell surface binding of costimulatory molecules (e.g., specially designed bispecific antibodies (bsAb)). The modification is independent of tumor cell proliferation and laborious recombinant gene technology and can be applied directly to freshly isolated and gamma-irradiated patient-derived tumor cells as an autologous cancer vaccine. Following the infection of tumor cells with a nonvirulent strain of Newcastle disease virus, the cells are washed and then further modified by coincubation with bsAbs, which attach with one arm to the viral hemagglutinin-neuraminidase (HN) molecule on the infected tumor cells. The second specificity of one bsAb (bs HN x CD28) is directed against CD28 to augment antitumor T-cell responses by selectively channeling positive costimulatory signals via the CD28 pathway. A second bsAb (bs HN x CD3) was produced to deliver T-cell receptor-mediated signals either alone (bsCD3 vaccine) or in combination with anti-CD28 (bsCD3 vaccine plus bsCD28 vaccine). In human T-cell stimulation studies in vitro, the bsCD28 vaccine caused an up-regulation of early (CD69) and late (CD25) T-cell activation markers on CD4 and CD8 T lymphocytes from either normal healthy donors or cancer patients (autologous system) and induced tumor cytostasis in nonmodified bystander tumor cells. In addition, in combination with the bsCD3 vaccine, augmented antitumor cytotoxicity and T-cell proliferative responses were observed. This tumor vaccine modification procedure is highly specific, quick, economic, and has a broad range of clinical applications.  (+info)

Carcinoembryonic antigen (CEA)-specific T-cell activation in colon carcinoma induced by anti-CD3 x anti-CEA bispecific diabodies and B7 x anti-CEA bispecific fusion proteins. (4/429)

Two bispecific recombinant molecules, an anti-CD3 x anti-carcinoembryogenic antigen (CEA) diabody and a B7 x anti-CEA fusion protein, were tested for their capacity to specifically activate T cells in the presence of CEA-expressing colon carcinoma cells. T-cell activation by the anti-CD3 x anti-CEA diabody required close contact to CEA-positive cells and resulted in diabody-mediated cytotoxicity against the target cells. Additionally, CD28-mediated costimulation in combination with anti-CD3 x anti-CEA diabodies induced activation of autologous T cells in CEA-positive primary colon carcinoma specimens, as determined by flow cytometry. The high specificity of the bispecific diabody approach could be further enhanced by the use of B7 x anti-CEA fusion proteins because the costimulatory CD28-signaling to the T cells strictly depended on the expression of CEA on the target cells. We demonstrate that displaying engagement sites for the T-cell antigens CD3 and CD28 on the surface of colon carcinoma cells is a suitable way to activate and retarget T cells in a highly tumor-specific manner. For clinical purposes, B7 x anti-tumor-associated antigen (TAA) fusion proteins, which are equally effective but more specific compared with anti-CD28 monoclonal anti-bodies, thus may improve the tumor specificity of anti-CD3 x anti-TAA bispecific antibodies. Furthermore, B7-negative tumors can be converted into B7-positive tumors by B7 x anti-TAA fusion proteins without the need for B7 gene transfer to the malignant cells.  (+info)

Two-step targeting and dosimetry for small cell lung cancer xenograft with anti-NCAM/antihistamine bispecific antibody and radioiodinated bivalent hapten. (5/429)

The "affinity enhancement system," a two-step targeting technique using bispecific antibody and radiolabeled bivalent hapten, has been reported to be useful for carcinoembryonic antigen-expressing tumors. The purpose of this study was to evaluate the efficacy of this method for targeting human small cell lung cancer using an antineural cell adhesion molecule antibody. METHODS: Antineural cell adhesion molecule/antihistamine bispecific antibody NK1NBL1-679 was prepared by coupling an equimolecular quantity of a Fab' fragment of NK1NBL1 to a Fab fragment of antihistamine 679. Athymic mice inoculated with NCI-H69 small cell lung cancer cells expressing neural cell adhesion molecule were administered bispecific antibody and then 48 h later 125I-labeled bivalent histamine hapten. 125I-labeled intact NK1NBL1 was injected into other groups of mice. Biodistributions were examined as a function of time. RESULTS: In mice of the two-step targeting, tumor uptake was 2.5 +/- 0.2, 3.2 +/- 0.4, 6.4 +/- 2.0, 7.2 +/- 2.7, 6.1 +/- 2.1 and 2.2 +/- 0.4 %ID/g at 5, 30 min, 5, 24, 48 and 96 h, and tumor-to-blood, tumor-to-liver and tumor-to-kidney ratios were 1.4 +/- 1.1, 10.8 +/- 13.2 and 4.6 +/- 4.7, respectively, at 5 h, whereas 125I-labeled NK1NBL1 showed a tumor uptake of 5.7 +/- 0.4 %ID/g and tumor-to-blood, tumor-to-liver and tumor-to-kidney ratios of 0.3 +/- 0.1, 1.1 +/- 0.2 and 0.9 +/- 0.1, respectively, at 5 h. These results were confirmed by autoradiographic studies, which demonstrated clear tumor-to-normal tissue contrast. Dosimetry showed that the affinity enhancement system could enhance the therapeutic potential of the antineural cell adhesion molecule antibody NK1NBL1. CONCLUSION: This two-step targeting method seems promising for the diagnosis and therapy of small cell lung cancer.  (+info)

Expression and characterization of bispecific single-chain Fv fragments produced in transgenic plants. (6/429)

We describe the expression of the bispecific antibody biscFv2429 in transgenic suspension culture cells and tobacco plants. biscFv2429 consists of two single-chain antibodies, scFv24 and scFv29, connected by the Trichoderma reesi cellobiohydrolase I linker. biscFv2429 binds two epitopes of tobacco mosaic virus (TMV): the scFv24 domain recognizes neotopes of intact virions, and the scFv29 domain recognizes a cryptotope of the TMV coat protein monomer. biscFv2429 was functionally expressed either in the cytosol (biscFv2429-cyt) or targeted to the apoplast using a murine leader peptide sequence (biscFv2429-apoplast). A third construct contained the C-terminal KDEL sequence for retention in the ER (biscFv2429-KDEL). Levels of cytoplasmic biscFv2429 expression levels were low. The highest levels of antibody expression were for apoplast-targeted biscFv2429-apoplast and ER-retained biscFv2429-KDEL that reached a maximum expression level of 1.65% total soluble protein in transgenic plants. Plant-expressed biscFv2429 retained both epitope specificities, and bispecificity and bivalency were confirmed by ELISA and surface plasmon resonance analysis. This study establishes plant cells as an expression system for bispecific single-chain antibodies for use in medical and biological applications.  (+info)

Novel tetravalent and bispecific IgG-like antibody molecules combining single-chain diabodies with the immunoglobulin gamma1 Fc or CH3 region. (7/429)

Although bispecific IgG molecules have been successfully applied for antibody-mediated immunotherapy of tumours, applicability is hampered by the difficulties associated with their generation. In the present study, we have used a bispecific single-chain diabody (scDb) directed against carcinoembryonic antigen and Escherichia coli beta-galactosidase as a model to generate bispecific IgG-like antibody molecules. We show that the fusion of this single-chain diabody to the Fc (scDb-Fc) or CH3 (scDb-CH3) region of the human immunoglobulin gamma1 chain results in the expression of dimeric fusion proteins exhibiting four functional antigen binding sites with increased functional affinity. This strategy represents a new and convenient way to generate IgG-like multivalent and bispecific molecules that are efficiently secreted from mammalian cells.  (+info)

Simultaneous activation of T cells and accessory cells by a new class of intact bispecific antibody results in efficient tumor cell killing. (8/429)

Bispecific Abs (bsAb) are promising immunological tools for the elimination of tumor cells in minimal residual disease situations. In principle, they target an Ag on tumor cells and recruit one class of effector cell. Because immune reactions in vivo are more complex and are mediated by different classes of effector cell, we argue that conventional bsAb might not yield optimal immune responses at the tumor site. We therefore constructed a bsAb that combines the two potent effector subclasses mouse IgG2a and rat IgG2b. This bispecific molecule not only recruits T cells via its one binding arm, but simultaneously activates FcgammaR+ accessory cells via its Fc region. We demonstrate here that the activation of both T lymphocytes and accessory cells leads to production of immunomodulating cytokines like IL-1beta, IL-2, IL-6, IL-12, and DC-CK1. Thus this new class of bsAb elicits excellent antitumor activity in vitro even without the addition of exogenous IL-2, and therefore represents a totally self-supporting system.  (+info)

Bispecific antibodies are a type of artificial protein that have been engineered to recognize and bind to two different antigens simultaneously. They are created by combining two separate antibody molecules, each with a unique binding site, into a single entity. This allows the bispecific antibody to link two cells or proteins together, bringing them into close proximity and facilitating various biological processes.

In the context of medicine and immunotherapy, bispecific antibodies are being investigated as a potential treatment for cancer and other diseases. For example, a bispecific antibody can be designed to recognize a specific tumor-associated antigen on the surface of cancer cells, while also binding to a component of the immune system, such as a T cell. This brings the T cell into close contact with the cancer cell, activating the immune system and triggering an immune response against the tumor.

Bispecific antibodies have several potential advantages over traditional monoclonal antibodies, which only recognize a single antigen. By targeting two different epitopes or antigens, bispecific antibodies can increase the specificity and affinity of the interaction, reducing off-target effects and improving therapeutic efficacy. Additionally, bispecific antibodies can bring together multiple components of the immune system, amplifying the immune response and enhancing the destruction of cancer cells.

Overall, bispecific antibodies represent a promising new class of therapeutics that have the potential to revolutionize the treatment of cancer and other diseases. However, further research is needed to fully understand their mechanisms of action and optimize their clinical use.

Antibodies are proteins produced by the immune system in response to the presence of a foreign substance, such as a bacterium or virus. They are capable of identifying and binding to specific antigens (foreign substances) on the surface of these invaders, marking them for destruction by other immune cells. Antibodies are also known as immunoglobulins and come in several different types, including IgA, IgD, IgE, IgG, and IgM, each with a unique function in the immune response. They are composed of four polypeptide chains, two heavy chains and two light chains, that are held together by disulfide bonds. The variable regions of the heavy and light chains form the antigen-binding site, which is specific to a particular antigen.

Antibody specificity refers to the ability of an antibody to bind to a specific epitope or antigenic determinant on an antigen. Each antibody has a unique structure that allows it to recognize and bind to a specific region of an antigen, typically a small portion of the antigen's surface made up of amino acids or sugar residues. This highly specific binding is mediated by the variable regions of the antibody's heavy and light chains, which form a pocket that recognizes and binds to the epitope.

The specificity of an antibody is determined by its unique complementarity-determining regions (CDRs), which are loops of amino acids located in the variable domains of both the heavy and light chains. The CDRs form a binding site that recognizes and interacts with the epitope on the antigen. The precise fit between the antibody's binding site and the epitope is critical for specificity, as even small changes in the structure of either can prevent binding.

Antibody specificity is important in immune responses because it allows the immune system to distinguish between self and non-self antigens. This helps to prevent autoimmune reactions where the immune system attacks the body's own cells and tissues. Antibody specificity also plays a crucial role in diagnostic tests, such as ELISA assays, where antibodies are used to detect the presence of specific antigens in biological samples.

Monoclonal antibodies are a type of antibody that are identical because they are produced by a single clone of cells. They are laboratory-produced molecules that act like human antibodies in the immune system. They can be designed to attach to specific proteins found on the surface of cancer cells, making them useful for targeting and treating cancer. Monoclonal antibodies can also be used as a therapy for other diseases, such as autoimmune disorders and inflammatory conditions.

Monoclonal antibodies are produced by fusing a single type of immune cell, called a B cell, with a tumor cell to create a hybrid cell, or hybridoma. This hybrid cell is then able to replicate indefinitely, producing a large number of identical copies of the original antibody. These antibodies can be further modified and engineered to enhance their ability to bind to specific targets, increase their stability, and improve their effectiveness as therapeutic agents.

Monoclonal antibodies have several mechanisms of action in cancer therapy. They can directly kill cancer cells by binding to them and triggering an immune response. They can also block the signals that promote cancer growth and survival. Additionally, monoclonal antibodies can be used to deliver drugs or radiation directly to cancer cells, increasing the effectiveness of these treatments while minimizing their side effects on healthy tissues.

Monoclonal antibodies have become an important tool in modern medicine, with several approved for use in cancer therapy and other diseases. They are continuing to be studied and developed as a promising approach to treating a wide range of medical conditions.

Antibodies, viral are proteins produced by the immune system in response to an infection with a virus. These antibodies are capable of recognizing and binding to specific antigens on the surface of the virus, which helps to neutralize or destroy the virus and prevent its replication. Once produced, these antibodies can provide immunity against future infections with the same virus.

Viral antibodies are typically composed of four polypeptide chains - two heavy chains and two light chains - that are held together by disulfide bonds. The binding site for the antigen is located at the tip of the Y-shaped structure, formed by the variable regions of the heavy and light chains.

There are five classes of antibodies in humans: IgA, IgD, IgE, IgG, and IgM. Each class has a different function and is distributed differently throughout the body. For example, IgG is the most common type of antibody found in the bloodstream and provides long-term immunity against viruses, while IgA is found primarily in mucous membranes and helps to protect against respiratory and gastrointestinal infections.

In addition to their role in the immune response, viral antibodies can also be used as diagnostic tools to detect the presence of a specific virus in a patient's blood or other bodily fluids.

Bacterial antibodies are a type of antibodies produced by the immune system in response to an infection caused by bacteria. These antibodies are proteins that recognize and bind to specific antigens on the surface of the bacterial cells, marking them for destruction by other immune cells. Bacterial antibodies can be classified into several types based on their structure and function, including IgG, IgM, IgA, and IgE. They play a crucial role in the body's defense against bacterial infections and provide immunity to future infections with the same bacteria.

Single-chain antibodies (scFvs) are small, artificial protein molecules that contain the antigen-binding sites of immunoglobulins. They are formed by linking the variable regions of the heavy and light chains of an antibody via a flexible peptide linker, creating a single polypeptide chain. This design allows scFvs to maintain the specificity of traditional antibodies while being significantly smaller in size, more stable, and easier to produce. They have various applications in research, diagnostics, and therapeutics, including targeted drug delivery, tumor imaging, and the development of novel therapies for cancer and other diseases.

Antibody affinity refers to the strength and specificity of the interaction between an antibody and its corresponding antigen at a molecular level. It is a measure of how strongly and selectively an antibody binds to its target antigen. A higher affinity indicates a more stable and specific binding, while a lower affinity suggests weaker and less specific interactions. Affinity is typically measured in terms of the dissociation constant (Kd), which describes the concentration of antigen needed to achieve half-maximal binding to an antibody. Generally, a smaller Kd value corresponds to a higher affinity, indicating a tighter and more selective bond. This parameter is crucial in the development of diagnostic and therapeutic applications, such as immunoassays and targeted therapies, where high-affinity antibodies are preferred for improved sensitivity and specificity.

Antibody formation, also known as humoral immune response, is the process by which the immune system produces proteins called antibodies in response to the presence of a foreign substance (antigen) in the body. This process involves several steps:

1. Recognition: The antigen is recognized and bound by a type of white blood cell called a B lymphocyte or B cell, which then becomes activated.
2. Differentiation: The activated B cell undergoes differentiation to become a plasma cell, which is a type of cell that produces and secretes large amounts of antibodies.
3. Antibody production: The plasma cells produce and release antibodies, which are proteins made up of four polypeptide chains (two heavy chains and two light chains) arranged in a Y-shape. Each antibody has two binding sites that can recognize and bind to specific regions on the antigen called epitopes.
4. Neutralization or elimination: The antibodies bind to the antigens, neutralizing them or marking them for destruction by other immune cells. This helps to prevent the spread of infection and protect the body from harmful substances.

Antibody formation is an important part of the adaptive immune response, which allows the body to specifically recognize and respond to a wide variety of pathogens and foreign substances.

Neutralizing antibodies are a type of antibody that defends against pathogens such as viruses or bacteria by neutralizing their ability to infect cells. They do this by binding to specific regions on the surface proteins of the pathogen, preventing it from attaching to and entering host cells. This renders the pathogen ineffective and helps to prevent or reduce the severity of infection. Neutralizing antibodies can be produced naturally in response to an infection or vaccination, or they can be generated artificially for therapeutic purposes.

A binding site on an antibody refers to the specific region on the surface of the antibody molecule that can recognize and bind to a specific antigen. Antibodies are proteins produced by the immune system in response to the presence of foreign substances called antigens. They have two main functions: to neutralize the harmful effects of antigens and to help eliminate them from the body.

The binding site of an antibody is located at the tips of its Y-shaped structure, formed by the variable regions of the heavy and light chains of the antibody molecule. These regions contain unique amino acid sequences that determine the specificity of the antibody for a particular antigen. The binding site can recognize and bind to a specific epitope or region on the antigen, forming an antigen-antibody complex.

The binding between the antibody and antigen is highly specific and depends on non-covalent interactions such as hydrogen bonds, van der Waals forces, and electrostatic attractions. This interaction plays a crucial role in the immune response, as it allows the immune system to recognize and eliminate pathogens and other foreign substances from the body.

Anti-idiotypic antibodies are a type of immune protein that recognizes and binds to the unique identifying region (idiotype) of another antibody. These antibodies are produced by the immune system as part of a regulatory feedback mechanism, where they can modulate or inhibit the activity of the original antibody. They have been studied for their potential use in immunotherapy and vaccine development.

CD3 antigens are a group of proteins found on the surface of T-cells, which are a type of white blood cell that plays a central role in the immune response. The CD3 antigens are composed of several different subunits (ε, δ, γ, and α) that associate to form the CD3 complex, which is involved in T-cell activation and signal transduction.

The CD3 complex is associated with the T-cell receptor (TCR), which recognizes and binds to specific antigens presented by antigen-presenting cells. When the TCR binds to an antigen, it triggers a series of intracellular signaling events that lead to T-cell activation and the initiation of an immune response.

CD3 antigens are important targets for immunotherapy in some diseases, such as certain types of cancer. For example, monoclonal antibodies that target CD3 have been developed to activate T-cells and enhance their ability to recognize and destroy tumor cells. However, CD3-targeted therapies can also cause side effects, such as cytokine release syndrome, which can be serious or life-threatening in some cases.

Immunoglobulin fragments refer to the smaller protein units that are formed by the digestion or break-down of an intact immunoglobulin, also known as an antibody. Immunoglobulins are large Y-shaped proteins produced by the immune system to identify and neutralize foreign substances such as pathogens or toxins. They consist of two heavy chains and two light chains, held together by disulfide bonds.

The digestion or break-down of an immunoglobulin can occur through enzymatic cleavage, which results in the formation of distinct fragments. The most common immunoglobulin fragments are:

1. Fab (Fragment, antigen binding) fragments: These are formed by the digestion of an intact immunoglobulin using the enzyme papain. Each Fab fragment contains a single antigen-binding site, consisting of a portion of one heavy chain and one light chain. The Fab fragments retain their ability to bind to specific antigens.
2. Fc (Fragment, crystallizable) fragments: These are formed by the digestion of an intact immunoglobulin using the enzyme pepsin or through the natural breakdown process in the body. The Fc fragment contains the constant region of both heavy chains and is responsible for effector functions such as complement activation, binding to Fc receptors on immune cells, and antibody-dependent cellular cytotoxicity (ADCC).

These immunoglobulin fragments play crucial roles in various immune responses and diagnostic applications. For example, Fab fragments can be used in immunoassays for the detection of specific antigens, while Fc fragments can mediate effector functions that help eliminate pathogens or damaged cells from the body.

The Fluorescent Antibody Technique (FAT) is a type of immunofluorescence assay used in laboratory medicine and pathology for the detection and localization of specific antigens or antibodies in tissues, cells, or microorganisms. In this technique, a fluorescein-labeled antibody is used to selectively bind to the target antigen or antibody, forming an immune complex. When excited by light of a specific wavelength, the fluorescein label emits light at a longer wavelength, typically visualized as green fluorescence under a fluorescence microscope.

The FAT is widely used in diagnostic microbiology for the identification and characterization of various bacteria, viruses, fungi, and parasites. It has also been applied in the diagnosis of autoimmune diseases and certain cancers by detecting specific antibodies or antigens in patient samples. The main advantage of FAT is its high sensitivity and specificity, allowing for accurate detection and differentiation of various pathogens and disease markers. However, it requires specialized equipment and trained personnel to perform and interpret the results.

'Antibodies, Neoplasm' is a medical term that refers to abnormal antibodies produced by neoplastic cells, which are cells that have undergone uncontrolled division and form a tumor or malignancy. These antibodies can be produced in large quantities and may have altered structures or functions compared to normal antibodies.

Neoplastic antibodies can arise from various types of malignancies, including leukemias, lymphomas, and multiple myeloma. In some cases, these abnormal antibodies can interfere with the normal functioning of the immune system and contribute to the progression of the disease.

In addition, neoplastic antibodies can also be used as tumor markers for diagnostic purposes. For example, certain types of monoclonal gammopathy, such as multiple myeloma, are characterized by the overproduction of a single type of immunoglobulin, which can be detected in the blood or urine and used to monitor the disease.

Overall, 'Antibodies, Neoplasm' is a term that encompasses a wide range of abnormal antibodies produced by neoplastic cells, which can have significant implications for both the diagnosis and treatment of malignancies.

A hapten is a small molecule that can elicit an immune response only when it is attached to a larger carrier protein. On its own, a hapten is too small to be recognized by the immune system as a foreign substance. However, when it binds to a carrier protein, it creates a new antigenic site that can be detected by the immune system. This process is known as haptenization.

Haptens are important in the study of immunology and allergies because they can cause an allergic response when they bind to proteins in the body. For example, certain chemicals found in cosmetics, drugs, or industrial products can act as haptens and trigger an allergic reaction when they come into contact with the skin or mucous membranes. The resulting immune response can cause symptoms such as rash, itching, or inflammation.

Haptens can also be used in the development of vaccines and diagnostic tests, where they are attached to carrier proteins to stimulate an immune response and produce specific antibodies that can be measured or used for therapy.

Immunoglobulin (Ig) Fab fragments are the antigen-binding portions of an antibody that result from the digestion of the whole antibody molecule by enzymes such as papain. An antibody, also known as an immunoglobulin, is a Y-shaped protein produced by the immune system to identify and neutralize foreign substances like bacteria, viruses, or toxins. The antibody has two identical antigen-binding sites, located at the tips of the two shorter arms, which can bind specifically to a target antigen.

Fab fragments are formed when an antibody is cleaved by papain, resulting in two Fab fragments and one Fc fragment. Each Fab fragment contains one antigen-binding site, composed of a variable region (Fv) and a constant region (C). The Fv region is responsible for the specificity and affinity of the antigen binding, while the C region contributes to the effector functions of the antibody.

Fab fragments are often used in various medical applications, such as immunodiagnostics and targeted therapies, due to their ability to bind specifically to target antigens without triggering an immune response or other effector functions associated with the Fc region.

An epitope is a specific region on the surface of an antigen (a molecule that can trigger an immune response) that is recognized by an antibody, B-cell receptor, or T-cell receptor. It is also commonly referred to as an antigenic determinant. Epitopes are typically composed of linear amino acid sequences or conformational structures made up of discontinuous amino acids in the antigen. They play a crucial role in the immune system's ability to differentiate between self and non-self molecules, leading to the targeted destruction of foreign substances like viruses and bacteria. Understanding epitopes is essential for developing vaccines, diagnostic tests, and immunotherapies.

HIV antibodies are proteins produced by the immune system in response to the presence of HIV (Human Immunodeficiency Virus) in the body. These antibodies are designed to recognize and bind to specific parts of the virus, known as antigens, in order to neutralize or eliminate it.

There are several types of HIV antibodies that can be produced, including:

1. Anti-HIV-1 and anti-HIV-2 antibodies: These are antibodies that specifically target the HIV-1 and HIV-2 viruses, respectively.
2. Antibodies to HIV envelope proteins: These antibodies recognize and bind to the outer envelope of the virus, which is covered in glycoprotein spikes that allow the virus to attach to and enter host cells.
3. Antibodies to HIV core proteins: These antibodies recognize and bind to the interior of the viral particle, where the genetic material of the virus is housed.

The presence of HIV antibodies in the blood can be detected through a variety of tests, including enzyme-linked immunosorbent assay (ELISA) and Western blot. A positive test result for HIV antibodies indicates that an individual has been infected with the virus, although it may take several weeks or months after infection for the antibodies to become detectable.

Antibodies, protozoan, refer to the immune system's response to an infection caused by a protozoan organism. Protozoa are single-celled microorganisms that can cause various diseases in humans, such as malaria, giardiasis, and toxoplasmosis.

When the body is infected with a protozoan, the immune system responds by producing specific proteins called antibodies. Antibodies are produced by a type of white blood cell called a B-cell, and they recognize and bind to specific antigens on the surface of the protozoan organism.

There are five main types of antibodies: IgA, IgD, IgE, IgG, and IgM. Each type of antibody has a different role in the immune response. For example, IgG is the most common type of antibody and provides long-term immunity to previously encountered pathogens. IgM is the first antibody produced in response to an infection and is important for activating the complement system, which helps to destroy the protozoan organism.

Overall, the production of antibodies against protozoan organisms is a critical part of the immune response and helps to protect the body from further infection.

Antibody-Dependent Cell Cytotoxicity (ADCC) is a type of immune response in which the effector cells of the immune system, such as natural killer (NK) cells, cytotoxic T-cells or macrophages, recognize and destroy virus-infected or cancer cells that are coated with antibodies.

In this process, an antibody produced by B-cells binds specifically to an antigen on the surface of a target cell. The other end of the antibody then interacts with Fc receptors found on the surface of effector cells. This interaction triggers the effector cells to release cytotoxic substances, such as perforins and granzymes, which create pores in the target cell membrane and induce apoptosis (programmed cell death).

ADCC plays an important role in the immune defense against viral infections and cancer. It is also a mechanism of action for some monoclonal antibody therapies used in cancer treatment.

Antinuclear antibodies (ANA) are a type of autoantibody that target structures found in the nucleus of a cell. These antibodies are produced by the immune system and attack the body's own cells and tissues, leading to inflammation and damage. The presence of ANA is often used as a marker for certain autoimmune diseases, such as systemic lupus erythematosus (SLE), Sjogren's syndrome, rheumatoid arthritis, scleroderma, and polymyositis.

ANA can be detected through a blood test called the antinuclear antibody test. A positive result indicates the presence of ANA in the blood, but it does not necessarily mean that a person has an autoimmune disease. Further testing is usually needed to confirm a diagnosis and determine the specific type of autoantibodies present.

It's important to note that ANA can also be found in healthy individuals, particularly as they age. Therefore, the test results should be interpreted in conjunction with other clinical findings and symptoms.

Cross reactions, in the context of medical diagnostics and immunology, refer to a situation where an antibody or a immune response directed against one antigen also reacts with a different antigen due to similarities in their molecular structure. This can occur in allergy testing, where a person who is allergic to a particular substance may have a positive test result for a different but related substance because of cross-reactivity between them. For example, some individuals who are allergic to birch pollen may also have symptoms when eating certain fruits, such as apples, due to cross-reactive proteins present in both.

Radioimmunotherapy (RIT) is a medical treatment that combines the specificity of antibodies and the therapeutic effects of radiation to target and destroy cancer cells. It involves the use of radioactive isotopes, which are attached to monoclonal antibodies, that recognize and bind to antigens expressed on the surface of cancer cells. Once bound, the radioactivity emitted from the isotope irradiates the cancer cells, causing damage to their DNA and leading to cell death. This targeted approach helps minimize radiation exposure to healthy tissues and reduces side effects compared to conventional radiotherapy techniques. RIT has been used in the treatment of various hematological malignancies, such as non-Hodgkin lymphoma, and is being investigated for solid tumors as well.

Immunoglobulin G (IgG) is a type of antibody, which is a protective protein produced by the immune system in response to foreign substances like bacteria or viruses. IgG is the most abundant type of antibody in human blood, making up about 75-80% of all antibodies. It is found in all body fluids and plays a crucial role in fighting infections caused by bacteria, viruses, and toxins.

IgG has several important functions:

1. Neutralization: IgG can bind to the surface of bacteria or viruses, preventing them from attaching to and infecting human cells.
2. Opsonization: IgG coats the surface of pathogens, making them more recognizable and easier for immune cells like neutrophils and macrophages to phagocytose (engulf and destroy) them.
3. Complement activation: IgG can activate the complement system, a group of proteins that work together to help eliminate pathogens from the body. Activation of the complement system leads to the formation of the membrane attack complex, which creates holes in the cell membranes of bacteria, leading to their lysis (destruction).
4. Antibody-dependent cellular cytotoxicity (ADCC): IgG can bind to immune cells like natural killer (NK) cells and trigger them to release substances that cause target cells (such as virus-infected or cancerous cells) to undergo apoptosis (programmed cell death).
5. Immune complex formation: IgG can form immune complexes with antigens, which can then be removed from the body through various mechanisms, such as phagocytosis by immune cells or excretion in urine.

IgG is a critical component of adaptive immunity and provides long-lasting protection against reinfection with many pathogens. It has four subclasses (IgG1, IgG2, IgG3, and IgG4) that differ in their structure, function, and distribution in the body.

A hybridoma is a type of hybrid cell that is created in a laboratory by fusing a cancer cell (usually a B cell) with a normal immune cell. The resulting hybrid cell combines the ability of the cancer cell to grow and divide indefinitely with the ability of the immune cell to produce antibodies, which are proteins that help the body fight infection.

Hybridomas are commonly used to produce monoclonal antibodies, which are identical copies of a single antibody produced by a single clone of cells. These antibodies can be used for a variety of purposes, including diagnostic tests and treatments for diseases such as cancer and autoimmune disorders.

To create hybridomas, B cells are first isolated from the spleen or blood of an animal that has been immunized with a specific antigen (a substance that triggers an immune response). The B cells are then fused with cancer cells using a chemical agent such as polyethylene glycol. The resulting hybrid cells are called hybridomas and are grown in culture medium, where they can be selected for their ability to produce antibodies specific to the antigen of interest. These antibody-producing hybridomas can then be cloned to produce large quantities of monoclonal antibodies.

Radioimmunodetection (RID) is a medical diagnostic technique that combines the specificity of antibodies with the sensitivity of radioisotopes to detect and locate antigens or tumor markers within the body. This technique involves labeling antibodies with radioactive isotopes, which are then introduced into the patient's body. The labeled antibodies bind to the target antigens, allowing for their detection and localization using external gamma cameras.

The process typically begins with the production of monoclonal or polyclonal antibodies that specifically recognize and bind to a particular antigen associated with a disease or condition. These antibodies are then labeled with radioisotopes such as technetium-99m, iodine-131, or indium-111, which emit gamma rays that can be detected by external imaging devices.

Once the labeled antibodies have been administered to the patient, they circulate throughout the body and bind to their respective antigens. The bound radioactive antibodies can then be imaged using a gamma camera or single-photon emission computed tomography (SPECT) scanner, providing information about the location, size, and distribution of the target antigens within the body.

Radioimmunodetection has been widely used in the detection and monitoring of various malignancies, including cancerous tumors and metastases, as well as inflammatory and infectious diseases. It offers several advantages over other diagnostic techniques, such as high sensitivity, specificity, and non-invasiveness, making it an essential tool in modern medical imaging and diagnostics.

Immunotherapy is a type of medical treatment that uses the body's own immune system to fight against diseases, such as cancer. It involves the use of substances (like vaccines, medications, or immune cells) that stimulate or suppress the immune system to help it recognize and destroy harmful disease-causing cells or agents, like tumor cells.

Immunotherapy can work in several ways:

1. Activating the immune system: Certain immunotherapies boost the body's natural immune responses, helping them recognize and attack cancer cells more effectively.
2. Suppressing immune system inhibitors: Some immunotherapies target and block proteins or molecules that can suppress the immune response, allowing the immune system to work more efficiently against diseases.
3. Replacing or enhancing specific immune cells: Immunotherapy can also involve administering immune cells (like T-cells) that have been genetically engineered or modified to recognize and destroy cancer cells.

Immunotherapies have shown promising results in treating various types of cancer, autoimmune diseases, and allergies. However, they can also cause side effects, as an overactive immune system may attack healthy tissues and organs. Therefore, careful monitoring is necessary during immunotherapy treatment.

Molecular sequence data refers to the specific arrangement of molecules, most commonly nucleotides in DNA or RNA, or amino acids in proteins, that make up a biological macromolecule. This data is generated through laboratory techniques such as sequencing, and provides information about the exact order of the constituent molecules. This data is crucial in various fields of biology, including genetics, evolution, and molecular biology, allowing for comparisons between different organisms, identification of genetic variations, and studies of gene function and regulation.

Immunotoxins are biomolecules that combine the specificity of an antibody with the toxicity of a toxin. They are created by chemically linking a monoclonal antibody (that recognizes and binds to a specific cell surface antigen) to a protein toxin (that inhibits protein synthesis in cells). The immunotoxin selectively binds to the target cell, gets internalized, and releases the toxin into the cytosol, leading to cell death. Immunotoxins have been explored as potential therapeutic agents for targeted cancer therapy and treatment of other diseases.

IgG receptors, also known as Fcγ receptors (Fc gamma receptors), are specialized protein molecules found on the surface of various immune cells, such as neutrophils, monocytes, macrophages, and some lymphocytes. These receptors recognize and bind to the Fc region of IgG antibodies, one of the five classes of immunoglobulins in the human body.

IgG receptors play a crucial role in immune responses by mediating different effector functions, including:

1. Antibody-dependent cellular cytotoxicity (ADCC): IgG receptors on natural killer (NK) cells and other immune cells bind to IgG antibodies coated on the surface of virus-infected or cancer cells, leading to their destruction.
2. Phagocytosis: When IgG antibodies tag pathogens or foreign particles, phagocytes like neutrophils and macrophages recognize and bind to these immune complexes via IgG receptors, facilitating the engulfment and removal of the targeted particles.
3. Antigen presentation: IgG receptors on antigen-presenting cells (APCs) can internalize immune complexes, process the antigens, and present them to T cells, thereby initiating adaptive immune responses.
4. Inflammatory response regulation: IgG receptors can modulate inflammation by activating or inhibiting downstream signaling pathways in immune cells, depending on the specific type of Fcγ receptor and its activation state.

There are several types of IgG receptors (FcγRI, FcγRII, FcγRIII, and FcγRIV) with varying affinities for different subclasses of IgG antibodies (IgG1, IgG2, IgG3, and IgG4). The distinct functions and expression patterns of these receptors contribute to the complexity and fine-tuning of immune responses in the human body.

BALB/c is an inbred strain of laboratory mouse that is widely used in biomedical research. The strain was developed at the Institute of Cancer Research in London by Henry Baldwin and his colleagues in the 1920s, and it has since become one of the most commonly used inbred strains in the world.

BALB/c mice are characterized by their black coat color, which is determined by a recessive allele at the tyrosinase locus. They are also known for their docile and friendly temperament, making them easy to handle and work with in the laboratory.

One of the key features of BALB/c mice that makes them useful for research is their susceptibility to certain types of tumors and immune responses. For example, they are highly susceptible to developing mammary tumors, which can be induced by chemical carcinogens or viral infection. They also have a strong Th2-biased immune response, which makes them useful models for studying allergic diseases and asthma.

BALB/c mice are also commonly used in studies of genetics, neuroscience, behavior, and infectious diseases. Because they are an inbred strain, they have a uniform genetic background, which makes it easier to control for genetic factors in experiments. Additionally, because they have been bred in the laboratory for many generations, they are highly standardized and reproducible, making them ideal subjects for scientific research.

An antigen-antibody reaction is a specific immune response that occurs when an antigen (a foreign substance, such as a protein or polysaccharide on the surface of a bacterium or virus) comes into contact with a corresponding antibody (a protective protein produced by the immune system in response to the antigen). The antigen and antibody bind together, forming an antigen-antibody complex. This interaction can neutralize the harmful effects of the antigen, mark it for destruction by other immune cells, or activate complement proteins to help eliminate the antigen from the body. Antigen-antibody reactions are a crucial part of the adaptive immune response and play a key role in the body's defense against infection and disease.

Pentetic Acid, also known as DTPA (Diethylenetriaminepentaacetic acid), is not a medication itself but a chelating agent used in the preparation of pharmaceutical products. A chelating agent is a compound that can form multiple bonds with metal ions, allowing them to be excreted from the body.

Pentetic Acid is used in medical treatments to remove or decrease the levels of certain toxic metals, such as lead, plutonium, americium, and curium, from the body. It can be given intravenously or orally, depending on the specific situation and the formulation of the medication.

It is important to note that the use of Pentetic Acid should be under the supervision of a healthcare professional, as it can also bind to essential metals like zinc, calcium, and iron, which can lead to deficiencies if not properly managed.

CD19 is a type of protein found on the surface of B cells, which are a type of white blood cell that plays a key role in the body's immune response. CD19 is a marker that helps identify and distinguish B cells from other types of cells in the body. It is also a target for immunotherapy in certain diseases, such as B-cell malignancies.

An antigen is any substance that can stimulate an immune response, particularly the production of antibodies. In the context of CD19, antigens refer to substances that can bind to CD19 and trigger a response from the immune system. This can include proteins, carbohydrates, or other molecules found on the surface of bacteria, viruses, or cancer cells.

Therefore, 'antigens, CD19' refers to any substances that can bind to the CD19 protein on B cells and trigger an immune response. These antigens may be used in the development of immunotherapies for the treatment of B-cell malignancies or other diseases.

An amino acid sequence is the specific order of amino acids in a protein or peptide molecule, formed by the linking of the amino group (-NH2) of one amino acid to the carboxyl group (-COOH) of another amino acid through a peptide bond. The sequence is determined by the genetic code and is unique to each type of protein or peptide. It plays a crucial role in determining the three-dimensional structure and function of proteins.

Protein engineering is a branch of molecular biology that involves the modification of proteins to achieve desired changes in their structure and function. This can be accomplished through various techniques, including site-directed mutagenesis, gene shuffling, directed evolution, and rational design. The goal of protein engineering may be to improve the stability, activity, specificity, or other properties of a protein for therapeutic, diagnostic, industrial, or research purposes. It is an interdisciplinary field that combines knowledge from genetics, biochemistry, structural biology, and computational modeling.

Siglec-2, also known as CD22, is a type of cell surface protein that belongs to the sialic acid-binding immunoglobulin-like lectins (Siglecs) family. It is primarily expressed on mature B cells and plays a crucial role in regulating B cell activation and function. Siglec-2 recognizes and binds to sialic acid residues on glycoproteins and gangliosides, which are sugars that are attached to proteins and lipids on the surface of cells. This binding can lead to inhibitory signals that dampen B cell activation and help prevent autoimmunity. Siglec-2 has also been implicated in the regulation of B cell migration and adhesion.

Immunoglobulin M (IgM) is a type of antibody that is primarily found in the blood and lymph fluid. It is the first antibody to be produced in response to an initial exposure to an antigen, making it an important part of the body's primary immune response. IgM antibodies are large molecules that are composed of five basic units, giving them a pentameric structure. They are primarily found on the surface of B cells as membrane-bound immunoglobulins (mlgM), where they function as receptors for antigens. Once an mlgM receptor binds to an antigen, it triggers the activation and differentiation of the B cell into a plasma cell that produces and secretes large amounts of soluble IgM antibodies.

IgM antibodies are particularly effective at agglutination (clumping) and complement activation, which makes them important in the early stages of an immune response to help clear pathogens from the bloodstream. However, they are not as stable or long-lived as other types of antibodies, such as IgG, and their levels tend to decline after the initial immune response has occurred.

In summary, Immunoglobulin M (IgM) is a type of antibody that plays a crucial role in the primary immune response to antigens by agglutination and complement activation. It is primarily found in the blood and lymph fluid, and it is produced by B cells after they are activated by an antigen.

Neoplasm antigens, also known as tumor antigens, are substances that are produced by cancer cells (neoplasms) and can stimulate an immune response. These antigens can be proteins, carbohydrates, or other molecules that are either unique to the cancer cells or are overexpressed or mutated versions of normal cellular proteins.

Neoplasm antigens can be classified into two main categories: tumor-specific antigens (TSAs) and tumor-associated antigens (TAAs). TSAs are unique to cancer cells and are not expressed by normal cells, while TAAs are present at low levels in normal cells but are overexpressed or altered in cancer cells.

TSAs can be further divided into viral antigens and mutated antigens. Viral antigens are produced when cancer is caused by a virus, such as human papillomavirus (HPV) in cervical cancer. Mutated antigens are the result of genetic mutations that occur during cancer development and are unique to each patient's tumor.

Neoplasm antigens play an important role in the immune response against cancer. They can be recognized by the immune system, leading to the activation of immune cells such as T cells and natural killer (NK) cells, which can then attack and destroy cancer cells. However, cancer cells often develop mechanisms to evade the immune response, allowing them to continue growing and spreading.

Understanding neoplasm antigens is important for the development of cancer immunotherapies, which aim to enhance the body's natural immune response against cancer. These therapies include checkpoint inhibitors, which block proteins that inhibit T cell activation, and therapeutic vaccines, which stimulate an immune response against specific tumor antigens.

Autoantibodies are defined as antibodies that are produced by the immune system and target the body's own cells, tissues, or organs. These antibodies mistakenly identify certain proteins or molecules in the body as foreign invaders and attack them, leading to an autoimmune response. Autoantibodies can be found in various autoimmune diseases such as rheumatoid arthritis, lupus, and thyroiditis. The presence of autoantibodies can also be used as a diagnostic marker for certain conditions.

Fungal antibodies are a type of protein called immunoglobulins that are produced by the immune system in response to the presence of fungi in the body. These antibodies are specifically designed to recognize and bind to antigens on the surface of fungal cells, marking them for destruction by other immune cells.

There are several types of fungal antibodies, including IgA, IgG, IgM, and IgE, each with a specific role in the immune response. For example, IgG antibodies are the most common type of antibody found in the blood and provide long-term immunity to fungi, while IgE antibodies are associated with allergic reactions to fungi.

Fungal antibodies can be measured in the blood or other bodily fluids to help diagnose fungal infections, monitor the effectiveness of treatment, or assess immune function in individuals who are at risk for fungal infections, such as those with weakened immune systems due to HIV/AIDS, cancer, or organ transplantation.

Carcinoembryonic antigen (CEA) is a protein that is normally produced in small amounts during fetal development. In adults, low levels of CEA can be found in the blood, but elevated levels are typically associated with various types of cancer, particularly colon, rectal, and breast cancer.

Measurement of CEA levels in the blood is sometimes used as a tumor marker to monitor response to treatment, detect recurrence, or screen for secondary cancers in patients with a history of certain types of cancer. However, it's important to note that CEA is not a specific or sensitive indicator of cancer and can be elevated in various benign conditions such as inflammation, smoking, and some gastrointestinal diseases. Therefore, the test should be interpreted in conjunction with other clinical and diagnostic findings.

Single-domain antibodies (sdAbs), also known as nanobodies or VHHs, are antigen-binding fragments derived from the heavy-chain only antibodies found in camelids (camels, llamas, and alpacas) and some shark species. These unique antibodies lack light chains and consist of a single variable domain (VHH) that can bind to specific antigens with high affinity and stability. The small size (12-15 kDa), robustness, and solubility make sdAbs attractive for various biotechnological and therapeutic applications, including diagnostics, targeted drug delivery, and protein engineering.

An antigen-antibody complex is a type of immune complex that forms when an antibody binds to a specific antigen. An antigen is any substance that triggers an immune response, while an antibody is a protein produced by the immune system to neutralize or destroy foreign substances like antigens.

When an antibody binds to an antigen, it forms a complex that can be either soluble or insoluble. Soluble complexes are formed when the antigen is small and can move freely through the bloodstream. Insoluble complexes, on the other hand, are formed when the antigen is too large to move freely, such as when it is part of a bacterium or virus.

The formation of antigen-antibody complexes plays an important role in the immune response. Once formed, these complexes can be recognized and cleared by other components of the immune system, such as phagocytes, which help to prevent further damage to the body. However, in some cases, the formation of large numbers of antigen-antibody complexes can lead to inflammation and tissue damage, contributing to the development of certain autoimmune diseases.

The Immunoglobulin (Ig) variable region is the antigen-binding part of an antibody, which is highly variable in its amino acid sequence and therefore specific to a particular epitope (the site on an antigen that is recognized by the antigen-binding site of an antibody). This variability is generated during the process of V(D)J recombination in the maturation of B cells, allowing for a diverse repertoire of antibodies to be produced and recognizing a wide range of potential pathogens.

The variable region is composed of several sub-regions including:

1. The heavy chain variable region (VH)
2. The light chain variable region (VL)
3. The heavy chain joining region (JH)
4. The light chain joining region (JL)

These regions are further divided into framework regions and complementarity-determining regions (CDRs). The CDRs, particularly CDR3, contain the most variability and are primarily responsible for antigen recognition.

Neutralization tests are a type of laboratory assay used in microbiology and immunology to measure the ability of a substance, such as an antibody or antitoxin, to neutralize the activity of a toxin or infectious agent. In these tests, the substance to be tested is mixed with a known quantity of the toxin or infectious agent, and the mixture is then incubated under controlled conditions. After incubation, the mixture is tested for residual toxicity or infectivity using a variety of methods, such as cell culture assays, animal models, or biochemical assays.

The neutralization titer is then calculated based on the highest dilution of the test substance that completely neutralizes the toxin or infectious agent. Neutralization tests are commonly used in the diagnosis and evaluation of immune responses to vaccines, as well as in the detection and quantification of toxins and other harmful substances.

Examples of neutralization tests include the serum neutralization test for measles antibodies, the plaque reduction neutralization test (PRNT) for dengue virus antibodies, and the cytotoxicity neutralization assay for botulinum neurotoxins.

T-lymphocytes, also known as T-cells, are a type of white blood cell that plays a key role in the adaptive immune system's response to infection. They are produced in the bone marrow and mature in the thymus gland. There are several different types of T-cells, including CD4+ helper T-cells, CD8+ cytotoxic T-cells, and regulatory T-cells (Tregs).

CD4+ helper T-cells assist in activating other immune cells, such as B-lymphocytes and macrophages. They also produce cytokines, which are signaling molecules that help coordinate the immune response. CD8+ cytotoxic T-cells directly kill infected cells by releasing toxic substances. Regulatory T-cells help maintain immune tolerance and prevent autoimmune diseases by suppressing the activity of other immune cells.

T-lymphocytes are important in the immune response to viral infections, cancer, and other diseases. Dysfunction or depletion of T-cells can lead to immunodeficiency and increased susceptibility to infections. On the other hand, an overactive T-cell response can contribute to autoimmune diseases and chronic inflammation.

Immunologic cytotoxicity refers to the damage or destruction of cells that occurs as a result of an immune response. This process involves the activation of immune cells, such as cytotoxic T cells and natural killer (NK) cells, which release toxic substances, such as perforins and granzymes, that can kill target cells.

In addition, antibodies produced by B cells can also contribute to immunologic cytotoxicity by binding to antigens on the surface of target cells and triggering complement-mediated lysis or antibody-dependent cellular cytotoxicity (ADCC) by activating immune effector cells.

Immunologic cytotoxicity plays an important role in the body's defense against viral infections, cancer cells, and other foreign substances. However, it can also contribute to tissue damage and autoimmune diseases if the immune system mistakenly targets healthy cells or tissues.

CD20 is not a medical definition of an antigen, but rather it is a cell surface marker that helps identify a specific type of white blood cell called B-lymphocytes or B-cells. These cells are part of the adaptive immune system and play a crucial role in producing antibodies to fight off infections.

CD20 is a protein found on the surface of mature B-cells, and it is used as a target for monoclonal antibody therapies in the treatment of certain types of cancer and autoimmune diseases. Rituximab is an example of a monoclonal antibody that targets CD20 and is used to treat conditions such as non-Hodgkin lymphoma, chronic lymphocytic leukemia, and rheumatoid arthritis.

While CD20 itself is not an antigen, it can be recognized by the immune system as a foreign substance when a monoclonal antibody such as rituximab binds to it. This binding can trigger an immune response, leading to the destruction of the B-cells that express CD20 on their surface.

Indium radioisotopes refer to specific types of radioactive indium atoms, which are unstable and emit radiation as they decay. Indium is a chemical element with the symbol In and atomic number 49. Its radioisotopes are often used in medical imaging and therapy due to their unique properties.

For instance, one commonly used indium radioisotope is Indium-111 (^111In), which has a half-life of approximately 2.8 days. It emits gamma rays, making it useful for diagnostic imaging techniques such as single-photon emission computed tomography (SPECT). In clinical applications, indium-111 is often attached to specific molecules or antibodies that target particular cells or tissues in the body, allowing medical professionals to monitor biological processes and identify diseases like cancer.

Another example is Indium-113m (^113mIn), which has a half-life of about 99 minutes. It emits low-energy gamma rays and is used as a source for in vivo counting, typically in the form of indium chloride (InCl3) solution. This radioisotope can be used to measure blood flow, ventilation, and other physiological parameters.

It's important to note that handling and using radioisotopes require proper training and safety measures due to their ionizing radiation properties.

Monoclonal antibodies are laboratory-produced proteins that mimic the immune system's ability to fight off harmful antigens such as viruses and cancer cells. They are created by fusing a single B cell (the type of white blood cell responsible for producing antibodies) with a tumor cell, resulting in a hybrid cell called a hybridoma. This hybridoma can then be cloned to produce a large number of identical cells, all producing the same antibody, hence "monoclonal."

Humanized monoclonal antibodies are a type of monoclonal antibody that have been genetically engineered to include human components. This is done to reduce the risk of an adverse immune response in patients receiving the treatment. In this process, the variable region of the mouse monoclonal antibody, which contains the antigen-binding site, is grafted onto a human constant region. The resulting humanized monoclonal antibody retains the ability to bind to the target antigen while minimizing the immunogenicity associated with murine (mouse) antibodies.

In summary, "antibodies, monoclonal, humanized" refers to a type of laboratory-produced protein that mimics the immune system's ability to fight off harmful antigens, but with reduced immunogenicity due to the inclusion of human components in their structure.

An Enzyme-Linked Immunosorbent Assay (ELISA) is a type of analytical biochemistry assay used to detect and quantify the presence of a substance, typically a protein or peptide, in a liquid sample. It takes its name from the enzyme-linked antibodies used in the assay.

In an ELISA, the sample is added to a well containing a surface that has been treated to capture the target substance. If the target substance is present in the sample, it will bind to the surface. Next, an enzyme-linked antibody specific to the target substance is added. This antibody will bind to the captured target substance if it is present. After washing away any unbound material, a substrate for the enzyme is added. If the enzyme is present due to its linkage to the antibody, it will catalyze a reaction that produces a detectable signal, such as a color change or fluorescence. The intensity of this signal is proportional to the amount of target substance present in the sample, allowing for quantification.

ELISAs are widely used in research and clinical settings to detect and measure various substances, including hormones, viruses, and bacteria. They offer high sensitivity, specificity, and reproducibility, making them a reliable choice for many applications.

Recombinant proteins are artificially created proteins produced through the use of recombinant DNA technology. This process involves combining DNA molecules from different sources to create a new set of genes that encode for a specific protein. The resulting recombinant protein can then be expressed, purified, and used for various applications in research, medicine, and industry.

Recombinant proteins are widely used in biomedical research to study protein function, structure, and interactions. They are also used in the development of diagnostic tests, vaccines, and therapeutic drugs. For example, recombinant insulin is a common treatment for diabetes, while recombinant human growth hormone is used to treat growth disorders.

The production of recombinant proteins typically involves the use of host cells, such as bacteria, yeast, or mammalian cells, which are engineered to express the desired protein. The host cells are transformed with a plasmid vector containing the gene of interest, along with regulatory elements that control its expression. Once the host cells are cultured and the protein is expressed, it can be purified using various chromatography techniques.

Overall, recombinant proteins have revolutionized many areas of biology and medicine, enabling researchers to study and manipulate proteins in ways that were previously impossible.

'Tumor cells, cultured' refers to the process of removing cancerous cells from a tumor and growing them in controlled laboratory conditions. This is typically done by isolating the tumor cells from a patient's tissue sample, then placing them in a nutrient-rich environment that promotes their growth and multiplication.

The resulting cultured tumor cells can be used for various research purposes, including the study of cancer biology, drug development, and toxicity testing. They provide a valuable tool for researchers to better understand the behavior and characteristics of cancer cells outside of the human body, which can lead to the development of more effective cancer treatments.

It is important to note that cultured tumor cells may not always behave exactly the same way as they do in the human body, so findings from cell culture studies must be validated through further research, such as animal models or clinical trials.

Blocking antibodies are a type of antibody that binds to a specific antigen but does not cause the immune system to directly attack the antigen. Instead, blocking antibodies prevent the antigen from interacting with other molecules or receptors, effectively "blocking" its activity. This can be useful in therapeutic settings, where blocking antibodies can be used to inhibit the activity of harmful proteins or toxins.

For example, some blocking antibodies have been developed to target and block the activity of specific cytokines, which are signaling molecules involved in inflammation and immune responses. By blocking the interaction between the cytokine and its receptor, these antibodies can help to reduce inflammation and alleviate symptoms in certain autoimmune diseases or chronic inflammatory conditions.

It's important to note that while blocking antibodies can be useful for therapeutic purposes, they can also have unintended consequences if they block the activity of essential proteins or molecules. Therefore, careful consideration and testing are required before using blocking antibodies as a treatment.

Recombinant fusion proteins are artificially created biomolecules that combine the functional domains or properties of two or more different proteins into a single protein entity. They are generated through recombinant DNA technology, where the genes encoding the desired protein domains are linked together and expressed as a single, chimeric gene in a host organism, such as bacteria, yeast, or mammalian cells.

The resulting fusion protein retains the functional properties of its individual constituent proteins, allowing for novel applications in research, diagnostics, and therapeutics. For instance, recombinant fusion proteins can be designed to enhance protein stability, solubility, or immunogenicity, making them valuable tools for studying protein-protein interactions, developing targeted therapies, or generating vaccines against infectious diseases or cancer.

Examples of recombinant fusion proteins include:

1. Etaglunatide (ABT-523): A soluble Fc fusion protein that combines the heavy chain fragment crystallizable region (Fc) of an immunoglobulin with the extracellular domain of the human interleukin-6 receptor (IL-6R). This fusion protein functions as a decoy receptor, neutralizing IL-6 and its downstream signaling pathways in rheumatoid arthritis.
2. Etanercept (Enbrel): A soluble TNF receptor p75 Fc fusion protein that binds to tumor necrosis factor-alpha (TNF-α) and inhibits its proinflammatory activity, making it a valuable therapeutic option for treating autoimmune diseases like rheumatoid arthritis, ankylosing spondylitis, and psoriasis.
3. Abatacept (Orencia): A fusion protein consisting of the extracellular domain of cytotoxic T-lymphocyte antigen 4 (CTLA-4) linked to the Fc region of an immunoglobulin, which downregulates T-cell activation and proliferation in autoimmune diseases like rheumatoid arthritis.
4. Belimumab (Benlysta): A monoclonal antibody that targets B-lymphocyte stimulator (BLyS) protein, preventing its interaction with the B-cell surface receptor and inhibiting B-cell activation in systemic lupus erythematosus (SLE).
5. Romiplostim (Nplate): A fusion protein consisting of a thrombopoietin receptor agonist peptide linked to an immunoglobulin Fc region, which stimulates platelet production in patients with chronic immune thrombocytopenia (ITP).
6. Darbepoetin alfa (Aranesp): A hyperglycosylated erythropoiesis-stimulating protein that functions as a longer-acting form of recombinant human erythropoietin, used to treat anemia in patients with chronic kidney disease or cancer.
7. Palivizumab (Synagis): A monoclonal antibody directed against the F protein of respiratory syncytial virus (RSV), which prevents RSV infection and is administered prophylactically to high-risk infants during the RSV season.
8. Ranibizumab (Lucentis): A recombinant humanized monoclonal antibody fragment that binds and inhibits vascular endothelial growth factor A (VEGF-A), used in the treatment of age-related macular degeneration, diabetic retinopathy, and other ocular disorders.
9. Cetuximab (Erbitux): A chimeric monoclonal antibody that binds to epidermal growth factor receptor (EGFR), used in the treatment of colorectal cancer and head and neck squamous cell carcinoma.
10. Adalimumab (Humira): A fully humanized monoclonal antibody that targets tumor necrosis factor-alpha (TNF-α), used in the treatment of various inflammatory diseases, including rheumatoid arthritis, psoriasis, and Crohn's disease.
11. Bevacizumab (Avastin): A recombinant humanized monoclonal antibody that binds to VEGF-A, used in the treatment of various cancers, including colorectal, lung, breast, and kidney cancer.
12. Trastuzumab (Herceptin): A humanized monoclonal antibody that targets HER2/neu receptor, used in the treatment of breast cancer.
13. Rituximab (Rituxan): A chimeric monoclonal antibody that binds to CD20 antigen on B cells, used in the treatment of non-Hodgkin's lymphoma and rheumatoid arthritis.
14. Palivizumab (Synagis): A humanized monoclonal antibody that binds to the F protein of respiratory syncytial virus, used in the prevention of respiratory syncytial virus infection in high-risk infants.
15. Infliximab (Remicade): A chimeric monoclonal antibody that targets TNF-α, used in the treatment of various inflammatory diseases, including Crohn's disease, ulcerative colitis, rheumatoid arthritis, and ankylosing spondylitis.
16. Natalizumab (Tysabri): A humanized monoclonal antibody that binds to α4β1 integrin, used in the treatment of multiple sclerosis and Crohn's disease.
17. Adalimumab (Humira): A fully human monoclonal antibody that targets TNF-α, used in the treatment of various inflammatory diseases, including rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, and ulcerative colitis.
18. Golimumab (Simponi): A fully human monoclonal antibody that targets TNF-α, used in the treatment of rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, and ulcerative colitis.
19. Certolizumab pegol (Cimzia): A PEGylated Fab' fragment of a humanized monoclonal antibody that targets TNF-α, used in the treatment of rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, and Crohn's disease.
20. Ustekinumab (Stelara): A fully human monoclonal antibody that targets IL-12 and IL-23, used in the treatment of psoriasis, psoriatic arthritis, and Crohn's disease.
21. Secukinumab (Cosentyx): A fully human monoclonal antibody that targets IL-17A, used in the treatment of psoriasis, psoriatic arthritis, and ankylosing spondylitis.
22. Ixekizumab (Taltz): A fully human monoclonal antibody that targets IL-17A, used in the treatment of psoriasis and psoriatic arthritis.
23. Brodalumab (Siliq): A fully human monoclonal antibody that targets IL-17 receptor A, used in the treatment of psoriasis.
24. Sarilumab (Kevzara): A fully human monoclonal antibody that targets the IL-6 receptor, used in the treatment of rheumatoid arthritis.
25. Tocilizumab (Actemra): A humanized monoclonal antibody that targets the IL-6 receptor, used in the treatment of rheumatoid arthritis, systemic juvenile idiopathic arthritis, polyarticular juvenile idiopathic arthritis, giant cell arteritis, and chimeric antigen receptor T-cell-induced cytokine release syndrome.
26. Siltuximab (Sylvant): A chimeric monoclonal antibody that targets IL-6, used in the treatment of multicentric Castleman disease.
27. Satralizumab (Enspryng): A humanized monoclonal antibody that targets IL-6 receptor alpha, used in the treatment of neuromyelitis optica spectrum disorder.
28. Sirukumab (Plivensia): A human monoclonal antibody that targets IL-6, used in the treatment

Passive immunization is a type of temporary immunity that is transferred to an individual through the injection of antibodies produced outside of the body, rather than through the active production of antibodies in the body in response to vaccination or infection. This can be done through the administration of preformed antibodies, such as immune globulins, which contain a mixture of antibodies that provide immediate protection against specific diseases.

Passive immunization is often used in situations where individuals have been exposed to a disease and do not have time to develop their own active immune response, or in cases where individuals are unable to produce an adequate immune response due to certain medical conditions. It can also be used as a short-term measure to provide protection until an individual can receive a vaccination that will confer long-term immunity.

Passive immunization provides immediate protection against disease, but the protection is typically short-lived, lasting only a few weeks or months. This is because the transferred antibodies are gradually broken down and eliminated by the body over time. In contrast, active immunization confers long-term immunity through the production of memory cells that can mount a rapid and effective immune response upon re-exposure to the same pathogen in the future.

A cell line is a culture of cells that are grown in a laboratory for use in research. These cells are usually taken from a single cell or group of cells, and they are able to divide and grow continuously in the lab. Cell lines can come from many different sources, including animals, plants, and humans. They are often used in scientific research to study cellular processes, disease mechanisms, and to test new drugs or treatments. Some common types of human cell lines include HeLa cells (which come from a cancer patient named Henrietta Lacks), HEK293 cells (which come from embryonic kidney cells), and HUVEC cells (which come from umbilical vein endothelial cells). It is important to note that cell lines are not the same as primary cells, which are cells that are taken directly from a living organism and have not been grown in the lab.

"Nude mice" is a term used in the field of laboratory research to describe a strain of mice that have been genetically engineered to lack a functional immune system. Specifically, nude mice lack a thymus gland and have a mutation in the FOXN1 gene, which results in a failure to develop a mature T-cell population. This means that they are unable to mount an effective immune response against foreign substances or organisms.

The name "nude" refers to the fact that these mice also have a lack of functional hair follicles, resulting in a hairless or partially hairless phenotype. This feature is actually a secondary consequence of the same genetic mutation that causes their immune deficiency.

Nude mice are commonly used in research because their weakened immune system makes them an ideal host for transplanted tumors, tissues, and cells from other species, including humans. This allows researchers to study the behavior of these foreign substances in a living organism without the complication of an immune response. However, it's important to note that because nude mice lack a functional immune system, they must be kept in sterile conditions and are more susceptible to infection than normal mice.

Heterophile antibodies are a type of antibody that can react with antigens from more than one source, rather than being specific to a single antigen. They are produced in response to an initial infection or immunization, but can also cross-react with antigens from unrelated organisms or substances. A common example of heterophile antibodies are those that are produced in response to Epstein-Barr virus (EBV) infection, which can cause infectious mononucleosis. These antibodies, known as Paul-Bunnell antibodies, can agglutinate (clump together) sheep or horse red blood cells, which is the basis for a diagnostic test for EBV infection called the Monospot test. However, it's important to note that not all cases of infectious mononucleosis are caused by EBV, and other infections or conditions can also cause the production of heterophile antibodies, leading to false-positive results.

Catalytic antibodies, also known as abzymes or catalytic immune proteins, are a type of antibody that possesses enzymatic activity. They are capable of accelerating specific chemical reactions in a manner similar to traditional enzymes. This unique property arises from the ability of certain antibodies to bind substrates and promote their conversion into products through a series of chemical transformations.

Catalytic antibodies are generated by immunizing an organism with a transition state analogue, a molecule that mimics the high-energy, transient structure of a substrate during a chemical reaction. The immune system recognizes this analogue as foreign and produces antibodies against it. Some of these antibodies will bind to the transition state analogue in a way that stabilizes its geometry and lowers the energy barrier for the conversion of the substrate into the product. This results in the formation of a catalytic antibody, which can then accelerate this specific chemical reaction when presented with the appropriate substrate.

These specialized antibodies have attracted significant interest in the fields of chemistry, biochemistry, and immunology due to their potential applications in various areas, including drug design, diagnostics, and environmental monitoring. However, it is important to note that catalytic antibodies are still a subject of ongoing research, and their use as practical tools in these applications is not yet widespread.

I believe there may be some confusion in your question. "Rabbits" is a common name used to refer to the Lagomorpha species, particularly members of the family Leporidae. They are small mammals known for their long ears, strong legs, and quick reproduction.

However, if you're referring to "rabbits" in a medical context, there is a term called "rabbit syndrome," which is a rare movement disorder characterized by repetitive, involuntary movements of the fingers, resembling those of a rabbit chewing. It is also known as "finger-chewing chorea." This condition is usually associated with certain medications, particularly antipsychotics, and typically resolves when the medication is stopped or adjusted.

A cell line that is derived from tumor cells and has been adapted to grow in culture. These cell lines are often used in research to study the characteristics of cancer cells, including their growth patterns, genetic changes, and responses to various treatments. They can be established from many different types of tumors, such as carcinomas, sarcomas, and leukemias. Once established, these cell lines can be grown and maintained indefinitely in the laboratory, allowing researchers to conduct experiments and studies that would not be feasible using primary tumor cells. It is important to note that tumor cell lines may not always accurately represent the behavior of the original tumor, as they can undergo genetic changes during their time in culture.

Lymphocyte activation is the process by which B-cells and T-cells (types of lymphocytes) become activated to perform effector functions in an immune response. This process involves the recognition of specific antigens presented on the surface of antigen-presenting cells, such as dendritic cells or macrophages.

The activation of B-cells leads to their differentiation into plasma cells that produce antibodies, while the activation of T-cells results in the production of cytotoxic T-cells (CD8+ T-cells) that can directly kill infected cells or helper T-cells (CD4+ T-cells) that assist other immune cells.

Lymphocyte activation involves a series of intracellular signaling events, including the binding of co-stimulatory molecules and the release of cytokines, which ultimately result in the expression of genes involved in cell proliferation, differentiation, and effector functions. The activation process is tightly regulated to prevent excessive or inappropriate immune responses that can lead to autoimmunity or chronic inflammation.

Immunoglobulin Fc fragments are the crystallizable fragment of an antibody that is responsible for effector functions such as engagement with Fc receptors on immune cells, activation of the complement system, and neutralization of toxins. The Fc region is located at the tail end of the Y-shaped immunoglobulin molecule, and it is made up of constant regions of the heavy chains of the antibody.

When an antibody binds to its target antigen, the Fc region can interact with other proteins in the immune system, leading to a variety of responses such as phagocytosis, antibody-dependent cellular cytotoxicity (ADCC), and complement activation. These effector functions help to eliminate pathogens and infected cells from the body.

Immunoglobulin Fc fragments can be produced artificially through enzymatic digestion of intact antibodies, resulting in a fragment that retains the ability to interact with Fc receptors and other proteins involved in immune responses. These fragments have potential therapeutic applications in a variety of diseases, including autoimmune disorders, inflammatory conditions, and cancer.

Leukemia, B-cell is a type of cancer that affects the blood and bone marrow, characterized by an overproduction of abnormal B-lymphocytes, a type of white blood cell. These abnormal cells accumulate in the bone marrow and interfere with the production of normal blood cells, leading to anemia, infection, and bleeding.

B-cells are a type of lymphocyte that plays a crucial role in the immune system by producing antibodies to help fight off infections. In B-cell leukemia, the cancerous B-cells do not mature properly and accumulate in the bone marrow, leading to a decrease in the number of healthy white blood cells, red blood cells, and platelets.

There are several types of B-cell leukemia, including acute lymphoblastic leukemia (ALL) and chronic lymphocytic leukemia (CLL). ALL is more common in children and young adults, while CLL is more common in older adults. Treatment options for B-cell leukemia depend on the type and stage of the disease and may include chemotherapy, radiation therapy, stem cell transplantation, or targeted therapies.

Tissue distribution, in the context of pharmacology and toxicology, refers to the way that a drug or xenobiotic (a chemical substance found within an organism that is not naturally produced by or expected to be present within that organism) is distributed throughout the body's tissues after administration. It describes how much of the drug or xenobiotic can be found in various tissues and organs, and is influenced by factors such as blood flow, lipid solubility, protein binding, and the permeability of cell membranes. Understanding tissue distribution is important for predicting the potential effects of a drug or toxin on different parts of the body, and for designing drugs with improved safety and efficacy profiles.

Neoplasm transplantation is not a recognized or established medical procedure in the field of oncology. The term "neoplasm" refers to an abnormal growth of cells, which can be benign or malignant (cancerous). "Transplantation" typically refers to the surgical transfer of living cells, tissues, or organs from one part of the body to another or between individuals.

The concept of neoplasm transplantation may imply the transfer of cancerous cells or tissues from a donor to a recipient, which is not a standard practice due to ethical considerations and the potential harm it could cause to the recipient. In some rare instances, researchers might use laboratory animals to study the transmission and growth of human cancer cells, but this is done for scientific research purposes only and under strict regulatory guidelines.

In summary, there is no medical definition for 'Neoplasm Transplantation' as it does not represent a standard or ethical medical practice.

Iodine radioisotopes are radioactive isotopes of the element iodine, which decays and emits radiation in the form of gamma rays. Some commonly used iodine radioisotopes include I-123, I-125, I-131. These radioisotopes have various medical applications such as in diagnostic imaging, therapy for thyroid disorders, and cancer treatment.

For example, I-131 is commonly used to treat hyperthyroidism and differentiated thyroid cancer due to its ability to destroy thyroid tissue. On the other hand, I-123 is often used in nuclear medicine scans of the thyroid gland because it emits gamma rays that can be detected by a gamma camera, allowing for detailed images of the gland's structure and function.

It is important to note that handling and administering radioisotopes require specialized training and safety precautions due to their radiation-emitting properties.

SCID mice is an acronym for Severe Combined Immunodeficiency mice. These are genetically modified mice that lack a functional immune system due to the mutation or knockout of several key genes required for immunity. This makes them ideal for studying the human immune system, infectious diseases, and cancer, as well as testing new therapies and treatments in a controlled environment without the risk of interference from the mouse's own immune system. SCID mice are often used in xenotransplantation studies, where human cells or tissues are transplanted into the mouse to study their behavior and interactions with the human immune system.

Immunization is defined medically as the process where an individual is made immune or resistant to an infectious disease, typically through the administration of a vaccine. The vaccine stimulates the body's own immune system to recognize and fight off the specific disease-causing organism, thereby preventing or reducing the severity of future infections with that organism.

Immunization can be achieved actively, where the person is given a vaccine to trigger an immune response, or passively, where antibodies are transferred to the person through immunoglobulin therapy. Immunizations are an important part of preventive healthcare and have been successful in controlling and eliminating many infectious diseases worldwide.

B-cell lymphoma is a type of cancer that originates from the B-lymphocytes, which are a part of the immune system and play a crucial role in fighting infections. These cells can develop mutations in their DNA, leading to uncontrolled growth and division, resulting in the formation of a tumor.

B-cell lymphomas can be classified into two main categories: Hodgkin's lymphoma and non-Hodgkin's lymphoma. B-cell lymphomas are further divided into subtypes based on their specific characteristics, such as the appearance of the cells under a microscope, the genetic changes present in the cancer cells, and the aggressiveness of the disease.

Some common types of B-cell lymphomas include diffuse large B-cell lymphoma, follicular lymphoma, mantle cell lymphoma, and Burkitt lymphoma. Treatment options for B-cell lymphomas depend on the specific subtype, stage of the disease, and other individual factors. Treatment may include chemotherapy, radiation therapy, immunotherapy, targeted therapy, or stem cell transplantation.

An antigen is a substance (usually a protein) that is recognized as foreign by the immune system and stimulates an immune response, leading to the production of antibodies or activation of T-cells. Antigens can be derived from various sources, including bacteria, viruses, fungi, parasites, and tumor cells. They can also come from non-living substances such as pollen, dust mites, or chemicals.

Antigens contain epitopes, which are specific regions on the antigen molecule that are recognized by the immune system. The immune system's response to an antigen depends on several factors, including the type of antigen, its size, and its location in the body.

In general, antigens can be classified into two main categories:

1. T-dependent antigens: These require the help of T-cells to stimulate an immune response. They are typically larger, more complex molecules that contain multiple epitopes capable of binding to both MHC class II molecules on antigen-presenting cells and T-cell receptors on CD4+ T-cells.
2. T-independent antigens: These do not require the help of T-cells to stimulate an immune response. They are usually smaller, simpler molecules that contain repetitive epitopes capable of cross-linking B-cell receptors and activating them directly.

Understanding antigens and their properties is crucial for developing vaccines, diagnostic tests, and immunotherapies.

CD (cluster of differentiation) antigens are cell-surface proteins that are expressed on leukocytes (white blood cells) and can be used to identify and distinguish different subsets of these cells. They are important markers in the field of immunology and hematology, and are commonly used to diagnose and monitor various diseases, including cancer, autoimmune disorders, and infectious diseases.

CD antigens are designated by numbers, such as CD4, CD8, CD19, etc., which refer to specific proteins found on the surface of different types of leukocytes. For example, CD4 is a protein found on the surface of helper T cells, while CD8 is found on cytotoxic T cells.

CD antigens can be used as targets for immunotherapy, such as monoclonal antibody therapy, in which antibodies are designed to bind to specific CD antigens and trigger an immune response against cancer cells or infected cells. They can also be used as markers to monitor the effectiveness of treatments and to detect minimal residual disease (MRD) after treatment.

It's important to note that not all CD antigens are exclusive to leukocytes, some can be found on other cell types as well, and their expression can vary depending on the activation state or differentiation stage of the cells.

Flow cytometry is a medical and research technique used to measure physical and chemical characteristics of cells or particles, one cell at a time, as they flow in a fluid stream through a beam of light. The properties measured include:

* Cell size (light scatter)
* Cell internal complexity (granularity, also light scatter)
* Presence or absence of specific proteins or other molecules on the cell surface or inside the cell (using fluorescent antibodies or other fluorescent probes)

The technique is widely used in cell counting, cell sorting, protein engineering, biomarker discovery and monitoring disease progression, particularly in hematology, immunology, and cancer research.

Heterocyclic compounds are organic molecules that contain a ring structure made up of at least one atom that is not carbon, known as a heteroatom. These heteroatoms can include nitrogen, oxygen, sulfur, or other elements. In the case of "1-ring" heterocyclic compounds, the molecule contains a single ring structure composed of these heteroatoms and carbon atoms. Examples of 1-ring heterocyclic compounds include pyridine (contains one nitrogen atom in the ring), furan (contains one oxygen atom in the ring), and thiophene (contains one sulfur atom in the ring). These compounds play important roles in various biological processes and are also found in many drugs, dyes, and materials.

Immunoglobulin A (IgA) is a type of antibody that plays a crucial role in the immune function of the human body. It is primarily found in external secretions, such as saliva, tears, breast milk, and sweat, as well as in mucous membranes lining the respiratory and gastrointestinal tracts. IgA exists in two forms: a monomeric form found in serum and a polymeric form found in secretions.

The primary function of IgA is to provide immune protection at mucosal surfaces, which are exposed to various environmental antigens, such as bacteria, viruses, parasites, and allergens. By doing so, it helps prevent the entry and colonization of pathogens into the body, reducing the risk of infections and inflammation.

IgA functions by binding to antigens present on the surface of pathogens or allergens, forming immune complexes that can neutralize their activity. These complexes are then transported across the epithelial cells lining mucosal surfaces and released into the lumen, where they prevent the adherence and invasion of pathogens.

In summary, Immunoglobulin A (IgA) is a vital antibody that provides immune defense at mucosal surfaces by neutralizing and preventing the entry of harmful antigens into the body.

The Fluorescent Antibody Technique (FAT), Indirect is a type of immunofluorescence assay used to detect the presence of specific antigens in a sample. In this method, the sample is first incubated with a primary antibody that binds to the target antigen. After washing to remove unbound primary antibodies, a secondary fluorescently labeled antibody is added, which recognizes and binds to the primary antibody. This indirect labeling approach allows for amplification of the signal, making it more sensitive than direct methods. The sample is then examined under a fluorescence microscope to visualize the location and amount of antigen based on the emitted light from the fluorescent secondary antibody. It's commonly used in diagnostic laboratories for detection of various bacteria, viruses, and other antigens in clinical specimens.

'Immune sera' refers to the serum fraction of blood that contains antibodies produced in response to an antigenic stimulus, such as a vaccine or an infection. These antibodies are proteins known as immunoglobulins, which are secreted by B cells (a type of white blood cell) and can recognize and bind to specific antigens. Immune sera can be collected from an immunized individual and used as a source of passive immunity to protect against infection or disease. It is often used in research and diagnostic settings to identify or measure the presence of specific antigens or antibodies.

Western blotting is a laboratory technique used in molecular biology to detect and quantify specific proteins in a mixture of many different proteins. This technique is commonly used to confirm the expression of a protein of interest, determine its size, and investigate its post-translational modifications. The name "Western" blotting distinguishes this technique from Southern blotting (for DNA) and Northern blotting (for RNA).

The Western blotting procedure involves several steps:

1. Protein extraction: The sample containing the proteins of interest is first extracted, often by breaking open cells or tissues and using a buffer to extract the proteins.
2. Separation of proteins by electrophoresis: The extracted proteins are then separated based on their size by loading them onto a polyacrylamide gel and running an electric current through the gel (a process called sodium dodecyl sulfate-polyacrylamide gel electrophoresis or SDS-PAGE). This separates the proteins according to their molecular weight, with smaller proteins migrating faster than larger ones.
3. Transfer of proteins to a membrane: After separation, the proteins are transferred from the gel onto a nitrocellulose or polyvinylidene fluoride (PVDF) membrane using an electric current in a process called blotting. This creates a replica of the protein pattern on the gel but now immobilized on the membrane for further analysis.
4. Blocking: The membrane is then blocked with a blocking agent, such as non-fat dry milk or bovine serum albumin (BSA), to prevent non-specific binding of antibodies in subsequent steps.
5. Primary antibody incubation: A primary antibody that specifically recognizes the protein of interest is added and allowed to bind to its target protein on the membrane. This step may be performed at room temperature or 4°C overnight, depending on the antibody's properties.
6. Washing: The membrane is washed with a buffer to remove unbound primary antibodies.
7. Secondary antibody incubation: A secondary antibody that recognizes the primary antibody (often coupled to an enzyme or fluorophore) is added and allowed to bind to the primary antibody. This step may involve using a horseradish peroxidase (HRP)-conjugated or alkaline phosphatase (AP)-conjugated secondary antibody, depending on the detection method used later.
8. Washing: The membrane is washed again to remove unbound secondary antibodies.
9. Detection: A detection reagent is added to visualize the protein of interest by detecting the signal generated from the enzyme-conjugated or fluorophore-conjugated secondary antibody. This can be done using chemiluminescent, colorimetric, or fluorescent methods.
10. Analysis: The resulting image is analyzed to determine the presence and quantity of the protein of interest in the sample.

Western blotting is a powerful technique for identifying and quantifying specific proteins within complex mixtures. It can be used to study protein expression, post-translational modifications, protein-protein interactions, and more. However, it requires careful optimization and validation to ensure accurate and reproducible results.

A peptide library is a collection of a large number of peptides, which are short chains of amino acids. Each peptide in the library is typically composed of a defined length and sequence, and may contain a variety of different amino acids. Peptide libraries can be synthesized using automated techniques and are often used in scientific research to identify potential ligands (molecules that bind to specific targets) or to study the interactions between peptides and other molecules.

In a peptide library, each peptide is usually attached to a solid support, such as a resin bead, and the entire library can be created using split-and-pool synthesis techniques. This allows for the rapid and efficient synthesis of a large number of unique peptides, which can then be screened for specific activities or properties.

Peptide libraries are used in various fields such as drug discovery, proteomics, and molecular biology to identify potential therapeutic targets, understand protein-protein interactions, and develop new diagnostic tools.

Epitope mapping is a technique used in immunology to identify the specific portion or regions (called epitopes) on an antigen that are recognized and bind to antibodies or T-cell receptors. This process helps to understand the molecular basis of immune responses against various pathogens, allergens, or transplanted tissues.

Epitope mapping can be performed using different methods such as:

1. Peptide scanning: In this method, a series of overlapping peptides spanning the entire length of the antigen are synthesized and tested for their ability to bind to antibodies or T-cell receptors. The peptide that shows binding is considered to contain the epitope.
2. Site-directed mutagenesis: In this approach, specific amino acids within the antigen are altered, and the modified antigens are tested for their ability to bind to antibodies or T-cell receptors. This helps in identifying the critical residues within the epitope.
3. X-ray crystallography and NMR spectroscopy: These techniques provide detailed information about the three-dimensional structure of antigen-antibody complexes, allowing for accurate identification of epitopes at an atomic level.

The results from epitope mapping can be useful in various applications, including vaccine design, diagnostic test development, and understanding the basis of autoimmune diseases.

Antiphospholipid antibodies are a type of autoantibody that targets and binds to certain proteins found in the blood that attach to phospholipids (a type of fat molecule). These antibodies are associated with an increased risk of developing antiphospholipid syndrome, a disorder characterized by abnormal blood clotting.

There are several types of antiphospholipid antibodies, including:

1. Lupus anticoagulant: This type of antiphospholipid antibody can interfere with blood clotting tests and may increase the risk of thrombosis (blood clots) in both arteries and veins.
2. Anticardiolipin antibodies: These antibodies target a specific phospholipid called cardiolipin, which is found in the inner membrane of mitochondria. High levels of anticardiolipin antibodies are associated with an increased risk of thrombosis and pregnancy complications such as recurrent miscarriage.
3. Anti-β2 glycoprotein I antibodies: These antibodies target a protein called β2 glycoprotein I, which binds to negatively charged phospholipids on the surface of cells. High levels of anti-β2 glycoprotein I antibodies are associated with an increased risk of thrombosis and pregnancy complications.

The exact mechanism by which antiphospholipid antibodies cause blood clotting is not fully understood, but it is thought to involve the activation of platelets, the inhibition of natural anticoagulants, and the promotion of inflammation. Antiphospholipid syndrome can be treated with medications that thin the blood or prevent clots from forming, such as aspirin, warfarin, or heparin.

"Cells, cultured" is a medical term that refers to cells that have been removed from an organism and grown in controlled laboratory conditions outside of the body. This process is called cell culture and it allows scientists to study cells in a more controlled and accessible environment than they would have inside the body. Cultured cells can be derived from a variety of sources, including tissues, organs, or fluids from humans, animals, or cell lines that have been previously established in the laboratory.

Cell culture involves several steps, including isolation of the cells from the tissue, purification and characterization of the cells, and maintenance of the cells in appropriate growth conditions. The cells are typically grown in specialized media that contain nutrients, growth factors, and other components necessary for their survival and proliferation. Cultured cells can be used for a variety of purposes, including basic research, drug development and testing, and production of biological products such as vaccines and gene therapies.

It is important to note that cultured cells may behave differently than they do in the body, and results obtained from cell culture studies may not always translate directly to human physiology or disease. Therefore, it is essential to validate findings from cell culture experiments using additional models and ultimately in clinical trials involving human subjects.

A xenograft model antitumor assay is a type of preclinical cancer research study that involves transplanting human tumor cells or tissues into an immunodeficient mouse. This model allows researchers to study the effects of various treatments, such as drugs or immune therapies, on human tumors in a living organism.

In this assay, human tumor cells or tissues are implanted into the mouse, typically under the skin or in another organ, where they grow and form a tumor. Once the tumor has established, the mouse is treated with the experimental therapy, and the tumor's growth is monitored over time. The response of the tumor to the treatment is then assessed by measuring changes in tumor size or weight, as well as other parameters such as survival rate and metastasis.

Xenograft model antitumor assays are useful for evaluating the efficacy and safety of new cancer therapies before they are tested in human clinical trials. They provide valuable information on how the tumors respond to treatment, drug pharmacokinetics, and toxicity, which can help researchers optimize dosing regimens and identify potential side effects. However, it is important to note that xenograft models have limitations, such as differences in tumor biology between mice and humans, and may not always predict how well a therapy will work in human patients.

Electrophoresis, polyacrylamide gel (EPG) is a laboratory technique used to separate and analyze complex mixtures of proteins or nucleic acids (DNA or RNA) based on their size and electrical charge. This technique utilizes a matrix made of cross-linked polyacrylamide, a type of gel, which provides a stable and uniform environment for the separation of molecules.

In this process:

1. The polyacrylamide gel is prepared by mixing acrylamide monomers with a cross-linking agent (bis-acrylamide) and a catalyst (ammonium persulfate) in the presence of a buffer solution.
2. The gel is then poured into a mold and allowed to polymerize, forming a solid matrix with uniform pore sizes that depend on the concentration of acrylamide used. Higher concentrations result in smaller pores, providing better resolution for separating smaller molecules.
3. Once the gel has set, it is placed in an electrophoresis apparatus containing a buffer solution. Samples containing the mixture of proteins or nucleic acids are loaded into wells on the top of the gel.
4. An electric field is applied across the gel, causing the negatively charged molecules to migrate towards the positive electrode (anode) while positively charged molecules move toward the negative electrode (cathode). The rate of migration depends on the size, charge, and shape of the molecules.
5. Smaller molecules move faster through the gel matrix and will migrate farther from the origin compared to larger molecules, resulting in separation based on size. Proteins and nucleic acids can be selectively stained after electrophoresis to visualize the separated bands.

EPG is widely used in various research fields, including molecular biology, genetics, proteomics, and forensic science, for applications such as protein characterization, DNA fragment analysis, cloning, mutation detection, and quality control of nucleic acid or protein samples.

Immunohistochemistry (IHC) is a technique used in pathology and laboratory medicine to identify specific proteins or antigens in tissue sections. It combines the principles of immunology and histology to detect the presence and location of these target molecules within cells and tissues. This technique utilizes antibodies that are specific to the protein or antigen of interest, which are then tagged with a detection system such as a chromogen or fluorophore. The stained tissue sections can be examined under a microscope, allowing for the visualization and analysis of the distribution and expression patterns of the target molecule in the context of the tissue architecture. Immunohistochemistry is widely used in diagnostic pathology to help identify various diseases, including cancer, infectious diseases, and immune-mediated disorders.

Neoplasms are abnormal growths of cells or tissues in the body that serve no physiological function. They can be benign (non-cancerous) or malignant (cancerous). Benign neoplasms are typically slow growing and do not spread to other parts of the body, while malignant neoplasms are aggressive, invasive, and can metastasize to distant sites.

Neoplasms occur when there is a dysregulation in the normal process of cell division and differentiation, leading to uncontrolled growth and accumulation of cells. This can result from genetic mutations or other factors such as viral infections, environmental exposures, or hormonal imbalances.

Neoplasms can develop in any organ or tissue of the body and can cause various symptoms depending on their size, location, and type. Treatment options for neoplasms include surgery, radiation therapy, chemotherapy, immunotherapy, and targeted therapy, among others.

B-lymphocytes, also known as B-cells, are a type of white blood cell that plays a key role in the immune system's response to infection. They are responsible for producing antibodies, which are proteins that help to neutralize or destroy pathogens such as bacteria and viruses.

When a B-lymphocyte encounters a pathogen, it becomes activated and begins to divide and differentiate into plasma cells, which produce and secrete large amounts of antibodies specific to the antigens on the surface of the pathogen. These antibodies bind to the pathogen, marking it for destruction by other immune cells such as neutrophils and macrophages.

B-lymphocytes also have a role in presenting antigens to T-lymphocytes, another type of white blood cell involved in the immune response. This helps to stimulate the activation and proliferation of T-lymphocytes, which can then go on to destroy infected cells or help to coordinate the overall immune response.

Overall, B-lymphocytes are an essential part of the adaptive immune system, providing long-lasting immunity to previously encountered pathogens and helping to protect against future infections.

Fc receptors (FcRs) are specialized proteins found on the surface of various immune cells, including neutrophils, monocytes, macrophages, eosinophils, basophils, mast cells, and B lymphocytes. They play a crucial role in the immune response by recognizing and binding to the Fc region of antibodies (IgG, IgA, and IgE) after they have interacted with their specific antigens.

FcRs can be classified into several types based on the class of antibody they bind:

1. FcγRs - bind to the Fc region of IgG antibodies
2. FcαRs - bind to the Fc region of IgA antibodies
3. FcεRs - bind to the Fc region of IgE antibodies

The binding of antibodies to Fc receptors triggers various cellular responses, such as phagocytosis, degranulation, and antibody-dependent cellular cytotoxicity (ADCC), which contribute to the elimination of pathogens, immune complexes, and other foreign substances. Dysregulation of Fc receptor function has been implicated in several diseases, including autoimmune disorders and allergies.

Immunoenzyme techniques are a group of laboratory methods used in immunology and clinical chemistry that combine the specificity of antibody-antigen reactions with the sensitivity and amplification capabilities of enzyme reactions. These techniques are primarily used for the detection, quantitation, or identification of various analytes (such as proteins, hormones, drugs, viruses, or bacteria) in biological samples.

In immunoenzyme techniques, an enzyme is linked to an antibody or antigen, creating a conjugate. This conjugate then interacts with the target analyte in the sample, forming an immune complex. The presence and amount of this immune complex can be visualized or measured by detecting the enzymatic activity associated with it.

There are several types of immunoenzyme techniques, including:

1. Enzyme-linked Immunosorbent Assay (ELISA): A widely used method for detecting and quantifying various analytes in a sample. In ELISA, an enzyme is attached to either the capture antibody or the detection antibody. After the immune complex formation, a substrate is added that reacts with the enzyme, producing a colored product that can be measured spectrophotometrically.
2. Immunoblotting (Western blot): A method used for detecting specific proteins in a complex mixture, such as a protein extract from cells or tissues. In this technique, proteins are separated by gel electrophoresis and transferred to a membrane, where they are probed with an enzyme-conjugated antibody directed against the target protein.
3. Immunohistochemistry (IHC): A method used for detecting specific antigens in tissue sections or cells. In IHC, an enzyme-conjugated primary or secondary antibody is applied to the sample, and the presence of the antigen is visualized using a chromogenic substrate that produces a colored product at the site of the antigen-antibody interaction.
4. Immunofluorescence (IF): A method used for detecting specific antigens in cells or tissues by employing fluorophore-conjugated antibodies. The presence of the antigen is visualized using a fluorescence microscope.
5. Enzyme-linked immunosorbent assay (ELISA): A method used for detecting and quantifying specific antigens or antibodies in liquid samples, such as serum or culture supernatants. In ELISA, an enzyme-conjugated detection antibody is added after the immune complex formation, and a substrate is added that reacts with the enzyme to produce a colored product that can be measured spectrophotometrically.

These techniques are widely used in research and diagnostic laboratories for various applications, including protein characterization, disease diagnosis, and monitoring treatment responses.

Immunoglobulin heavy chains are proteins that make up the framework of antibodies, which are Y-shaped immune proteins. These heavy chains, along with light chains, form the antigen-binding sites of an antibody, which recognize and bind to specific foreign substances (antigens) in order to neutralize or remove them from the body.

The heavy chain is composed of a variable region, which contains the antigen-binding site, and constant regions that determine the class and function of the antibody. There are five classes of immunoglobulins (IgA, IgD, IgE, IgG, and IgM) that differ in their heavy chain constant regions and therefore have different functions in the immune response.

Immunoglobulin heavy chains are synthesized by B cells, a type of white blood cell involved in the adaptive immune response. The genetic rearrangement of immunoglobulin heavy chain genes during B cell development results in the production of a vast array of different antibodies with unique antigen-binding sites, allowing for the recognition and elimination of a wide variety of pathogens.

Heterologous transplantation is a type of transplantation where an organ or tissue is transferred from one species to another. This is in contrast to allogeneic transplantation, where the donor and recipient are of the same species, or autologous transplantation, where the donor and recipient are the same individual.

In heterologous transplantation, the immune systems of the donor and recipient are significantly different, which can lead to a strong immune response against the transplanted organ or tissue. This is known as a graft-versus-host disease (GVHD), where the immune cells in the transplanted tissue attack the recipient's body.

Heterologous transplantation is not commonly performed in clinical medicine due to the high risk of rejection and GVHD. However, it may be used in research settings to study the biology of transplantation and to develop new therapies for transplant rejection.

Bacterial antigens are substances found on the surface or produced by bacteria that can stimulate an immune response in a host organism. These antigens can be proteins, polysaccharides, teichoic acids, lipopolysaccharides, or other molecules that are recognized as foreign by the host's immune system.

When a bacterial antigen is encountered by the host's immune system, it triggers a series of responses aimed at eliminating the bacteria and preventing infection. The host's immune system recognizes the antigen as foreign through the use of specialized receptors called pattern recognition receptors (PRRs), which are found on various immune cells such as macrophages, dendritic cells, and neutrophils.

Once a bacterial antigen is recognized by the host's immune system, it can stimulate both the innate and adaptive immune responses. The innate immune response involves the activation of inflammatory pathways, the recruitment of immune cells to the site of infection, and the production of antimicrobial peptides.

The adaptive immune response, on the other hand, involves the activation of T cells and B cells, which are specific to the bacterial antigen. These cells can recognize and remember the antigen, allowing for a more rapid and effective response upon subsequent exposures.

Bacterial antigens are important in the development of vaccines, as they can be used to stimulate an immune response without causing disease. By identifying specific bacterial antigens that are associated with virulence or pathogenicity, researchers can develop vaccines that target these antigens and provide protection against infection.

Surface antigens are molecules found on the surface of cells that can be recognized by the immune system as being foreign or different from the host's own cells. Antigens are typically proteins or polysaccharides that are capable of stimulating an immune response, leading to the production of antibodies and activation of immune cells such as T-cells.

Surface antigens are important in the context of infectious diseases because they allow the immune system to identify and target infected cells for destruction. For example, viruses and bacteria often display surface antigens that are distinct from those found on host cells, allowing the immune system to recognize and attack them. In some cases, these surface antigens can also be used as targets for vaccines or other immunotherapies.

In addition to their role in infectious diseases, surface antigens are also important in the context of cancer. Tumor cells often display abnormal surface antigens that differ from those found on normal cells, allowing the immune system to potentially recognize and attack them. However, tumors can also develop mechanisms to evade the immune system, making it difficult to mount an effective response.

Overall, understanding the properties and behavior of surface antigens is crucial for developing effective immunotherapies and vaccines against infectious diseases and cancer.

A base sequence in the context of molecular biology refers to the specific order of nucleotides in a DNA or RNA molecule. In DNA, these nucleotides are adenine (A), guanine (G), cytosine (C), and thymine (T). In RNA, uracil (U) takes the place of thymine. The base sequence contains genetic information that is transcribed into RNA and ultimately translated into proteins. It is the exact order of these bases that determines the genetic code and thus the function of the DNA or RNA molecule.

Immunoglobulin light chains are the smaller protein subunits of an immunoglobulin, also known as an antibody. They are composed of two polypeptide chains, called kappa (κ) and lambda (λ), which are produced by B cells during the immune response. Each immunoglobulin molecule contains either two kappa or two lambda light chains, in association with two heavy chains.

Light chains play a crucial role in the antigen-binding site of an antibody, where they contribute to the specificity and affinity of the interaction between the antibody and its target antigen. In addition to their role in immune function, abnormal production or accumulation of light chains can lead to various diseases, such as multiple myeloma and amyloidosis.

Protein binding, in the context of medical and biological sciences, refers to the interaction between a protein and another molecule (known as the ligand) that results in a stable complex. This process is often reversible and can be influenced by various factors such as pH, temperature, and concentration of the involved molecules.

In clinical chemistry, protein binding is particularly important when it comes to drugs, as many of them bind to proteins (especially albumin) in the bloodstream. The degree of protein binding can affect a drug's distribution, metabolism, and excretion, which in turn influence its therapeutic effectiveness and potential side effects.

Protein-bound drugs may be less available for interaction with their target tissues, as only the unbound or "free" fraction of the drug is active. Therefore, understanding protein binding can help optimize dosing regimens and minimize adverse reactions.

An immunoassay is a biochemical test that measures the presence or concentration of a specific protein, antibody, or antigen in a sample using the principles of antibody-antigen reactions. It is commonly used in clinical laboratories to diagnose and monitor various medical conditions such as infections, hormonal disorders, allergies, and cancer.

Immunoassays typically involve the use of labeled reagents, such as enzymes, radioisotopes, or fluorescent dyes, that bind specifically to the target molecule. The amount of label detected is proportional to the concentration of the target molecule in the sample, allowing for quantitative analysis.

There are several types of immunoassays, including enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), fluorescence immunoassay (FIA), and chemiluminescent immunoassay (CLIA). Each type has its own advantages and limitations, depending on the sensitivity, specificity, and throughput required for a particular application.

In the field of medicine, "time factors" refer to the duration of symptoms or time elapsed since the onset of a medical condition, which can have significant implications for diagnosis and treatment. Understanding time factors is crucial in determining the progression of a disease, evaluating the effectiveness of treatments, and making critical decisions regarding patient care.

For example, in stroke management, "time is brain," meaning that rapid intervention within a specific time frame (usually within 4.5 hours) is essential to administering tissue plasminogen activator (tPA), a clot-busting drug that can minimize brain damage and improve patient outcomes. Similarly, in trauma care, the "golden hour" concept emphasizes the importance of providing definitive care within the first 60 minutes after injury to increase survival rates and reduce morbidity.

Time factors also play a role in monitoring the progression of chronic conditions like diabetes or heart disease, where regular follow-ups and assessments help determine appropriate treatment adjustments and prevent complications. In infectious diseases, time factors are crucial for initiating antibiotic therapy and identifying potential outbreaks to control their spread.

Overall, "time factors" encompass the significance of recognizing and acting promptly in various medical scenarios to optimize patient outcomes and provide effective care.

"Competitive binding" is a term used in pharmacology and biochemistry to describe the behavior of two or more molecules (ligands) competing for the same binding site on a target protein or receptor. In this context, "binding" refers to the physical interaction between a ligand and its target.

When a ligand binds to a receptor, it can alter the receptor's function, either activating or inhibiting it. If multiple ligands compete for the same binding site, they will compete to bind to the receptor. The ability of each ligand to bind to the receptor is influenced by its affinity for the receptor, which is a measure of how strongly and specifically the ligand binds to the receptor.

In competitive binding, if one ligand is present in high concentrations, it can prevent other ligands with lower affinity from binding to the receptor. This is because the higher-affinity ligand will have a greater probability of occupying the binding site and blocking access to the other ligands. The competition between ligands can be described mathematically using equations such as the Langmuir isotherm, which describes the relationship between the concentration of ligand and the fraction of receptors that are occupied by the ligand.

Competitive binding is an important concept in drug development, as it can be used to predict how different drugs will interact with their targets and how they may affect each other's activity. By understanding the competitive binding properties of a drug, researchers can optimize its dosage and delivery to maximize its therapeutic effect while minimizing unwanted side effects.

Muromonab-CD3 is a type of immunosuppressant medication that is used in the treatment of acute organ rejection in patients who have received organ transplants. It is a monoclonal antibody that specifically targets and binds to the CD3 receptor found on the surface of T-lymphocytes, which are a type of white blood cell that plays a central role in the immune response.

By binding to the CD3 receptor, Muromonab-CD3 inhibits the activation and proliferation of T-lymphocytes, thereby suppressing the immune system's ability to recognize and attack the transplanted organ. This helps to prevent or reverse the process of acute organ rejection.

Muromonab-CD3 is administered intravenously and is typically given as a series of doses over several days. It may be used in combination with other immunosuppressive drugs to achieve optimal results. As with any medication, Muromonab-CD3 can have side effects, including fever, chills, nausea, and headache. More serious side effects, such as anaphylaxis or severe infections, may also occur, and patients should be closely monitored during treatment.

Antigens are substances (usually proteins) on the surface of cells, viruses, fungi, or bacteria that the immune system recognizes as foreign and mounts a response against.

Differentiation in the context of T-lymphocytes refers to the process by which immature T-cells mature and develop into different types of T-cells with specific functions, such as CD4+ helper T-cells or CD8+ cytotoxic T-cells.

T-lymphocytes, also known as T-cells, are a type of white blood cell that plays a central role in cell-mediated immunity. They are produced in the bone marrow and mature in the thymus gland. Once mature, they circulate throughout the body in search of foreign antigens to attack and destroy.

Therefore, 'Antigens, Differentiation, T-Lymphocyte' refers to the process by which T-lymphocytes mature and develop the ability to recognize and respond to specific foreign antigens.

Molecular weight, also known as molecular mass, is the mass of a molecule. It is expressed in units of atomic mass units (amu) or daltons (Da). Molecular weight is calculated by adding up the atomic weights of each atom in a molecule. It is a useful property in chemistry and biology, as it can be used to determine the concentration of a substance in a solution, or to calculate the amount of a substance that will react with another in a chemical reaction.

Monoclonal murine-derived antibodies are a type of laboratory-produced antibody that is identical in structure, having been derived from a single clone of cells. These antibodies are created using mouse cells and are therefore composed entirely of mouse immune proteins. They are designed to bind specifically to a particular target protein or antigen, making them useful tools for research, diagnostic testing, and therapeutic applications.

Monoclonal antibodies offer several advantages over polyclonal antibodies (which are derived from multiple clones of cells and can recognize multiple epitopes on an antigen). Monoclonal antibodies have a consistent and uniform structure, making them more reliable for research and diagnostic purposes. They also have higher specificity and affinity for their target antigens, allowing for more sensitive detection and measurement.

However, there are some limitations to using monoclonal murine-derived antibodies in therapeutic applications. Because they are composed entirely of mouse proteins, they can elicit an immune response in humans, leading to the production of human anti-mouse antibodies (HAMA) that can neutralize their effectiveness. To overcome this limitation, researchers have developed chimeric and humanized monoclonal antibodies that incorporate human protein sequences, reducing the risk of an immune response.

Immunoglobulins (Igs), also known as antibodies, are glycoprotein molecules produced by the immune system's B cells in response to the presence of foreign substances, such as bacteria, viruses, and toxins. These Y-shaped proteins play a crucial role in identifying and neutralizing pathogens and other antigens, thereby protecting the body against infection and disease.

Immunoglobulins are composed of four polypeptide chains: two identical heavy chains and two identical light chains, held together by disulfide bonds. The variable regions of these chains form the antigen-binding sites, which recognize and bind to specific epitopes on antigens. Based on their heavy chain type, immunoglobulins are classified into five main isotypes or classes: IgA, IgD, IgE, IgG, and IgM. Each class has distinct functions in the immune response, such as providing protection in different body fluids and tissues, mediating hypersensitivity reactions, and aiding in the development of immunological memory.

In medical settings, immunoglobulins can be administered therapeutically to provide passive immunity against certain diseases or to treat immune deficiencies, autoimmune disorders, and other conditions that may benefit from immunomodulation.

A dose-response relationship in immunology refers to the quantitative relationship between the dose or amount of an antigen (a substance that triggers an immune response) and the magnitude or strength of the resulting immune response. Generally, as the dose of an antigen increases, the intensity and/or duration of the immune response also increase, up to a certain point. This relationship helps in determining the optimal dosage for vaccines and immunotherapies, ensuring sufficient immune activation while minimizing potential adverse effects.

An antigen is any substance that can stimulate an immune response, particularly the production of antibodies. Viral antigens are antigens that are found on or produced by viruses. They can be proteins, glycoproteins, or carbohydrates present on the surface or inside the viral particle.

Viral antigens play a crucial role in the immune system's recognition and response to viral infections. When a virus infects a host cell, it may display its antigens on the surface of the infected cell. This allows the immune system to recognize and target the infected cells for destruction, thereby limiting the spread of the virus.

Viral antigens are also important targets for vaccines. Vaccines typically work by introducing a harmless form of a viral antigen to the body, which then stimulates the production of antibodies and memory T-cells that can recognize and respond quickly and effectively to future infections with the actual virus.

It's worth noting that different types of viruses have different antigens, and these antigens can vary between strains of the same virus. This is why there are often different vaccines available for different viral diseases, and why flu vaccines need to be updated every year to account for changes in the circulating influenza virus strains.

Immunoblotting, also known as western blotting, is a laboratory technique used in molecular biology and immunogenetics to detect and quantify specific proteins in a complex mixture. This technique combines the electrophoretic separation of proteins by gel electrophoresis with their detection using antibodies that recognize specific epitopes (protein fragments) on the target protein.

The process involves several steps: first, the protein sample is separated based on size through sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Next, the separated proteins are transferred onto a nitrocellulose or polyvinylidene fluoride (PVDF) membrane using an electric field. The membrane is then blocked with a blocking agent to prevent non-specific binding of antibodies.

After blocking, the membrane is incubated with a primary antibody that specifically recognizes the target protein. Following this, the membrane is washed to remove unbound primary antibodies and then incubated with a secondary antibody conjugated to an enzyme such as horseradish peroxidase (HRP) or alkaline phosphatase (AP). The enzyme catalyzes a colorimetric or chemiluminescent reaction that allows for the detection of the target protein.

Immunoblotting is widely used in research and clinical settings to study protein expression, post-translational modifications, protein-protein interactions, and disease biomarkers. It provides high specificity and sensitivity, making it a valuable tool for identifying and quantifying proteins in various biological samples.

Radioimmunoassay (RIA) is a highly sensitive analytical technique used in clinical and research laboratories to measure concentrations of various substances, such as hormones, vitamins, drugs, or tumor markers, in biological samples like blood, urine, or tissues. The method relies on the specific interaction between an antibody and its corresponding antigen, combined with the use of radioisotopes to quantify the amount of bound antigen.

In a typical RIA procedure, a known quantity of a radiolabeled antigen (also called tracer) is added to a sample containing an unknown concentration of the same unlabeled antigen. The mixture is then incubated with a specific antibody that binds to the antigen. During the incubation period, the antibody forms complexes with both the radiolabeled and unlabeled antigens.

After the incubation, the unbound (free) radiolabeled antigen is separated from the antibody-antigen complexes, usually through a precipitation or separation step involving centrifugation, filtration, or chromatography. The amount of radioactivity in the pellet (containing the antibody-antigen complexes) is then measured using a gamma counter or other suitable radiation detection device.

The concentration of the unlabeled antigen in the sample can be determined by comparing the ratio of bound to free radiolabeled antigen in the sample to a standard curve generated from known concentrations of unlabeled antigen and their corresponding bound/free ratios. The higher the concentration of unlabeled antigen in the sample, the lower the amount of radiolabeled antigen that will bind to the antibody, resulting in a lower bound/free ratio.

Radioimmunoassays offer high sensitivity, specificity, and accuracy, making them valuable tools for detecting and quantifying low levels of various substances in biological samples. However, due to concerns about radiation safety and waste disposal, alternative non-isotopic immunoassay techniques like enzyme-linked immunosorbent assays (ELISAs) have become more popular in recent years.

Cell adhesion molecules (CAMs) are a type of protein found on the surface of cells that mediate the attachment or adhesion of cells to either other cells or to the extracellular matrix (ECM), which is the network of proteins and carbohydrates that provides structural and biochemical support to surrounding cells.

CAMs play crucial roles in various biological processes, including tissue development, differentiation, repair, and maintenance of tissue architecture and function. They are also involved in cell signaling, migration, and regulation of the immune response.

There are several types of CAMs, classified based on their structure and function, such as immunoglobulin-like CAMs (IgCAMs), cadherins, integrins, and selectins. Dysregulation of CAMs has been implicated in various diseases, including cancer, inflammation, and neurological disorders.

The complement system is a group of proteins found in the blood and on the surface of cells that when activated, work together to help eliminate pathogens such as bacteria, viruses, and fungi from the body. The proteins are normally inactive in the bloodstream. When they encounter an invading microorganism or foreign substance, a series of reactions take place leading to the activation of the complement system. Activation results in the production of effector molecules that can punch holes in the cell membranes of pathogens, recruit and activate immune cells, and help remove debris and dead cells from the body.

There are three main pathways that can lead to complement activation: the classical pathway, the lectin pathway, and the alternative pathway. Each pathway involves a series of proteins that work together in a cascade-like manner to amplify the response and generate effector molecules. The three main effector molecules produced by the complement system are C3b, C4b, and C5b. These molecules can bind to the surface of pathogens, marking them for destruction by other immune cells.

Complement proteins also play a role in the regulation of the immune response. They help to prevent excessive activation of the complement system, which could damage host tissues. Dysregulation of the complement system has been implicated in a number of diseases, including autoimmune disorders and inflammatory conditions.

In summary, Complement System Proteins are a group of proteins that play a crucial role in the immune response by helping to eliminate pathogens and regulate the immune response. They can be activated through three different pathways, leading to the production of effector molecules that mark pathogens for destruction. Dysregulation of the complement system has been linked to various diseases.

Molecular cloning is a laboratory technique used to create multiple copies of a specific DNA sequence. This process involves several steps:

1. Isolation: The first step in molecular cloning is to isolate the DNA sequence of interest from the rest of the genomic DNA. This can be done using various methods such as PCR (polymerase chain reaction), restriction enzymes, or hybridization.
2. Vector construction: Once the DNA sequence of interest has been isolated, it must be inserted into a vector, which is a small circular DNA molecule that can replicate independently in a host cell. Common vectors used in molecular cloning include plasmids and phages.
3. Transformation: The constructed vector is then introduced into a host cell, usually a bacterial or yeast cell, through a process called transformation. This can be done using various methods such as electroporation or chemical transformation.
4. Selection: After transformation, the host cells are grown in selective media that allow only those cells containing the vector to grow. This ensures that the DNA sequence of interest has been successfully cloned into the vector.
5. Amplification: Once the host cells have been selected, they can be grown in large quantities to amplify the number of copies of the cloned DNA sequence.

Molecular cloning is a powerful tool in molecular biology and has numerous applications, including the production of recombinant proteins, gene therapy, functional analysis of genes, and genetic engineering.

Antineoplastic agents are a class of drugs used to treat malignant neoplasms or cancer. These agents work by inhibiting the growth and proliferation of cancer cells, either by killing them or preventing their division and replication. Antineoplastic agents can be classified based on their mechanism of action, such as alkylating agents, antimetabolites, topoisomerase inhibitors, mitotic inhibitors, and targeted therapy agents.

Alkylating agents work by adding alkyl groups to DNA, which can cause cross-linking of DNA strands and ultimately lead to cell death. Antimetabolites interfere with the metabolic processes necessary for DNA synthesis and replication, while topoisomerase inhibitors prevent the relaxation of supercoiled DNA during replication. Mitotic inhibitors disrupt the normal functioning of the mitotic spindle, which is essential for cell division. Targeted therapy agents are designed to target specific molecular abnormalities in cancer cells, such as mutated oncogenes or dysregulated signaling pathways.

It's important to note that antineoplastic agents can also affect normal cells and tissues, leading to various side effects such as nausea, vomiting, hair loss, and myelosuppression (suppression of bone marrow function). Therefore, the use of these drugs requires careful monitoring and management of their potential adverse effects.

Immunoconjugates are biomolecules created by the conjugation (coupling) of an antibody or antibody fragment with a cytotoxic agent, such as a drug, radionuclide, or toxin. This coupling is designed to direct the cytotoxic agent specifically to target cells, usually cancer cells, against which the antibody is directed, thereby increasing the effectiveness and reducing the side effects of the therapy.

The antibody part of the immunoconjugate recognizes and binds to specific antigens (proteins or other molecules) on the surface of the target cells, while the cytotoxic agent part enters the cell and disrupts its function, leading to cell death. The linker between the two parts is designed to be stable in circulation but can release the cytotoxic agent once inside the target cell.

Immunoconjugates are a promising area of research in targeted cancer therapy, as they offer the potential for more precise and less toxic treatments compared to traditional chemotherapy. However, their development and use also pose challenges, such as ensuring that the immunoconjugate binds specifically to the target cells and not to normal cells, optimizing the dose and schedule of treatment, and minimizing the risk of resistance to the therapy.

Sensitivity and specificity are statistical measures used to describe the performance of a diagnostic test or screening tool in identifying true positive and true negative results.

* Sensitivity refers to the proportion of people who have a particular condition (true positives) who are correctly identified by the test. It is also known as the "true positive rate" or "recall." A highly sensitive test will identify most or all of the people with the condition, but may also produce more false positives.
* Specificity refers to the proportion of people who do not have a particular condition (true negatives) who are correctly identified by the test. It is also known as the "true negative rate." A highly specific test will identify most or all of the people without the condition, but may also produce more false negatives.

In medical testing, both sensitivity and specificity are important considerations when evaluating a diagnostic test. High sensitivity is desirable for screening tests that aim to identify as many cases of a condition as possible, while high specificity is desirable for confirmatory tests that aim to rule out the condition in people who do not have it.

It's worth noting that sensitivity and specificity are often influenced by factors such as the prevalence of the condition in the population being tested, the threshold used to define a positive result, and the reliability and validity of the test itself. Therefore, it's important to consider these factors when interpreting the results of a diagnostic test.

Complement fixation tests are a type of laboratory test used in immunology and serology to detect the presence of antibodies in a patient's serum. These tests are based on the principle of complement activation, which is a part of the immune response. The complement system consists of a group of proteins that work together to help eliminate pathogens from the body.

In a complement fixation test, the patient's serum is mixed with a known antigen and complement proteins. If the patient has antibodies against the antigen, they will bind to it and activate the complement system. This results in the consumption or "fixation" of the complement proteins, which are no longer available to participate in a secondary reaction.

A second step involves adding a fresh source of complement proteins and a dye-labeled antibody that recognizes a specific component of the complement system. If complement was fixed during the first step, it will not be available for this secondary reaction, and the dye-labeled antibody will remain unbound. Conversely, if no antibodies were present in the patient's serum, the complement proteins would still be available for the second reaction, leading to the binding of the dye-labeled antibody.

The mixture is then examined under a microscope or using a spectrophotometer to determine whether the dye-labeled antibody has bound. If it has not, this indicates that the patient's serum contains antibodies specific to the antigen used in the test, and a positive result is recorded.

Complement fixation tests have been widely used for the diagnosis of various infectious diseases, such as syphilis, measles, and influenza. However, they have largely been replaced by more modern serological techniques, like enzyme-linked immunosorbent assays (ELISAs) and nucleic acid amplification tests (NAATs), due to their increased sensitivity, specificity, and ease of use.

"ErbB-2" is also known as "HER2" or "human epidermal growth factor receptor 2." It is a type of receptor tyrosine kinase (RTK) found on the surface of some cells. ErbB-2 does not bind to any known ligands, but it can form heterodimers with other ErbB family members, such as ErbB-3 and ErbB-4, which do have identified ligands. When a ligand binds to one of these receptors, it causes a conformational change that allows the ErbB-2 receptor to become activated through transphosphorylation. This activation triggers a signaling cascade that regulates cell growth, differentiation, and survival.

Overexpression or amplification of the ERBB2 gene, which encodes the ErbB-2 protein, is observed in approximately 20-30% of breast cancers and is associated with a more aggressive disease phenotype and poorer prognosis. Therefore, ErbB-2 has become an important target for cancer therapy, and several drugs that target this receptor have been developed, including trastuzumab (Herceptin), lapatinib (Tykerb), and pertuzumab (Perjeta).

Tumor-associated carbohydrate antigens (TACAs) are a type of tumor antigen that are expressed on the surface of cancer cells. These antigens are abnormal forms of carbohydrates, also known as glycans, which are attached to proteins and lipids on the cell surface.

TACAs are often overexpressed or expressed in a different form on cancer cells compared to normal cells. This makes them attractive targets for cancer immunotherapy because they can be recognized by the immune system as foreign and elicit an immune response. Some examples of TACAs include gangliosides, fucosylated glycans, and sialylated glycans.

Tumor-associated carbohydrate antigens have been studied as potential targets for cancer vaccines, antibody therapies, and other immunotherapeutic approaches. However, their use as targets for cancer therapy is still in the early stages of research and development.

A peptide fragment is a short chain of amino acids that is derived from a larger peptide or protein through various biological or chemical processes. These fragments can result from the natural breakdown of proteins in the body during regular physiological processes, such as digestion, or they can be produced experimentally in a laboratory setting for research or therapeutic purposes.

Peptide fragments are often used in research to map the structure and function of larger peptides and proteins, as well as to study their interactions with other molecules. In some cases, peptide fragments may also have biological activity of their own and can be developed into drugs or diagnostic tools. For example, certain peptide fragments derived from hormones or neurotransmitters may bind to receptors in the body and mimic or block the effects of the full-length molecule.

Hemagglutination tests are laboratory procedures used to detect the presence of antibodies or antigens in a sample, typically in blood serum. These tests rely on the ability of certain substances, such as viruses or bacteria, to agglutinate (clump together) red blood cells.

In a hemagglutination test, a small amount of the patient's serum is mixed with a known quantity of red blood cells that have been treated with a specific antigen. If the patient has antibodies against that antigen in their serum, they will bind to the antigens on the red blood cells and cause them to agglutinate. This clumping can be observed visually, indicating a positive test result.

Hemagglutination tests are commonly used to diagnose infectious diseases caused by viruses or bacteria that have hemagglutinating properties, such as influenza, parainfluenza, and HIV. They can also be used in blood typing and cross-matching before transfusions.

Hemagglutination inhibition (HI) tests are a type of serological assay used in medical laboratories to detect and measure the amount of antibodies present in a patient's serum. These tests are commonly used to diagnose viral infections, such as influenza or HIV, by identifying the presence of antibodies that bind to specific viral antigens and prevent hemagglutination (the agglutination or clumping together of red blood cells).

In an HI test, a small amount of the patient's serum is mixed with a known quantity of the viral antigen, which has been treated to attach to red blood cells. If the patient's serum contains antibodies that bind to the viral antigen, they will prevent the antigen from attaching to the red blood cells and inhibit hemagglutination. The degree of hemagglutination inhibition can be measured and used to estimate the amount of antibody present in the patient's serum.

HI tests are relatively simple and inexpensive to perform, but they have some limitations. For example, they may not detect early-stage infections before the body has had a chance to produce antibodies, and they may not be able to distinguish between different strains of the same virus. Nonetheless, HI tests remain an important tool for diagnosing viral infections and monitoring immune responses to vaccination or infection.

A genetic vector is a vehicle, often a plasmid or a virus, that is used to introduce foreign DNA into a host cell as part of genetic engineering or gene therapy techniques. The vector contains the desired gene or genes, along with regulatory elements such as promoters and enhancers, which are needed for the expression of the gene in the target cells.

The choice of vector depends on several factors, including the size of the DNA to be inserted, the type of cell to be targeted, and the efficiency of uptake and expression required. Commonly used vectors include plasmids, adenoviruses, retroviruses, and lentiviruses.

Plasmids are small circular DNA molecules that can replicate independently in bacteria. They are often used as cloning vectors to amplify and manipulate DNA fragments. Adenoviruses are double-stranded DNA viruses that infect a wide range of host cells, including human cells. They are commonly used as gene therapy vectors because they can efficiently transfer genes into both dividing and non-dividing cells.

Retroviruses and lentiviruses are RNA viruses that integrate their genetic material into the host cell's genome. This allows for stable expression of the transgene over time. Lentiviruses, a subclass of retroviruses, have the advantage of being able to infect non-dividing cells, making them useful for gene therapy applications in post-mitotic tissues such as neurons and muscle cells.

Overall, genetic vectors play a crucial role in modern molecular biology and medicine, enabling researchers to study gene function, develop new therapies, and modify organisms for various purposes.

Antineutrophil cytoplasmic antibodies (ANCAs) are a type of autoantibody that specifically target certain proteins in the cytoplasm of neutrophils, which are a type of white blood cell. These antibodies are associated with several types of vasculitis, which is inflammation of the blood vessels.

There are two main types of ANCAs: perinuclear ANCAs (p-ANCAs) and cytoplasmic ANCAs (c-ANCAs). p-ANCAs are directed against myeloperoxidase, a protein found in neutrophil granules, while c-ANCAs target proteinase 3, another protein found in neutrophil granules.

The presence of ANCAs in the blood can indicate an increased risk for developing certain types of vasculitis, such as granulomatosis with polyangiitis (GPA), eosinophilic granulomatosis with polyangiitis (EGPA), and microscopic polyangiitis (MPA). ANCA testing is often used in conjunction with other clinical findings to help diagnose and manage these conditions.

It's important to note that while the presence of ANCAs can indicate an increased risk for vasculitis, not everyone with ANCAs will develop the condition. Additionally, ANCAs can also be found in some individuals without any associated disease, so their presence should be interpreted in the context of other clinical findings.

Peptides are short chains of amino acid residues linked by covalent bonds, known as peptide bonds. They are formed when two or more amino acids are joined together through a condensation reaction, which results in the elimination of a water molecule and the formation of an amide bond between the carboxyl group of one amino acid and the amino group of another.

Peptides can vary in length from two to about fifty amino acids, and they are often classified based on their size. For example, dipeptides contain two amino acids, tripeptides contain three, and so on. Oligopeptides typically contain up to ten amino acids, while polypeptides can contain dozens or even hundreds of amino acids.

Peptides play many important roles in the body, including serving as hormones, neurotransmitters, enzymes, and antibiotics. They are also used in medical research and therapeutic applications, such as drug delivery and tissue engineering.

Seroepidemiologic studies are a type of epidemiological study that measures the presence and levels of antibodies in a population's blood serum to investigate the prevalence, distribution, and transmission of infectious diseases. These studies help to identify patterns of infection and immunity within a population, which can inform public health policies and interventions.

Seroepidemiologic studies typically involve collecting blood samples from a representative sample of individuals in a population and testing them for the presence of antibodies against specific pathogens. The results are then analyzed to estimate the prevalence of infection and immunity within the population, as well as any factors associated with increased or decreased risk of infection.

These studies can provide valuable insights into the spread of infectious diseases, including emerging and re-emerging infections, and help to monitor the effectiveness of vaccination programs. Additionally, seroepidemiologic studies can also be used to investigate the transmission dynamics of infectious agents, such as identifying sources of infection or tracking the spread of antibiotic resistance.

C57BL/6 (C57 Black 6) is an inbred strain of laboratory mouse that is widely used in biomedical research. The term "inbred" refers to a strain of animals where matings have been carried out between siblings or other closely related individuals for many generations, resulting in a population that is highly homozygous at most genetic loci.

The C57BL/6 strain was established in 1920 by crossing a female mouse from the dilute brown (DBA) strain with a male mouse from the black strain. The resulting offspring were then interbred for many generations to create the inbred C57BL/6 strain.

C57BL/6 mice are known for their robust health, longevity, and ease of handling, making them a popular choice for researchers. They have been used in a wide range of biomedical research areas, including studies of cancer, immunology, neuroscience, cardiovascular disease, and metabolism.

One of the most notable features of the C57BL/6 strain is its sensitivity to certain genetic modifications, such as the introduction of mutations that lead to obesity or impaired glucose tolerance. This has made it a valuable tool for studying the genetic basis of complex diseases and traits.

Overall, the C57BL/6 inbred mouse strain is an important model organism in biomedical research, providing a valuable resource for understanding the genetic and molecular mechanisms underlying human health and disease.

The Epidermal Growth Factor Receptor (EGFR) is a type of receptor found on the surface of many cells in the body, including those of the epidermis or outer layer of the skin. It is a transmembrane protein that has an extracellular ligand-binding domain and an intracellular tyrosine kinase domain.

EGFR plays a crucial role in various cellular processes such as proliferation, differentiation, migration, and survival. When EGF (Epidermal Growth Factor) or other ligands bind to the extracellular domain of EGFR, it causes the receptor to dimerize and activate its intrinsic tyrosine kinase activity. This leads to the autophosphorylation of specific tyrosine residues on the receptor, which in turn recruits and activates various downstream signaling molecules, resulting in a cascade of intracellular signaling events that ultimately regulate gene expression and cell behavior.

Abnormal activation of EGFR has been implicated in several human diseases, including cancer. Overexpression or mutation of EGFR can lead to uncontrolled cell growth and division, angiogenesis, and metastasis, making it an important target for cancer therapy.

Immunoglobulin idiotypes refer to the unique antigenic determinants found on the variable regions of an immunoglobulin (antibody) molecule. These determinants are specific to each individual antibody and can be used to distinguish between different antibodies produced by a single individual or between antibodies produced by different individuals.

The variable region of an antibody is responsible for recognizing and binding to a specific antigen. The amino acid sequence in this region varies between different antibodies, and it is these variations that give rise to the unique idiotypes. Idiotypes can be used as markers to study the immune response, including the clonal selection and affinity maturation of B cells during an immune response.

Immunoglobulin idiotypes are also important in the development of monoclonal antibodies for therapeutic use. By identifying and isolating a specific antibody with the desired idiotype, it is possible to produce large quantities of identical antibodies that can be used to treat various diseases, including cancer and autoimmune disorders.

Immunologic techniques are a group of laboratory methods that utilize the immune system's ability to recognize and respond to specific molecules, known as antigens. These techniques are widely used in medicine, biology, and research to detect, measure, or identify various substances, including proteins, hormones, viruses, bacteria, and other antigens.

Some common immunologic techniques include:

1. Enzyme-linked Immunosorbent Assay (ELISA): A sensitive assay used to detect and quantify antigens or antibodies in a sample. This technique uses an enzyme linked to an antibody or antigen, which reacts with a substrate to produce a colored product that can be measured and quantified.
2. Immunofluorescence: A microscopic technique used to visualize the location of antigens or antibodies in tissues or cells. This technique uses fluorescent dyes conjugated to antibodies, which bind to specific antigens and emit light when excited by a specific wavelength of light.
3. Western Blotting: A laboratory technique used to detect and identify specific proteins in a sample. This technique involves separating proteins based on their size using electrophoresis, transferring them to a membrane, and then probing the membrane with antibodies that recognize the protein of interest.
4. Immunoprecipitation: A laboratory technique used to isolate and purify specific antigens or antibodies from a complex mixture. This technique involves incubating the mixture with an antibody that recognizes the antigen or antibody of interest, followed by precipitation of the antigen-antibody complex using a variety of methods.
5. Radioimmunoassay (RIA): A sensitive assay used to detect and quantify antigens or antibodies in a sample. This technique uses radioactively labeled antigens or antibodies, which bind to specific antigens or antibodies in the sample, allowing for detection and quantification using a scintillation counter.

These techniques are important tools in medical diagnosis, research, and forensic science.

Affinity chromatography is a type of chromatography technique used in biochemistry and molecular biology to separate and purify proteins based on their biological characteristics, such as their ability to bind specifically to certain ligands or molecules. This method utilizes a stationary phase that is coated with a specific ligand (e.g., an antibody, antigen, receptor, or enzyme) that selectively interacts with the target protein in a sample.

The process typically involves the following steps:

1. Preparation of the affinity chromatography column: The stationary phase, usually a solid matrix such as agarose beads or magnetic beads, is modified by covalently attaching the ligand to its surface.
2. Application of the sample: The protein mixture is applied to the top of the affinity chromatography column, allowing it to flow through the stationary phase under gravity or pressure.
3. Binding and washing: As the sample flows through the column, the target protein selectively binds to the ligand on the stationary phase, while other proteins and impurities pass through. The column is then washed with a suitable buffer to remove any unbound proteins and contaminants.
4. Elution of the bound protein: The target protein can be eluted from the column using various methods, such as changing the pH, ionic strength, or polarity of the buffer, or by introducing a competitive ligand that displaces the bound protein.
5. Collection and analysis: The eluted protein fraction is collected and analyzed for purity and identity, often through techniques like SDS-PAGE or mass spectrometry.

Affinity chromatography is a powerful tool in biochemistry and molecular biology due to its high selectivity and specificity, enabling the efficient isolation of target proteins from complex mixtures. However, it requires careful consideration of the binding affinity between the ligand and the protein, as well as optimization of the elution conditions to minimize potential damage or denaturation of the purified protein.

Antigens are substances that can stimulate an immune response, particularly the production of antibodies by B-lymphocytes. Differentiation refers to the process by which cells mature and become more specialized in their functions. In the context of B-lymphocytes, differentiation involves the maturation of naive B-cells into plasma cells that are capable of producing large amounts of antibodies in response to an antigenic stimulus.

B-lymphocytes, also known as B-cells, are a type of white blood cell that plays a critical role in the adaptive immune system. They are responsible for producing antibodies, which are proteins that recognize and bind to specific antigens, marking them for destruction by other immune cells.

When a B-lymphocyte encounters an antigen, it becomes activated and begins to differentiate into a plasma cell. During this process, the B-cell undergoes several changes, including an increase in size, the expression of new surface receptors, and the production of large amounts of antibodies specific to the antigen. These antibodies are then released into the bloodstream, where they can bind to the antigen and help to neutralize or eliminate it.

Overall, the differentiation of B-lymphocytes in response to antigens is a critical component of the adaptive immune system, allowing the body to mount targeted responses to specific pathogens and other foreign substances.

Ribosome-inactivating proteins (RIPs) are a type of protein that can inhibit the function of ribosomes, which are the cellular structures responsible for protein synthesis. Ribosome-inactivating proteins are classified into two types: Type 1 and Type 2.

Type 1 Ribosome-Inactivating Proteins (RIPs) are defined as single-chain proteins that inhibit protein synthesis by depurinating a specific adenine residue in the sarcin-ricin loop of the large rRNA molecule within the ribosome. This results in the irreversible inactivation of the ribosome, preventing it from participating in further protein synthesis.

Type 1 RIPs are found in various plant species and have been identified as potential therapeutic agents for cancer treatment due to their ability to selectively inhibit protein synthesis in cancer cells. However, they can also be toxic to normal cells, which limits their clinical use. Examples of Type 1 RIPs include dianthin, gelonin, and trichosanthin.

'Escherichia coli' (E. coli) is a type of gram-negative, facultatively anaerobic, rod-shaped bacterium that commonly inhabits the intestinal tract of humans and warm-blooded animals. It is a member of the family Enterobacteriaceae and one of the most well-studied prokaryotic model organisms in molecular biology.

While most E. coli strains are harmless and even beneficial to their hosts, some serotypes can cause various forms of gastrointestinal and extraintestinal illnesses in humans and animals. These pathogenic strains possess virulence factors that enable them to colonize and damage host tissues, leading to diseases such as diarrhea, urinary tract infections, pneumonia, and sepsis.

E. coli is a versatile organism with remarkable genetic diversity, which allows it to adapt to various environmental niches. It can be found in water, soil, food, and various man-made environments, making it an essential indicator of fecal contamination and a common cause of foodborne illnesses. The study of E. coli has contributed significantly to our understanding of fundamental biological processes, including DNA replication, gene regulation, and protein synthesis.

Immunosorbent techniques are a group of laboratory methods used in immunology and clinical chemistry to isolate or detect specific proteins, antibodies, or antigens from a complex mixture. These techniques utilize the specific binding properties of antibodies or antigens to capture and concentrate target molecules.

The most common immunosorbent technique is the Enzyme-Linked Immunosorbent Assay (ELISA), which involves coating a solid surface with a capture antibody, allowing the sample to bind, washing away unbound material, and then detecting bound antigens or antibodies using an enzyme-conjugated detection reagent. The enzyme catalyzes a colorimetric reaction that can be measured and quantified, providing a sensitive and specific assay for the target molecule.

Other immunosorbent techniques include Radioimmunoassay (RIA), Immunofluorescence Assay (IFA), and Lateral Flow Immunoassay (LFIA). These methods have wide-ranging applications in research, diagnostics, and drug development.

Lymphokine-activated killer (LAK) cells are a type of immune cell that has been activated to kill certain types of cells, including cancer cells and virus-infected cells. They are called "lymphokine-activated" because they are activated through the action of lymphokines, which are proteins secreted by other immune cells. LAK cells are a type of natural killer (NK) cell, which are a type of white blood cell that plays a role in the body's defense against viruses and cancer.

LAK cells are generated in the laboratory by incubating peripheral blood mononuclear cells (PBMCs), which include lymphocytes and monocytes, with high concentrations of interleukin-2 (IL-2) for several days. This process activates and expands the population of NK cells, resulting in the formation of LAK cells. These activated cells are then able to recognize and kill a wide range of tumor cells and virus-infected cells, regardless of whether they express specific antigens or not.

LAK cell therapy is an experimental form of cancer treatment that involves infusing patients with large numbers of LAK cells in order to enhance their immune response against cancer. While some studies have shown promising results, more research is needed to determine the safety and effectiveness of this approach.

A "camel" is a large, even-toed ungulate that belongs to the genus Camelus in the family Camelidae. There are two species of camels: the dromedary camel (Camelus dromedarius), also known as the Arabian camel, which has one hump, and the Bactrian camel (Camelus bactrianus), which has two humps.

Camels are well adapted to life in arid environments and are native to the Middle East and Central Asia. They have long legs, large, flat feet that help them walk on sand, and a thick coat of hair that helps protect them from the sun and cold temperatures. Camels are also known for their ability to store fat in their humps, which they can convert into water and energy when food and water are scarce.

Camels have been domesticated for thousands of years and have played an important role in human history as transportation, pack animals, and sources of meat, milk, and wool. They are also used in traditional medicine and religious ceremonies in some cultures.

The spleen is an organ in the upper left side of the abdomen, next to the stomach and behind the ribs. It plays multiple supporting roles in the body:

1. It fights infection by acting as a filter for the blood. Old red blood cells are recycled in the spleen, and platelets and white blood cells are stored there.
2. The spleen also helps to control the amount of blood in the body by removing excess red blood cells and storing platelets.
3. It has an important role in immune function, producing antibodies and removing microorganisms and damaged red blood cells from the bloodstream.

The spleen can be removed without causing any significant problems, as other organs take over its functions. This is known as a splenectomy and may be necessary if the spleen is damaged or diseased.

In the context of medicine and pharmacology, "kinetics" refers to the study of how a drug moves throughout the body, including its absorption, distribution, metabolism, and excretion (often abbreviated as ADME). This field is called "pharmacokinetics."

1. Absorption: This is the process of a drug moving from its site of administration into the bloodstream. Factors such as the route of administration (e.g., oral, intravenous, etc.), formulation, and individual physiological differences can affect absorption.

2. Distribution: Once a drug is in the bloodstream, it gets distributed throughout the body to various tissues and organs. This process is influenced by factors like blood flow, protein binding, and lipid solubility of the drug.

3. Metabolism: Drugs are often chemically modified in the body, typically in the liver, through processes known as metabolism. These changes can lead to the formation of active or inactive metabolites, which may then be further distributed, excreted, or undergo additional metabolic transformations.

4. Excretion: This is the process by which drugs and their metabolites are eliminated from the body, primarily through the kidneys (urine) and the liver (bile).

Understanding the kinetics of a drug is crucial for determining its optimal dosing regimen, potential interactions with other medications or foods, and any necessary adjustments for special populations like pediatric or geriatric patients, or those with impaired renal or hepatic function.

... entry in the public domain NCI Dictionary of Cancer Terms Bispecific+antibodies at the U.S. ... Suurs FV, Lub-de Hooge MN, de Vries EG, de Groot DJ (September 2019). "A review of bispecific antibodies and antibody ... The first bispecific antibody to gain regulatory approval, blinatumomab, targets CD19 on B cells and CD3 on T cells, leading to ... Bispecific antibodies have a wide variety of applications in diagnosis and therapy. BsAbs can be combined with HRPO, can be ...
"Zymeworks Announces Updated Single Agent Data for HER2-Targeted Bispecific Antibody ZW25 at European Society for Medical ... Holmes, David (2011). "Buy Buy Bispecific Antibodies". Nature Reviews Drug Discovery. 10 (11): 798-800. doi:10.1038/nrd3581. ... "Improving biophysical properties of a bispecific antibody scaffold to aid developability". mAbs. 5 (5): 646-654. doi:10.4161/ ... development and commercialization of bispecific antibodies across multiple disease areas. The extended agreement has meant that ...
These bispecific monoclonal antibodies are sometimes referred to as mAb2. Antibody Fab region Protein tag Janeway, CA, Jr.; et ... "MAb2 â„¢Bispecific Monoclonal Antibodies". Archived from the original on 2013-07-08. Retrieved 2013-08-13. (CS1 maint: multiple ... Fcab fragments can be inserted into a full immunoglobulin by swapping the Fc region, thus obtaining a bispecific antibody (with ... This region allows antibodies to activate the immune system, for example, through binding to Fc receptors. In IgG, IgA and IgD ...
"Bispecific Antibodies - Dual Targeting". N.V, Affimed (2015-06-23). "Affimed Announces Opening of US Operations and Key US- ... Kovacsovics-Bankowski, M.; Carini, C.; Kashala, O.; Essex, M. (1992). "Isotypic distribution of HIV-1-specific antibodies in ... correlation between HIV-1 cross-reactivity and antibodies to lipoarabinomannan". The Journal of Infectious Diseases. 169 (2): ...
... and there is extensive research for bispecific antibodies. Applications of bispecific antibodies cover a broad spectrum that ... Brinkmann, Ulrich; Kontermann, Roland E. (2017-02-17). "The making of bispecific antibodies". mAbs. 9 (2): 182-212. doi:10.1080 ... small bivalent and bispecific antibody fragments". Proceedings of the National Academy of Sciences. 90 (14): 6444-6448. Bibcode ... The most common example for multivalent binding are the antibodies, ...
Trifunctional antibodies were the first type of bispecific monoclonal antibodies to be produced. The first concepts date back ... antibody as well as pure mouse and pure rat antibody. The trifunctional antibody is extracted chromatographically with protein ... Chames, P; Baty, D (2009). "Bispecific antibodies for cancer therapy: the light at the end of the tunnel". mAbs. 1 (6): 1-9. ... Choi, BD; et, al.; Bigner, DD; Mehta, AI; Kuan, CT; Sampson, JH (2011). "Bispecific antibodies engage T cells for antitumor ...
The furthest developed of these are bispecific tandem di-scFvs, known as bi-specific T-cell engagers (BiTE antibody constructs ... "Expression and purification of monospecific and bispecific recombinant antibody fragments derived from antibodies that block ... Kufer, Peter; Lutterbüse, Ralf; Baeuerle, Patrick A. (2004). "A revival of bispecific antibodies" (PDF). Trends in ... Hollinger, Philipp; Prospero, T; Winter, G (July 1993). ""Diabodies": small bivalent and bispecific antibody fragments". ...
Bispecific antibodies combine two different antigen binding specificities within one molecule. The bispecific antibodies are ... The most commonly used are the scFv, Fab fragments and bispecific antibodies. scFv is the smallest of the recombinant antibody ... Recombinant antibodies are antibody fragments produced by using recombinant antibody coding genes. They mostly consist of a ... scFv or somatically fused antibodies create a bispecific antibody. The most commonly applied technology to produce recombinant ...
Baeuerle, Patrick A.; Reinhardt, Carsten (15 June 2009). "Bispecific T-Cell Engaging Antibodies for Cancer Therapy". Cancer ... Baeuerle, Patrick A.; Reinhardt, Carsten (15 June 2009). "Bispecific T-Cell Engaging Antibodies for Cancer Therapy". Cancer ... and the development of bispecific T-cell engaging antibodies for therapy of cancer. Baeuerle earned his diploma in biology from ... "Reinventing the Antibody". The Scientist. Retrieved 2017-10-04. Perkel, Jeffrey (14 August 2008). "New Lymphoma Drug Shows ...
An antibody can be called monospecific if it has specificity for the same antigen or epitope, or bispecific if they have ... antibody Neutralizing antibody Optimer Ligand Secondary antibodies Single-domain antibody Slope spectroscopy Synthetic antibody ... Antibody fragments, such as Fab and nanobodies are not considered as antibody mimetics. Common advantages over antibodies are ... Affimer Anti-mitochondrial antibodies Anti-nuclear antibodies Antibody mimetic Aptamer Colostrum ELISA Humoral immunity ...
31 (4). Guy DG, Uy GL (October 2018). "Bispecific Antibodies for the Treatment of Acute Myeloid Leukemia". Curr Hematol Malig ... Drugs that are a monoclonal antibody, All stub articles, Monoclonal antibody stubs, Monoclonal antibodies). ... Flotetuzumab (INN; development code MGD006) is a bispecific antibody designed for the treatment of acute myeloid leukemia. This ... a focus on monoclonal antibodies and immune checkpoint inhibitors". Curr. Opin. Hematol. 25 (2): 136-145. doi:10.1097/MOH. ...
Carvalho, Thiago (25 January 2023). "FDA approves Genentech's bispecific antibody for lymphoma". Nature Medicine. 29 (3): 507- ... Drugs that are a monoclonal antibody, Breakthrough therapy, Monoclonal antibodies for tumors, Orphan drugs, Genentech brands, ... Mosunetuzumab, sold under the brand name Lunsumio, is a monoclonal antibody used for the treatment of follicular lymphoma. It ... including an anti-CD20 monoclonal antibody and an alkylating agent. The US Food and Drug Administration (FDA) granted the ...
Holliger, Philipp (1994). Multivalent and bispecific antibody fragments from E.coli: new strategies for antibody-based ... Holliger, P.; Prospero, T.; Winter, G. (15 July 1993). ""Diabodies": small bivalent and bispecific antibody fragments". ... Holliger developed a new type of bispecific antibody fragment, called a diabody and worked on elucidating the infection pathway ...
The beginning of the pretargeting concept was based on bispecific antibodies which were able to bind a specific target antigen ... Owing to the high molecular weight of antibodies and the Fc domain of the antibody, a slow clearance from the blood and non- ... However, these types of antibodies turned out to be quite troublesome, due to the triggering of the human anti-murine antibody ... "Recombinant Bispecific Monoclonal Antibodies Prepared by the Dock-and-Lock Strategy for Pretargeted Radioimmunotherapy". ...
Also in 2022, Samsung Biologics launched S-DUAL, a bispecific antibody platform, and its DEVELOPICK platform, a service that ... Byung-yeul, Baek (7 October 2022). "Samsung Biologics launches bispecific antibody platform S-DUAL". Korea Times. "New Samsung ... "Lilly partners with Samsung to boost COVID-19 antibody manufacturing amid supply concerns". Fierce Pharma. "Samsung Biologics ...
He received his doctorate in 1992 with a thesis on immunotherapy against tumor cells (bispecific monoclonal antibodies for the ... 12 June 2017). "Elimination of large tumors in mice by mRNA-encoded bispecific antibodies". Nature Medicine. 23 (7): 815-817. ... Antibody Shines in Advanced Gastric Cancer, 5 June 2016, retrieved 26 October 2020 "A Study of Zolbetuximab (IMAB362) Plus ... Ganymed developed the monoclonal antibody Zolbetuximab, for use against esophageal and gastrointestinal cancer. In 2016, after ...
Another technology recently developed is the OkapY bispecific antibody technology. MorphoSys' OkapY technology is a new ... bispecific antibody format with the hysicochemical properties to simplify the development and large-scale production of such ... Two antibodies developed by MorphoSys are already approved and marketed. Tafasitamab (MOR208) is a humanized monoclonal ... The company has various antibody, protein and peptide technologies that it uses to discover and develop both proprietary and ...
Bispecific Antibody Traps". Journal of the American Chemical Society. 142 (41): 17703-17713. doi:10.1021/jacs.0c08458. ISSN ... antibody-based chemically induced dimerizers (AbCID), antibody-Based PROTACs (AbTAC), antibody targeting a proteolytic ... "Recombinant Antibody Network". recombinant-antibodies.org. Retrieved 15 November 2022. Martinko, Alexander J.; Truillet, ... devoted to generating human recombinant antibodies at a proteome-wide scale using high throughput platforms for antibody phage ...
... (MM-141) is an experimental monoclonal antibody for the treatment of cancer. It is a bispecific antibody targeting ... Drugs that are a monoclonal antibody, Experimental cancer drugs, Monoclonal antibodies, Orphan drugs, All stub articles, ...
CD79B The bispecific antibodies have yielded promising results in clinical trials. In April 2009, the bispecific antibody ... Chames P, Baty D (2009). "Bispecific antibodies for cancer therapy: the light at the end of the tunnel?". mAbs. 1 (6): 539-47. ... Antibody-drug conjugates (ADCs) are antibodies linked to one or more drug molecules. Typically when the ADC meets the target ... Murine antibodies in vitro are thereby transformed into fully human antibodies. The heavy and light chains of human IgG ...
It is a bispecific GPRC5D-directed CD3 T-cell engager. Talquetamab is a bispecific antibody against two targets: human CD3, a T ... December 2022). "Talquetamab, a T-Cell-Redirecting GPRC5D Bispecific Antibody for Multiple Myeloma". The New England Journal of ... Drugs that are a monoclonal antibody, Breakthrough therapy, Monoclonal antibodies for tumors, Orphan drugs, All stub articles, ... Talquetamab, sold under the brand name Talvey, is a humanized monoclonal antibody used for the treatment of multiple myeloma. ...
Chari, Ajai (15 December 2022). "Talquetamab, a T-Cell-Redirecting GPRC5D Bispecific Antibody for Multiple Myeloma". New ... or sometimes simply bispecific antibodies. The first drug in this class approved for multiple myeloma is Teclistamab, but again ... "Infections and Severe Hypogammaglobulinemia in Multiple Myeloma Patients Treated with Anti-BCMA Bispecific Antibodies". ... An upcoming therapy that also leverages T-cell immune response is the class of drugs termed bispecific T-cell engagers (BITE), ...
"CDR loop interactions can determine heavy and light chain pairing preferences in bispecific antibodies". mAbs. 14 (1): 2024118 ... DeCaprio, James; Kohl, Thomas O. (2019-02-01). "Cross-Linking Antibodies to Beads with Disuccinimidyl Suberate (DSS)". Cold ...
... is a bispecific monoclonal antibody used for the treatment of diffuse large B-cell lymphoma. It is a bispecific CD20-directed ... application of CrossMab technology for the development of therapeutic bispecific antibodies and antibody fusion proteins". mAbs ... "FDA approves Roche's Columvi, the first and only bispecific antibody with a fixed-duration treatment for people with relapsed ... Salvaris R, Ong J, Gregory GP (April 2021). "Bispecific Antibodies: A Review of Development, Clinical Efficacy and Toxicity in ...
One of them is development of bispecific antibodies such as CD3/CLL-1 antibody. It can recruit unstimulated primary T cells in ... February 2017). "An anti-CD3/anti-CLL-1 bispecific antibody for the treatment of acute myeloid leukemia". Blood. 129 (5): 609- ...
... is a bispecific monoclonal antibody used to treat non-small cell lung cancer. Amivantamab is a bispecific epidermal growth ... Clinical trial number NCT02609776 for "Study of Amivantamab, a Human Bispecific EGFR and cMet Antibody, in Participants With ... August 2020). "Antitumor Activity of Amivantamab (JNJ-61186372), an EGFR-MET Bispecific Antibody, in Diverse Models of EGFR ... April 2021). "Discovery of amivantamab (JNJ-61186372), a bispecific antibody targeting EGFR and MET". The Journal of Biological ...
2014). "Generation of bispecific IgG antibodies by structure-based design of an orthogonal Fab interface". Nature Biotechnology ... designed fusions for bispecific antibodies, and progress toward vaccines. DeLano Award for Computational Biosciences Feynman ...
This approach is similar to the one taken in the development of bispecific monoclonal antibodies. In a study, the plasma half- ... Since they are not structurally related to antibodies, they are classified as a type of antibody mimetic. Avimers have been ... Half-life can be increased by binding them to antibodies. A library theoretically containing up to 1023 different A domains ... They have improved heat stability compared with antibodies, but limited plasma half-life because of their smaller size. ...
Morecki S, Lindhofer H, Yacovlev E, Gelfand Y, Slavin S (Feb 2006). "Use of trifunctional bispecific antibodies to prevent ... Krolick KA, Uhr JW, Slavin S, Vitetta ES (Jun 1982). "In vivo therapy of a murine B cell tumor (BCL1) using antibody-ricin A ... Herman Waldmann, Slavin was the first to introduce the use of an anti-CD52 monoclonal antibody (CAMPATH-1; Alemtuzumab and ... focusing on the use of activated donor lymphocytes targeted against cancer with monoclonal and bispecific antibodies for ...
Genentech was able to demonstrate in mouse models that the new bispecific antibody was able to reach therapeutic levels in the ... They utilized a mouse bispecific antibody with two active sites performing different functions. One arm had a low-affinity anti ... Researchers at Genentech proposed the creation of a bispecific antibody that could bind the BBB membrane, induce receptor- ... This way, the amount of transported antibody is based on the concentration of antibody on either side of the barrier. The other ...
  • In 1975, the problem of producing pure antibodies was solved by the creation of hybridoma technology, and the new era of monoclonal antibodies (MoAbs) came. (wikipedia.org)
  • The bispecific antibody demonstrated pharmacokinetic properties similar to those of conventional monoclonal antibodies, like rituximab. (pharmaceutical-journal.com)
  • CD20-TDB administration in cynomolgus monkeys demonstrated a pharmacokinetic profile typical of IgG1 monoclonal antibodies," writes Liping Sun, research oncologist at Genentech, California, and co-authors. (pharmaceutical-journal.com)
  • These are known as monoclonal antibodies (mAbs or Moabs). (cancer.org)
  • Monoclonal antibodies are used to treat many diseases, including some types of cancer. (cancer.org)
  • But other monoclonal antibodies act like immunotherapy because they help the immune system find and attack cancer cells more effectively. (cancer.org)
  • Monoclonal antibodies are man-made proteins that act like human antibodies in the immune system. (cancer.org)
  • We have produced several bispecific monoclonal antibodies (Bis-mAbs) that can effectively neutralize EBOV and SUDV, binding to a critical neutralization epitope at the viral glycoprotein (GP) base, as well as a suite of novel cross-reactive antibodies binding multiple filovirus species. (elsevierpure.com)
  • To this end, most often monoclonal antibodies are being used. (rug.nl)
  • Scientists at NIAID have developed recombinant monoclonal antibodies that are effective in vitro and in vivo at neutralizing SARS-CoV-2. (nih.gov)
  • These recombinant monoclonal antibodies can be used alone, in combination, or with other therapeutics for the treatment of SARS-COV-2. (nih.gov)
  • This Funding Opportunity Announcement (FOA) encourages applications to support the optimization of monoclonal antibodies (mAb) or mAb derivatives that recognize and eliminate cells comprising the HIV reservoir. (nih.gov)
  • Bispecific lentiviral anti-CD20/anti-CD19 (LV20.19) CAR T cells may prevent progression/relapse due to antigen escape. (nih.gov)
  • A bispecific monoclonal antibody (BsMAb, BsAb) is an artificial protein that can simultaneously bind to two different types of antigen or two different epitopes on the same antigen. (wikipedia.org)
  • Naturally occurring antibodies typically only target one antigen. (wikipedia.org)
  • The furthest developed of these newer formats are the bi-specific T-cell engagers (BiTEs), which uses the G4S linker to connect two ScFvs-one CD3 antibody ScFv and one tumor-associated antigen (TAA) or tumor-specific ScFv-to redirect T cells to cancer cells for target killing. (wikipedia.org)
  • The findings come from a first-in-human phase 1/2 study of REGN5459, a bispecific antibody targeting the B-cell maturation antigen (BCMA) on myeloma cells with a low-affinity binder to CD3 on T cells. (medscape.com)
  • Standard antibody drugs are designed to specifically target a single antigen. (amgen.com)
  • While natural antibodies have two targeting arms that bind to the same target antigen, bispecific antibodies are engineered hybrid molecules with two distinct binding domains that target two distinct antigens. (amgen.com)
  • For instance, in October 2022, the FDA approved Teclistamab-cqyv (Tecvayli, Janssen Biotech, Inc.), the first bispecific B-cell maturation antigen for adult patients with relapsed or refractory multiple myeloma. (grandviewresearch.com)
  • If you've had a BCMA bispecific, could you go on to a different target like cevostamab or talquetamab or other GPRC5D-targeting modalities, such as CAR [chimeric antigen receptor] T-cell therapy? (onclive.com)
  • Its broad portfolio of oncology product candidates includes individualized and off-the-shelf mRNA-based therapies, innovative chimeric antigen receptor (CAR) T cells, several protein-based therapeutics, including bispecific immune checkpoint modulators, targeted cancer antibodies and antibody-drug conjugate (ADC) therapeutics, as well as small molecules. (blogchicks.com.au)
  • A bispecific antibody is made by fusing fragments of two different antibodies, allowing it to simultaneously bind to two types of antigen. (pharmaceutical-journal.com)
  • An antibody is a protein that sticks to a specific protein called an antigen . (cancer.org)
  • Antibodies circulate throughout the body until they find and attach to the antigen. (cancer.org)
  • Researchers can design antibodies that specifically target a certain antigen, such as one found on cancer cells. (cancer.org)
  • To make a monoclonal antibody, researchers first have to identify the right antigen to attack. (cancer.org)
  • B-cell maturation antigen (BCMA)-directed T-cell immunotherapies, such as chimeric antigen receptor T-cells (CAR T-cells) and bispecific antibodies (BsAbs) have markedly improved the survival of triple-class refractory multiple myeloma (MM). However, the majority of patients still develops disease progression, underlining the need for new agents for these patients. (qxmd.com)
  • Trispecifc antibodies targeting two different MM-associated antigens to prevent antigen escape are in early clinical development, as well as trispecific antibodies (TsAbs) that provide an additional co-stimulatory signal to T-cells to prevent their exhaustion. (qxmd.com)
  • The inventors discovered that by administering a pharmaceutical composition comprising a bispecific antigen-binding molecule that recognizes (a) blood coagulation factor IX and/or activated blood coagulation factor IX and (b) blood coagulation factor X and/or activated blood coagulation factor X according to a given dosage regimen, bleeding, a disease accompanying bleeding, or a disease caused by bleeding can be prevented and/or treated more effectively. (justia.com)
  • 2 The first bispecific single-chain antibody was described in 1995 and was developed with the goal of directing CD3 T cells to tumor cells expressing the epithelial cell adhesion/activating molecule (EpCAM) cell surface antigen in colorectal cancer. (mhmedical.com)
  • Most serologic assays for flaviviruses measure antibodies binding to viral-envelope glycoprotein (E protein) because this antigen is a major target of human antibodies. (cdc.gov)
  • 13. A Novel T-Cell Engaging Bi-specific Antibody Targeting the Leukemia Antigen PR1/HLA-A2. (nih.gov)
  • 16. A bispecific diabody directed against prostate-specific membrane antigen and CD3 induces T-cell mediated lysis of prostate cancer cells. (nih.gov)
  • ORLANDO, Florida - For patients with heavily pretreated relapsed or refractory multiple myeloma, use of a novel bi-specific antibody designed to minimize the cytokine release syndrome (CRS) was associated with deep and durable clinical responses, and the safety profile was an acceptable. (medscape.com)
  • Last year, another bispecific antibody therapy targeting BCMA and CD3, teclistimab (Tecvayli), was approved by the US Food and Drug Administration to treat multiple myeloma after four or more lines of therapy, but CRS remains a common and potentially fatal side effect of immunotherapy. (medscape.com)
  • T-cell redirecting bispecific and trispecific antibodies in multiple myeloma beyond BCMA. (qxmd.com)
  • In October, the FDA approved the first bispecific antibody for multiple myeloma with a different target. (formularywatch.com)
  • Bispecific antibodies are showing promise in patients with hematologic malignancies, notably relapsed or refractory multiple myeloma. (valuebasedcancer.com)
  • A first-in-human study of talquetamab, a first-in-class bispecific antibody that binds to the orphan receptor GPRC5D and to CD3 on T-cells, showed a 66% ORR among patients with relapsed or refractory multiple myeloma who received the most active doses of this drug, said Ajai Chari, MD, Director of Clinical Research, Multiple Myeloma Program, Mount Sinai Medical Center, New York, NY. (valuebasedcancer.com)
  • Samer Al Hadidi, MD, University of Arkansas for Medical Sciences, Little Rock, AR, discusses the results of a recent analysis evaluating the risk of infections associated with the use of bispecific antibodies (BsAbs) in multiple myeloma. (vjhemonc.com)
  • Tecvayli is the first "off-the shelf" BCMA-targeted bispecific antibody approved for patients with heavily pretreated relapsed refractory multiple myeloma (RRMM). (themmrf.org)
  • Moreover, rising approval for several bispecific antibodies by regulatory bodies has surged the interest and confidence of companies to invest in research and development. (grandviewresearch.com)
  • There are several bispecific antibodies currently available through clinical trials for relapsed/refractory myeloma. (myeloma.org.uk)
  • Currently, there are several bispecific antibodies in clinical trials and many of them are using the inhibition of pathways as their mechanism and these antibodies seem very promising. (rug.nl)
  • This format retains the traditional monoclonal antibody (mAb) structure of two Fab arms and one Fc region, except the two Fab sites bind different antigens. (wikipedia.org)
  • The TandAb platform is formed by a tetravalent antibody molecule containing two binding sites for each of two antigens. (wikipedia.org)
  • As per the World Health Organization (WHO) report, cancer accounted for nearly 609,360 deaths in 2022 in the U.S. The significant burden of cancer drives the demand for innovative therapies, including bispecific antibodies, which can target multiple tumor-associated antigens simultaneously, enhancing therapeutic efficacy. (grandviewresearch.com)
  • Bispecific antibodies offer a promising approach for addressing this challenge by targeting multiple pathways or antigens involved in cancer progression, potentially leading to improved patient outcomes. (grandviewresearch.com)
  • Bispecific antibodies specifically target cancer cells or tumor-associated antigens while sparing healthy cells. (grandviewresearch.com)
  • The bsAb are a fast-growing group of therapeutic antibodies that is therapeutically promising due to their ability to bind to two different epitopes/antigens simultaneously. (a-star.edu.sg)
  • It was constructed using what is known as "knobs-into-holes" technology, which favours the heterodimer production essential for an antibody that recognises two different antigens at once. (pharmaceutical-journal.com)
  • They combine specificities of two antibodies and simultaneously bind to two different antigens or epitopes. (genekine.com)
  • In this approach, two heavy chains from existing antibodies to two different antigens are heterodimerized by KIH technology, and introduced into human naïve and synthesized light chain libraries. (genekine.com)
  • A leader in the bispecific antibody space, MacroGenics has extensive experience applying its proprietary DART platform to develop novel therapeutics. (gilead.com)
  • MacroGenics is a biopharmaceutical company focused on developing and commercializing innovative monoclonal antibody-based therapeutics for the treatment of cancer. (gilead.com)
  • Non-BCMA targeting bispecific antibodies are a relatively new class of therapeutics. (themmrf.org)
  • Celgene Inc. has formed a collaboration and licensing agreement with the biopharmaceutical company, Zymeworks, for the research, development, and commercialization of bi-specific antibody therapeutics enabled using Zymeworks' proprietary Azymetric platform. (dcatvci.org)
  • Antibody therapeutics have great importance for advanced cases of SARS-CoV-2 where a vaccine would not be effective and may be more effective than a vaccine in certain high-risk populations. (nih.gov)
  • In addition to their potential as therapeutics, these antibodies against SARS-CoV-2 can be used as prophylactics and in assay development. (nih.gov)
  • Bispecific Antibody: The Janus God Of Therapeutics? (synabs.be)
  • BOSTON, Jan. 19, 2021 /PRNewswire/ -- The leader in site specific bioconjugation and DAR (drug to antibody ratio) optimization in antibody drug conjugates (ADCs) and protein therapeutics, has created a novel class of bispecific antibodies that have the ability to be labeled and conjugated at two distinct sites. (brickbio.com)
  • The rate of innovation at BrickBio is on track to have a large impact in the field of antibody and protein therapeutics to positively affect patients' lives,' Boyce concluded. (brickbio.com)
  • Bispecific antibodies aim to treat multifaceted, complex diseases by engaging two disease targets with one molecule. (amgen.com)
  • Antibody CAP256-VRC26.25 targets the second hypervariable region (V2) at the apex of the HIV envelope (Env) trimer with extraordinary neutralization potency, although less than optimal breadth. (nih.gov)
  • Talquetamab is a 'first in class' bispecific antibody that targets the GPRC5D, a new target expressed on myeloma cells, and CD3, a protein expressed on T-cells. (healthtree.org)
  • Bi-specific antibodies developed using the Azymetric platform resemble conventional mono-specific antibodies while being able to simultaneously bind to two different targets, thereby resulting in additive or synergistic therapeutic responses. (dcatvci.org)
  • Talquetamab is a bispecific antibody that targets both CD3 on T cells and GPRC5D, which is overexpressed on myeloma cells. (formularywatch.com)
  • Furthermore, the specific antibody sequences and targets will inform vaccine development and establishment of long-term immunity. (nih.gov)
  • Despite their promise, CD3-based bispecific antibodies have demonstrated toxicity, particularly for targets with broad tissue expression, such as EGFR and EpCAM. (synabs.be)
  • This new class of bispecific antibody drug conjugate greatly expands the company's ability to optimize a number of key therapeutic properties while binding to multiple targets in order to increase efficacy,' Boyce continued. (brickbio.com)
  • Zymeworks' Azymetric platform forges antibodies able to bind to two different targets, spurring multiple effects while potentially reducing the chances of toxicity and drug resistance. (zist-fan.ir)
  • a new bispecific antibody that simultaneously targets CHI3L1 and CTLA4. (afternoonheadlines.com)
  • In vivo localization of a bispecific antibody which targets human T lymphocytes to lyse human colon cancer cells. (unil.ch)
  • Rising incidences of chronic diseases such as cancer, autoimmune disorders, and infectious diseases, rising demand for targeted therapies as well as advancements in antibody engineering technologies are some major driving factors that propel the market growth. (grandviewresearch.com)
  • This high target specificity minimizes off-target effects and reduces toxicity compared to traditional therapies, making bispecific antibodies an attractive option for cancer treatment. (grandviewresearch.com)
  • For more information on bispecific antibodies and other myeloma therapies, please visit our Treatment Options page https://themmrf.org/diagnosis-and-treatment/treatment-options/ . (themmrf.org)
  • In this innovative program integrating Virtual Reality animations, clinicians will visualize mechanisms of action for bispecific antibodies and recognize the rationale for application of these therapies in relapsed/refractory large B-cell lymphomas. (freecme.com)
  • By visualizing mechanisms, recognizing current clinical trial data informing the use of bispecific antibodies for large B-cell lymphomas, and understanding the potential for adverse events associated with bispecific antibodies and other therapies, clinicians will be better able to select appropriate therapy and monitor for potential adverse events in a highly multidisciplinary oncology practice setting. (freecme.com)
  • Focusing on recent clinical trial data, expert panelists take a broad look at how they might select and sequence bispecific antibody therapies in practice. (onclive.com)
  • CAR T-cell therapies are here to stay, but the question is whether bispecific antibodies will encroach on them," he commented. (ascopost.com)
  • Current filovirus antibody therapies are narrowly strain specific, and therefore have limited therapeutic utility. (elsevierpure.com)
  • These therapies will fill a major therapeutic gap and could not be obtained using conventional antibody isolation methods. (elsevierpure.com)
  • While duelling anti-BCMA Car-T therapies continued to do battle at Ash, yesterday Regeneron became the latest group to suggest that a bispecific MAb could be at least as good. (evaluate.com)
  • Bispecific antibodies (bsAbs) are emerging as the next generation of antibody-based therapies. (genekine.com)
  • Since the Fc region is still intact, this allows for the BsAb to trigger common immune responses when recognized by an Fc receptor, such as antibody-dependent cell-mediated cytotoxicity or complement-dependent cytotoxicity. (wikipedia.org)
  • In this article, explore the results from a collaboration with Alligator to develop an efficient downstream purification process for a cancer immunotherapy bispecific antibody (bsAb). (drugdiscoveryonline.com)
  • The bsAb by-products and impurities, especially mispaired homodimers, half antibodies and high levels of high molecular weight species, pose unique challenges to the downstream processing of bsAbs. (a-star.edu.sg)
  • A single-step downstream process using protein A chromatography to purify a bispecific antibody (BsAb) in early screening has been successfully developed to replace an existing three-step process. (processdevelopmentforum.com)
  • Bispecific antibodies recognize two different epitopes. (ascopost.com)
  • The Flavivirus E protein contains immunodominant antibody epitopes that are conserved (cross-reactive) between different flaviviruses or unique to each virus (type-specific) ( 10 - 12 ). (cdc.gov)
  • The original concept of BsAbs was proposed by Nisonoff and his collaborators in the 1960s, including the first idea of antibody architecture and other findings. (wikipedia.org)
  • Due to their ability to bind more than one target, bsAbs have demonstrated improved therapeutic efficacy and hold great promise in therapeutic applications that are difficult to achieve by single-targeting monospecific antibodies. (a-star.edu.sg)
  • Beyond the mechanistic advantages , bsAbs offer the possibility to combine in one single molecule the potential of two different antibodies without the related cost of two parallel development programs. (synabs.be)
  • Additionally, companies are actively engaging in collaborations and partnerships to leverage complementary expertise and technologies to develop bispecific antibodies. (grandviewresearch.com)
  • Dr. Chari began by discussing Talvey (talquetamab), a GPRC5D-targeting bispecific antibody approved for use in the US. (themmrf.org)
  • Talquetamab is a first-in-class, T-cell redirecting bispecific antibody targeting GPRC5D on myeloma cells and CD3 receptors on T cells. (themmrf.org)
  • Recombinant antibodies · 17. (synabs.be)
  • Advances in genetic engineering technology has resulted in a range of recombinant bispecific antibody formats. (genekine.com)
  • We describe a modified and improved recombinant envelope domain III-based serologic assay for detecting ZIKV type-specific antibodies in regions with endemic DENV transmission. (cdc.gov)
  • This essay presents an overview of the development, treatment efficacy, and potential pitfalls of using bispecific antibodies in cancer immunotherapy. (rug.nl)
  • This shows that bispecific antibodies are far from their optimal potential and efficacy. (rug.nl)
  • Overall, it was concluded that bispecific antibodies are very promising for the future, but a lot of work still needs to be done in their development but also in their efficacy. (rug.nl)
  • The accelerated approval of the monoclonal antibody (mAb) aducanumab as a treatment option for Alzheimer's Disease and the continued discussions about its efficacy have shown that a better understanding of immunotherapy for the treatment of neurodegenerative diseases is needed. (thno.org)
  • Updated results were presented on the MagnetisMM-1 trial, a phase 1 trial investigating the safety and efficacy of the BCMA-targeted bispecific antibody elranatamab as a single agent. (themmrf.org)
  • Catumaxomab, one of the first trifunctional antibodies approved for therapeutic use, binds both CD3 on cytotoxic T cells and EpCAM on human adenocarcinomas. (wikipedia.org)
  • FOSTER CITY, Calif. & ROCKVILLE, Md.--(BUSINESS WIRE)-- Gilead Sciences, Inc. (NASDAQ: GILD) and MacroGenics (NASDAQ: MGNX) today announced an exclusive option and collaboration agreement to develop MGD024, an investigational, bispecific antibody that binds CD123 and CD3 using MacroGenics' DART ® platform, and two additional bispecific research programs. (gilead.com)
  • This latest bispecific antibody binds to a target on the surface of a patient's T cells (CD3) and a target on their leukaemia or lymphoma cells (CD20). (pharmaceutical-journal.com)
  • Catumaxomab is a monoclonal antibody that binds to both the T-cell target recognised by CD20-TDB (CD3) and EpCAM, which is found in high levels on some types of cancer cell. (pharmaceutical-journal.com)
  • In this work, we explored a two-step "pretargeting" approach through engineering a bispecific antibody that binds both cell-surface ICAM-1 and polyethylene glycol (PEG) on the surface of nanoparticles, thereby effectively decoupling cell targeting from particle design and formulation. (nih.gov)
  • And using a bispecific agent earlier might help ameliorate T-cell exhaustion, one of the most significant challenges in immunotherapy, she added. (medscape.com)
  • BiTE antibodies represent the first immunotherapy with the ability to bind to and sustain T-cell engagement in vivo using an anti-CD3 T-cell single-chain antibody linked to a second tumor-specific antibody. (mhmedical.com)
  • Yana iya haÉ—awa da nau'ikan nau'ikan jiyya daban-daban waÉ—anda suka haÉ—a da chemotherapy, radiation, antibodies monoclonal, immunotherapy, hanyoyin da aka yi niyya, dashen Æ™wayoyin sel, magungunan CAR T-cell da Æ™ari. (lymphoma.org.au)
  • In the preclinical model, this molecule showed a lower rate of cytokine release, which is a problem with bispecific antibodies as a whole class of drugs. (medscape.com)
  • Advancing our bispecific DART molecule, MGD024, through a strategic collaboration with the team at Gilead will accelerate our ability to drive further development of MGD024 to the potential benefit of people living with blood cancers. (gilead.com)
  • But although automated high throughput screening approaches have become increasingly relevant and mature for pharmaceutical small molecule and classical antibody discovery, screening of bispecific antibodies is, however, up to now very limited by laborious preparation of the tremendous number of potential bispecific combinations. (tu-darmstadt.de)
  • Finally, the antibody sequences will be combined into one molecule that can both target and cleave amyloid beta. (brightfocus.org)
  • This so-called B cell-targeting anti-CD20/CD3 T cell-dependent bispecific antibody (CD20-TDB for short) is a full-length, humanised immunoglobulin G1 molecule with the near-native architecture of a typical antibody. (pharmaceutical-journal.com)
  • Azymetric antibodies spontaneously assemble into a single molecule with two different Fab domains comprising of unique heavy and light chain pairings. (dcatvci.org)
  • 11. Selective targeting and potent control of tumor growth using an EphA2/CD3-Bispecific single-chain antibody construct. (nih.gov)
  • For comparison, Glaxosmithkline has a BCMA-targeting antibody-drug conjugate, belantamab mafodotin, whose Dreamm-1 study yielded a 60% ORR . (evaluate.com)
  • In 2016, Zymeworks signed another bispecific antibody discovery deal with GlaxoSmithKline, worth up to $908 million in commercial bonuses, should everything pay off. (zist-fan.ir)
  • The Bi-Nanobody platform forms multi-specific binding through the connection between the VH regions of two or more antibody molecules. (wikipedia.org)
  • The key result is a process that generates fully functional bispecific molecules, a major step in advancing this highly promising drug modality. (amgen.com)
  • Marketwatch identified 37 bispecific antibody molecules currently in different stages of clinical development, of which eight or nine are expected to be launched by 2024. (pharmaceutical-journal.com)
  • Methods Anti-CTLA-4 nanobody lead molecules were generated from alpaca and anti-CCR8 antibodies were generated from hybridoma. (bmj.com)
  • However, heterodimeric IgG-like bispecific antibody, which is based on the heterodimerization of two different IgG molecules, is a promising format because it preserves the overall size and natural structure of human IgG with good stability, half-life, and pharmacokinetics profile. (genekine.com)
  • Zymeworks, based in Vancouver, Canada, is a privately held biotherapeutics company that is developing Azymetric bi-specific antibodies and antibody drug conjugates for the treatment of oncology, autoimmunity, and inflammatory diseases. (dcatvci.org)
  • In contrast, the [ 89 Zr]Zr-DFO-conjugates and the control antibody groups did not show any amyloid-beta related uptake pattern, indicating that DFO is inferior for 89 Zr-immuno-PET imaging of the brain in comparison to DFO* for Adu-8D3. (thno.org)
  • The developers of the new antibody, REGN5459, hypothesized that the low-affinity binder would activate T cells to deplete myeloma cells while minimizing CRS. (medscape.com)
  • Unlike CAR T-cells, which are engineered (over the course of 4-6 weeks) from a patient's own T-cells to better recognize and kill myeloma cells, bispecific antibodies are an "off-the-shelf" treatment that can bind to both myeloma cells and to T cells at the same time, bringing them in close proximity so that the T cells can recognize and kill the myeloma cells. (themmrf.org)
  • Bispecific antibodies (or T cell engagers) are another targeted approach to specifically kill myeloma cells. (myeloma.org.uk)
  • Recent study on the Bispecific Antibodies for Cancer Market comprises precise research on quantitative and qualitative aspects to examine the key drivers, opportunities, and threats in the overall development of the Bispecific Antibodies for Cancer Market on the global scale for the forecast period 2020 - 2025. (reportsinsights.com)
  • What is the estimated growth rate and Market share and size of the Bispecific Antibodies for Cancer Market for the forecast period 2020 - 2025? (reportsinsights.com)
  • What are the driving forces in the Bispecific Antibodies for Cancer Market for the forecast period 2020 - 2025? (reportsinsights.com)
  • Results from studies of 3 bispecific antibodies were presented at ASH 2020, showing deep and durable responses. (valuebasedcancer.com)
  • Azymetric antibodies are manufactured using conventional monoclonal antibody processes and can also be easily adapted to rapidly screen target and sequence combinations for bi-specific activities in the final therapeutic format, according to information from Zymeworks. (dcatvci.org)
  • CD14(+) accessory cells and the monocyte cell line THP-1 were activated via binding of the Fc region of Bi20, given that T cells were simultaneously engaged by the antibody. (nih.gov)
  • These antibodies are capable of binding in two separate ways and therefore are able to direct immune cells to the tumour, deliver payloads, and inhibit two different pathways simultaneously. (rug.nl)
  • bispecific antibodies that simultaneously target CHI3L1 and PD-1, and 3. (afternoonheadlines.com)
  • An efficient strategy to overcome these challenges is to use knobs-into-holes (KIH) technology to favor heterodimerization of two different heavy chains and combine with a common light chain, based on the widely accepted fact that the affinity and specificity of an antibody is mainly defined by the heavy chain, and it can be maintained when an antibody has a non-cognate light chain. (genekine.com)
  • 14. Specificity of bispecific T cell receptors and antibodies targeting peptide-HLA. (nih.gov)
  • Today, dozens of next-generation bispecific antibody constructs are in clinical trials, and they are poised to have a huge impact in the treatment of lymphoma and other hematologic malignancies, experts agreed. (ascopost.com)
  • Next, Dr. Trudel discussed cevostamab, a FcRH5-targeting bispecific antibody that is still in clinical trials. (themmrf.org)
  • Two bispecific antibodies have been FDA approved-blinatumomab and catumaxomab-but more than 30 are now in clinical trials, and two-thirds are anticancer agents. (ascopost.com)
  • Anti-CCR8 antibody monotherapy has shown significant anti-tumor activity in pre-clinical studies. (bmj.com)
  • A study now shows that stabilization of the iTCR-CD1d complex via a single-chain bi-specific antibody stimulates iNKT cell-mediated anti-tumor immunity. (nature.com)
  • Blinatumomab ( blincyto ) is a first-in-class bispecific antibody with high affinity toward both CD19 (expressed primarily on surface B cells) and CD3 (found on T lymphocytes) receptors. (mhmedical.com)
  • CD Targeted Bispecific Antibody Market Size, Share & Trends Analysis Report By Target Type (CD3, CD19 and CD30/CD16A), Region And Segment Forecasts, 2023-2031. (insightaceanalytic.com)
  • Based on the target type, the CD Targeted Bispecific Antibody Market is divided into CD3, CD19 and CD30/CD16A. (insightaceanalytic.com)
  • 2. A tetravalent bispecific TandAb (CD19/CD3), AFM11, efficiently recruits T cells for the potent lysis of CD19(+) tumor cells. (nih.gov)
  • The bispecific T-cell engager blinatumomab was the first such agent to be approved by the U.S. Food and Drug Administration (FDA) in patients with relapsed or refractory B-cell acute lymphocytic leukemia (ALL). (ascopost.com)
  • Previously, we constructed a bispecific NK-cell engager (BiKE) consisting of two scFV against CD16 (FcγRIII) on NK cells and EpCAM on tumor cells (EpCAM16). (aacrjournals.org)
  • First, without compromising cevostamab's effectiveness, pretreating patients with tocilizumab (an IL-6 antibody ) prior to cevostamab significantly reduced the incidence of CRS ( cytokine release syndrome), a flu-like reaction to the infusion that can cause chills, fever, and headaches. (themmrf.org)
  • Second, unlike the bispecific drug class, which typically treats patients continuously until the disease progresses, cevostamab patients received fixed-duration treatment. (themmrf.org)
  • Alligator Bioscience AB develops antibody-based pharmaceuticals for cancer treatment, specializing in mono- and bispecific antibodies for tumor-directed immunotherapies. (drugdiscoveryonline.com)
  • NOTE: Some m onoclonal antibodies used to treat cancer are sometimes considered to be a type of targeted therapy because they work by attaching to a specific target on a cancer cell and stopping it from functioning. (cancer.org)
  • However, it remains unclear to what extent these bispecific antibodies can be used to treat cancer. (rug.nl)
  • If this can be done the full potential of bispecific antibodies can be unleashed and this could possibly lead to curing some types of cancer. (rug.nl)
  • What are the Market trends influencing the progress of the Bispecific Antibodies for Cancer industry worldwide? (reportsinsights.com)
  • The report is distributed over 15 Chapters to display the analysis of the global Bispecific Antibodies for Cancer Market. (reportsinsights.com)
  • The CD Targeted Bispecific Antibody Market is driven by factors such as the rising prevalence of cancer, increasing research funding in the immune-oncology, and focus on research activities from government health institutions and pharma giants in Immuno-oncology therapy development. (insightaceanalytic.com)
  • The impressive ability of these bispecific antibodies to control primary and metastatic lung cancer in murine experimental modeling systems have been discussed in detail in articles in the Journal of Clinical Investigation , and Frontiers in Immunology . (afternoonheadlines.com)
  • The plethora of bispecific antibody architectures can be harnessed to elicit a broad variety of specific modes of actions, spanning from enhanced selectivity by simultaneous avid binding to distinct effector cell recruitment, all of which cannot be addressed by monospecific antibodies. (tu-darmstadt.de)
  • Despite their high potential value, discovery of bispecific antibodies as the identification of the best possible combination of two parental monospecific antibodies, however, remains challenging. (tu-darmstadt.de)
  • Discovery of two sets of monospecific antibodies followed by cloning, production and functional investigation of combinations is tedious and often resulting in undesired extended development times and increased expenses. (tu-darmstadt.de)
  • One challenge in making bispecific antibodies is getting cells to assemble the pieces of the antibody correctly. (amgen.com)
  • When cells are asked to incorporate subunits from two different antibodies into one structure, they generate 10 configurations, and just one has the correct and functional format (see insert). (amgen.com)
  • Bispecific antibodies belong to the drug class known as T-cell engagers, which also includes CAR T-cells. (themmrf.org)
  • Trifunctional bispecific antibodies can efficiently mediate tumor cell killing by redirecting T cells and immune accessory cells to the tumor cell. (nih.gov)
  • Here, we describe the new trifunctional antibody, Bi20 (FBTA05, anti-CD20 x anti-CD3), that connects B cells and T cells via its variable regions and recruits FcgammaRI(+) accessory immune cells via its Fc region. (nih.gov)
  • Mosunetuzumab is basically an antibody that uses the patient's own T cells to do what a CAR T cell would do. (ascopost.com)
  • We need more follow-up with these approaches to see tolerability and durability, but if bispecific antibodies can do what CAR T cells can do, that would be a big advance and would represent a paradigm shift. (ascopost.com)
  • Since its inception, Biotheus has established several innovative platforms for antibody discovery and a proprietary cell therapy that delivers T cell engagers into the tumor microenvironment to eradicate tumor cells. (blogchicks.com.au)
  • Researchers at Genentech have developed a bispecific antibody that redirects the immune system to attack leukaemia and lymphoma cells. (pharmaceutical-journal.com)
  • A bispecific antibody has been developed by researchers at US biotech company Genentech, who claim it is "highly active" against leukaemia and lymphoma cells. (pharmaceutical-journal.com)
  • Moreover, even though bispecific antibodies show great potential, the only approved ones are based on the mechanism of directing immune cells to the tumour, merely one of the many positive characteristics of bispecific antibodies. (rug.nl)
  • 12. Construction and characterization of a bispecific diabody for retargeting T cells to human carcinomas. (nih.gov)
  • Studies of second-generation bispecific antibodies were among the highlights of the 2019 American Society of Hematology (ASH) Annual Meeting & Exposition. (ascopost.com)
  • The global bispecific antibodies market size was valued at USD 5.73 billion in 2022 and is expected to grow at a compound annual growth rate (CAGR) of 44.0% from 2023 to 2030. (grandviewresearch.com)
  • Analysis of each segment of the global CD Targeted Bispecific Antibody Market expected to grow during the forecast period 2022-2031. (insightaceanalytic.com)
  • Crucially, these antibodies effectively neutralize the emerging B.1.1.7 and B.1.351 SARS-CoV-2 variants of concern. (nih.gov)
  • Dr Nizar Bahlis presented results from the MagnetisMM-3 trial ( ABSTRACT 159 ), a phase 2 study of elranatamab in patients with RRMM (exposed to at least one proteasome inhibitor, one immunomodulatory drug, and one anti-CD38 antibody). (themmrf.org)
  • InsightAce Analytic's "Global CD Targeted Bispecific Antibody Market Research Report 2023" is a comprehensive analysis, forecast, market dynamics and major trends report covering key market data on the CD Targeted Bispecific Antibody target type segments. (insightaceanalytic.com)
  • The goal of this proposal is to use an innovative antibody technology platform, bispecific antibody (Bis-mAb) engineering, to develop cross-neutralizing, therapeutic antibody cocktails effective against EBOV, SUDV, and BDBV. (elsevierpure.com)
  • The successful generation of this first-in-class therapeutic antibody has allowed the company to continue to the second phase of a therapeutic partnership with one of its large pharmaceutical partners. (brickbio.com)
  • In comparison, rituximab, a chimeric monoclonal CD20 antibody, demonstrated a significantly lower B-cell eradication rate. (nih.gov)
  • Anti-CD20/CD3 T cell-dependent bispecific antibody for the treatment of B cell malignancies. (pharmaceutical-journal.com)
  • One way the immune system protects the body from germs and other foreign substances is by making large numbers of antibodies . (cancer.org)
  • For some reason, they develop a heavy-chain antibody, and those antibodies develop quite a powerful immune system for them to protect them from any virus or pathogen disease, even for these precious species on the earth. (nih.gov)
  • Bispecific antibodies have been generating much interest since catumaxomab (Removab) was approved in Europe in 2009. (pharmaceutical-journal.com)
  • Based on whether they are mono-specific or bi-specific and where they bind to the SARS-CoV-2 virus, these antibodies can be subdivided into four groups that target (A) the receptor-binding-domain (RBD) of the SARS-COV-2 spike protein, (B) the N-terminal domain (NTD) of the SARS-COV-2 spike protein, (C) dual locations on the RBD, or (D) both the RBD and NTD. (nih.gov)
  • Antibody fragments are fused within the hinge region to a respective split intein part, capable to reconstitute two antibody fragments back to a full-length antibody format in vitro, without extensive cloning and manufacturing work. (tu-darmstadt.de)
  • The reconstitution method is furthermore amenable for automated high throughput screening providing the possibility to screen for bispecific combinations by combinatorial mixing of antibody fragments. (tu-darmstadt.de)
  • Fab fragments were combined with different Fc fragments as exemplary application for fast switch effector function screenings of monoclonal in addition to bispecific antibodies. (tu-darmstadt.de)
  • o One strategy to tackle the issue is the development of fragment-base formats, combining two antibody fragments Fab or ScFv without the Fc region . (synabs.be)
  • The antibody has a different mechanism of action to the monoclonal antibody rituximab, which is currently used to treat blood cancers, and preclinical testing suggests it could circumvent rituximab resistance. (pharmaceutical-journal.com)
  • Zymeworks unveiled new phase 1 clinical data from its lead bispecific antibody, ZW25, demonstrating an overall response rate of just 36%, the result of 12 partial responses, as a single-agent treatment in a variety of heavily pretreated, HER2-expressing cancers. (zist-fan.ir)
  • There are also fusion proteins mimicking the variable domains of two antibodies. (wikipedia.org)
  • The next step will be to identify antibody sequences that break down the amyloid beta, and to introduce mutations that increase the activity and targeting of the antibody so that it breaks down amyloid beta but not other proteins. (brightfocus.org)
  • Bispecific antibodies targeting distinct regions of the spike protein potently neutralize SARS-CoV-2 variants of concern. (nih.gov)
  • With ZW25 as its flagship, the biotech inked a bispecific antibody development deal with Janssen late last year, involving as many as six products and worth up to $1.1 billion, including sales royalties. (zist-fan.ir)
  • The Global CD Targeted Bispecific Antibody Market is segmented on the basis of target type and region. (insightaceanalytic.com)
  • Broadly neutralizing antibodies target the coronavirus fusion peptide. (nih.gov)
  • MacroGenics' bispecific expertise naturally complements Gilead's portfolio strengths in immuno-oncology and our growing hematology franchise," said Bill Grossman, MD, PhD, Senior Vice President, Oncology Clinical Development, Gilead Sciences. (gilead.com)
  • The method described could enable bispecific antibody high throughput binding and functional cellular screenings to greatly shorten development times and enhance the probability of identifying the optimal combination, ultimately leading to the generation of better biotherapeutics. (tu-darmstadt.de)
  • Learn from Jay Sinha at Incyte Corp as he presents development of an antibody expression platform using our CHO-K1 GS knockout cell line. (horizondiscovery.com)
  • Several cell line development systems were studied using a model bispecific antibody. (horizondiscovery.com)
  • But because of increased ease of development there recently has been more interest in bispecific antibodies. (rug.nl)
  • There have been quite some struggles with the development of bispecific antibodies over the last decades. (rug.nl)
  • Finally, we explore recent biotechnological advances such as the development of bispecific antibodies (ABT-122, COVA322), and their application to the treatment of IMIDs. (bmj.com)
  • Under the agreement, Zymeworks and Celgene will collaborate on the research and development of multiple bi-specific antibodies based on the Azymetric platform. (dcatvci.org)
  • In lay terms, can you describe the premise of your study that is looking at the development of neutralizing antibodies against SARS-CoV-2? (nih.gov)
  • Dr. Sierks' first step in producing such antibodies will be to identify cloned gene sequences of antibodies that will bind to certain regions of the amyloid beta protein. (brightfocus.org)
  • We previously described a serologic assay using domain III of the ZIKV E protein (EDIII) to detect ZIKV type-specific antibodies among persons in areas with DENV and ZIKV cocirculation ( 19 ). (cdc.gov)
  • 6. TCR-mimic bispecific antibodies targeting LMP2A show potent activity against EBV malignancies. (nih.gov)
  • The most common types are called trifunctional antibodies, as they have three unique binding sites on the antibody: the two Fab regions, and the Fc region. (wikipedia.org)