US20040072262A1 - Methods and systems for detecting MHC class I binding peptides - Google Patents

Methods and systems for detecting MHC class I binding peptides Download PDF

Info

Publication number
US20040072262A1
US20040072262A1 US10/269,473 US26947302A US2004072262A1 US 20040072262 A1 US20040072262 A1 US 20040072262A1 US 26947302 A US26947302 A US 26947302A US 2004072262 A1 US2004072262 A1 US 2004072262A1
Authority
US
United States
Prior art keywords
mhc
binding
monomer
monomers
hla
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/269,473
Other languages
English (en)
Inventor
Felix Montero-Julian
Sylvain Monseaux
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Beckman Coulter Inc
Original Assignee
Beckman Coulter Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Beckman Coulter Inc filed Critical Beckman Coulter Inc
Priority to US10/269,473 priority Critical patent/US20040072262A1/en
Assigned to BECKMAN COULTER INC. reassignment BECKMAN COULTER INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MONSEAUX, SYLVAIN, MONTERO-JULIAN, FELIX A.
Priority to JP2004543745A priority patent/JP4607590B2/ja
Priority to CA2501864A priority patent/CA2501864C/en
Priority to PCT/US2003/032370 priority patent/WO2004034033A2/en
Priority to AU2003284113A priority patent/AU2003284113B2/en
Priority to EP03776344A priority patent/EP1549952B1/en
Priority to CN2003801010208A priority patent/CN1703624B/zh
Priority to US10/684,268 priority patent/US20040137537A1/en
Publication of US20040072262A1 publication Critical patent/US20040072262A1/en
Priority to US13/029,902 priority patent/US8815528B2/en
Priority to US14/336,944 priority patent/US20150072886A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J19/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J19/0046Sequential or parallel reactions, e.g. for the synthesis of polypeptides or polynucleotides; Apparatus and devices for combinatorial chemistry or for making molecular arrays
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54306Solid-phase reaction mechanisms
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56966Animal cells
    • G01N33/56977HLA or MHC typing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00279Features relating to reactor vessels
    • B01J2219/00306Reactor vessels in a multiple arrangement
    • B01J2219/00313Reactor vessels in a multiple arrangement the reactor vessels being formed by arrays of wells in blocks
    • B01J2219/00315Microtiter plates
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00452Means for the recovery of reactants or products
    • B01J2219/00454Means for the recovery of reactants or products by chemical cleavage from the solid support
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00277Apparatus
    • B01J2219/00497Features relating to the solid phase supports
    • B01J2219/005Beads
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/0061The surface being organic
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00614Delimitation of the attachment areas
    • B01J2219/00621Delimitation of the attachment areas by physical means, e.g. trenches, raised areas
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00623Immobilisation or binding
    • B01J2219/0063Other, e.g. van der Waals forces, hydrogen bonding
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00583Features relative to the processes being carried out
    • B01J2219/00603Making arrays on substantially continuous surfaces
    • B01J2219/00605Making arrays on substantially continuous surfaces the compounds being directly bound or immobilised to solid supports
    • B01J2219/00632Introduction of reactive groups to the surface
    • B01J2219/00637Introduction of reactive groups to the surface by coating it with another layer
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00718Type of compounds synthesised
    • B01J2219/0072Organic compounds
    • B01J2219/00725Peptides
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00718Type of compounds synthesised
    • B01J2219/0072Organic compounds
    • B01J2219/0074Biological products
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/70539MHC-molecules, e.g. HLA-molecules
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2458/00Labels used in chemical analysis of biological material

Definitions

  • This invention relates generally to the field of immunoassays, especially using immunoassays to detect and measure binding of peptides to MHC alleles.
  • the Class I histocompatibility ternary complex consists of three parts associated by noncovalent bonds.
  • a transmembrane protein, called the MHC heavy chain is mostly exposed at the cell surface.
  • the cell surface domains of the MHC heavy chain contain two segments of alpha helix that form two ridges with a groove between them.
  • a short peptide binds noncovalently (“fits”) into this groove between the two alpha helices, and a molecule of beta-2 microglobulin binds noncovalently along side the outer two domains of the MHC monomer, forming a ternary complex.
  • Peptides that bind noncovalently to one MHC subtype heavy chain usually will not bind to another subtype. However, all bind with the same type of beta-2 microglobulin.
  • MHC molecules are synthesized and displayed by most of the cells of the body.
  • HLA molecules are referred to as HLA molecules. Humans primarily synthesize three different sub-types of MHC class I molecules designated HLA-A, HLA-B and HLA-C, differing only in the heavy chains.
  • the MHC works coordinately with a specialized type of T cell (the cytotoxic T cell) to rid the body of “nonself” or foreign viral proteins.
  • the antigen receptor on T-cells recognizes an epitope that is a mosaic of the bound peptide and portions of the alpha helices of the making up the groove flanking it.
  • the presentation of peptide fragments by the MHC molecule allows for MHC-restricted cytotoxic T cells to survey cells for the expression of “nonself” or foreign viral proteins.
  • a functional T-cell will exhibit a cytotoxic immune response upon recognition of an MHC molecule containing bound antigenic peptide for which the T-cell is specific.
  • HLA-A, B, and C heavy chains interact with a multitude of peptides of about 8 to about 10, possibly about 8 to about 11, or about 8 to about 12 amino acids in length. Only certain peptides bind into the binding pocket in the heavy chain of each HLA sub-type as the monomer folds, although certain subtypes cross-react.
  • complete coding region sequences had been determined for each of 43 HLA-A, 89 HLA-B and 11 HLA-C alleles (P. Parham et al., Imunology Review 143:141-180, 1995).
  • Class II histocompatibility molecules consist of two transmembrane polypeptides that interact to form a groove at their outer end which, like the groove in class I molecules, non-covalently associates with an antigenic peptide.
  • the antigenic peptides bound to class II molecules are derived from antigens that the cell has taken in from its surroundings.
  • peptides that bind to class II histocompatibility molecules are 15 to about 25 or to about 30 amino acids in length. Only cells, such as macrophages, dendritic cells and B lymphocytes, that specialize in taking up antigen from extracellular fluids, express class II molecules.
  • Another approach to identifying MHC-binding peptides uses a competition-based binding assay. All competition assays yield a comparison of binding affinities of different peptides. However, such assays do not yield an absolute dissociation constant since the result is dependent on the reference peptide used.
  • Still another approach used for determining MHC-binding peptides is the classical reconstitution assay, e.g. using “T2” cells, in which cells expressing an appropriate MHC allele are “stripped” of a native binding peptide by incubating at pH 2-3 for a short period of time. Then, to determine the binding affinity of a putative MHC-binding peptide for the same MHC allele, the stripped MHC monomer is combined in solution with the putative MHC-binding peptide, beta2-microglobulin and a conformation-dependent monoclonal antibody.
  • the difference in fluorescence intensity determined between cells incubated with and without the test binding peptide after labeling can be used to determine binding of the test peptide.
  • soluble MHC monomers stripped at low pH aggregate immediately, making their use in high through-put assays difficult and impractical.
  • in vitro assays for cell mediated immunity which use cells from the donor.
  • the assays include situations where the cells are from the donor, however, many assays provide a source of antigen presenting cells from other sources, e.g., B cell lines.
  • These in vitro assays include the cytotoxic T lymphocyte assay; lymphoproliferative assays, e.g., tritiated thymidine incorporation; the protein kinase assays, the ion transport assay and the lymphocyte migration inhibition function assay (Hickling, J. K. et al, J. Virol., 61: 3463 (1987); Hengel, H. et al, J.
  • MHC tetramers which are complexes of four MHC monomers with streptavidin, a molecule having tetrameric binding sites for biotin, to which is bound a fluorochrome, e.g., phycoerythrin.
  • a fluorochrome e.g., phycoerythrin.
  • soluble subunits of ⁇ 2-microglobulin, the peptide fragment containing a putative T-cell epitope, and of a MHC heavy chain corresponding to the predicted MHC subtype of the peptide fragment of interest are obtained by expression of the polypeptides in host cells.
  • Each of the four monomers contained in the MHC tetramer is produced as a monomer by refolding these soluble subunits under conditions that favor assembly of the soluble units into reconstituted monomers, each containing a beta2-microglobulin, a peptide fragment, and the corresponding MHC heavy chain.
  • An MHC tetramer is constructed from the monomers by biotinylation of the monomers and subsequent contact of the biotinylated reconstituted monomers with fluorochrome-labeled streptavidin.
  • T cells When contacted with a diverse population of T cells, such as is contained in a sample of the peripheral blood lymphocytes (PBLs) of a subject, those tetramers containing reconstituted monomers that are recognized by a T cell in the sample will bind to the matched T cell. Contents of the reaction is analyzed using fluorescence flow cytometry, to determine, quantify and/or isolate those T-cells having a MHC tetramer bound thereto (See U.S. Pat. No. 5,635,363).
  • PBLs peripheral blood lymphocytes
  • At least one other test is required to determine whether a test peptide recognized by a T-cell by the MHC tetramer assay will activate the T-cell to generate an immune response, a so-called “functional test”.
  • the enzyme-linked immunospot (ELISpot) assay has been adapted for the detection of individual cells secreting specific cytokines or other effector molecules by attachment of a monoclonal antibody specific for a cytokine or effector molecule on a microplate. Cells stimulated by an antigen are contacted with the immobilized antibody.
  • a tagged polyclonalantibody or more often, a monoclonal antibody, specific for the same cytokine or other effector molecule is added to the wells.
  • a colorant that binds to the tagged antibody is added such that a blue-black colored precipitate (or spot) forms at the sites of cytokine localization.
  • the spots can be counted manually or with automated ELISpot reader system to quantitated the response.
  • a final confirmation of T-cell activation by the test peptide may require in vivo testing, for example in a mouse model.
  • the route to final confirmation of the efficacy of a MHC-binding peptide is expensive and time consuming.
  • the present invention is based on the discovery that MHC class I monomers when immobilized to a solid surface are still capable of reconstituting to incorporate from solution an MHC-binding peptide and form a ternary complex.
  • the invention provides a system comprising a solid surface, wherein the surface has attached thereto one or more MHC monomer or modified MHC monomer, wherein the monomer denatures in a denaturing condition and reconstitutes to form a ternary complex containing a suitable MHC-binding peptide in the binding pocket under reconstituting conditions.
  • a kit comprising the invention system is also provided.
  • the invention provides methods for determining binding between a MHC monomer or modified MHC monomer and a putative MHC-binding peptide therefor.
  • a solid surface having attached thereto a plurality of previously denatured MHC monomers or modified MHC monomers is incubated under reconstituting conditions in the presence and absence of the putative MHC-binding peptide such that the monomers reconstitute to form a ternary complex containing a suitable MHC-binding peptide under the reconstituting conditions. Binding to the ternary complex of a monoclonal antibody that does not bind to dissociated components of the complex indicates binding between the putative MHC-binding peptide and the monomers.
  • the invention provides methods for determining the degree of binding affinity of an MHC monomer or modified MHC monomer for a putative MHC-binding peptide therefor.
  • at least one denatured MHC monomer or modified MHC monomer attached to a solid surface is incubated under reconstituting conditions with the putative MHC-binding peptide and a monoclonal antibody that specifically binds to a conformational epitope of a corresponding reconstituted MHC monomer that is not present in the denatured monomer.
  • Binding of the monoclonal antibody to a monomer that binds to the putative MHC-binding peptide is compared with binding of the monoclonal antibody to a corresponding monomer having a known MHC-binding peptide bound thereto. The difference in the binding indicates the relative degree of binding affinity of the reconstituted monomer for the putative MHC-binding peptide.
  • the invention provides methods for determining the stability at 37° C. of an MHC monomer or modified MHC monomer for a putative MHC-binding peptide therefor.
  • at least one denatured MHC monomer or modified MHC monomer attached to a solid surface is incubated under reconstituting conditions with the putative MHC-binding peptide and a monoclonal antibody that specifically binds to a conformational epitope of a corresponding reconstituted MHC monomer that is not present in the denatured monomer.
  • the reconstituted ternary complex with the monoclonal antibody is incubated at different temperatures and different times. The difference in the signal obtained at different temperatures and different times, indicates the relative stability of the reconstituted monomer for the putative MHC-binding peptide.
  • FIG. 1 is a schematic representation of the immunoassay.
  • FIG. 2 is a graph showing calibration of the anti-HLA-class I-FITC mAb for fluorometric assay.
  • FIG. 3 is a graph showing a decrease in binding of anti-HLA-class I-FITC mAb to a reconstituted HLA heavy chain monomer Mart1 26-35 with increasing temperature as determined by fluorescence of bound antibody.
  • FIG. 4 is a graph showing the binding of an anti-HLA-class I-FITC monoclonal antibody to human and mouse alleles as determined by fluorescence of bound antibody.
  • FIG. 5 is a graph showing renaturation in various buffer solutions of the MHC heavy chain monomers attached to a plate as detected by an anti-HLA-class I-FITC mAb.
  • FIG. 6 is a graph showing antibody binding to monomer at concentrations of anti-HLA-class I mAb of 1 to 2 ⁇ g/ml for various HLA heavy chain monomer concentrations to determine the optimal concentration of the anti-HLA-class I antibody for use with a microtiter plate assay.
  • FIGS. 7A and 7B are graphs showing the dose response curve obtained with two different HLA heavy chain monomers
  • FIG. 8 is a graph showing the specificity of the anti-HLA-class I antibody for various HLA-A and HLA-B alleles.
  • FIG. 9 is a schematic drawing showing formation of a human-mouse chimeric MHC modified monomer according to the invention.
  • FIGS. 10 A-D show graphs of the dissociation curves for renatured peptides (HBV core peptide; 26-35L; 26-35; 27-35, respectively).
  • FIGS. 10 E-H show graphs of the off rates for peptides HBV core; 26-35L; 26-35; and 27-35, respectively.
  • FIG. 10I shows the effect of temperature on monomer dissociation.
  • the present invention relates in general to immunoassays directed to detection and measurement of the binding affinity of MHC heavy chain monomers, especially MHC heavy chain monomers immobilized on a surface, for putative MHC-binding peptides. It is the discovery of the present invention that MHC heavy chain monomers and modified MHC monomers immobilized to a solid surface are still capable of refolding so as to bind from solution beta2-microglobulin and a MHC-binding peptide that has the requisite binding.
  • binding can be detected in an immunoassay format, such as one utilizing a conformation-dependent monoclonal antibody that specifically binds to a ternary complex containing such refolded or reconstituted MHC monomers but does not bind to dissociated components of the ternary complex,.
  • MHC monomer and HLA monomer refer to a class I MHC heavy chain that maintains the ability to assemble into a ternary complex with an appropriate MHC-binding or HLA-binding peptide and beta-2 microglobulin under renaturing conditions.
  • MHC monomer and HLA monomer are also used to refer to the denatured form of the monomer that results from subjecting the ternary complex to denaturing conditions, causing the monomer to unfold and dissociate from a MHC-binding peptide and from beta-2 microglobulin.
  • modified MHC monomer and “modified HLA monomer” refer to class I monomers as described above, but which have been engineered to introduce modifications as described below. These terms also encompass functional fragments of the MHC monomer that maintain the ability to assemble into a ternary complex with an appropriate MHC-binding or HLA-binding peptide and beta-2 microglobulin under renaturing conditions and to dissociate under denaturing conditions.
  • a functional fragment can comprise only the ⁇ 1 , ⁇ 2 , ⁇ 3 , domains, or only ⁇ 1 , ⁇ 2 domains, of the class I heavy chain, i.e., the cell surface domains, that participate in formation of the ternary complex.
  • modified MHC monomers can be class I heavy chain molecules, or functional fragments thereof, contained in a fusion protein or “single chain” molecule and may further include an amino acid sequence functioning as a linker between cell surface domains of the monomer, a detectable marker or as a ligand to attach the molecule to a solid support that is coated with a second ligand with which the ligand in the fusion protein reacts.
  • modified MHC monomer and “modified HLA monomer” are intended to encompass chimera containing domains of class I heavy chain molecules from more than one species or from more than one class I subclass.
  • the Class I MHC in humans is located on chromosome 6 and has three loci, HLA-, HLA-B, and HLA-C.
  • the first two loci have a large number of alleles encoding alloantigens. These are found to consist of a 44 Kd heavy chain subunit and a 12 Kd .beta 2 -microglobulin subunit which is common to all antigenic specificities.
  • soluble HLA-A2 can be purified after papain digestion of plasma membranes from the homozygous human lymphoblastoid cell line J-Y as described by Turner, M. J. et al., J. Biol. Chem. (1977) 252:7555-7567. Papain cleaves the 44 Kd heavy chain close to the transmembrane region, yielding a molecule comprised of ⁇ 1 , ⁇ 2 , ⁇ 3 domains and beta-2 microglobulin.
  • the MHC monomers can be isolated from appropriate cells or can be recombinantly produced, for example as described by Paul et al, Fundamental Immunology, 2d Ed., W. E. Paul, ed., Ravens Press N.Y. 1989, Chapters 16-18) and readily modified, as described below.
  • isolated refers to an MHC glycoprotein heavy chain of MHC class I, which is in other than its native state, for example, not associated with the cell membrane of a cell that normally expresses MHC.
  • This term embraces a full length subunit chain, as well as a functional fragment of the MHC monomer.
  • a functional fragment is one comprising an antigen binding site and sequences necessary for recognition by the appropriate T cell receptor. It typically comprises at least about 60-80%, typically 90-95% of the sequence of the full-length chain.
  • the “isolated” MHC subunit component may be recombinantly produced or solubilized from the appropriate cell source.
  • MHC glycoprotein monomers will vary somewhat in length because of deletions, substitutions, and insertions or additions of one or more amino acids in the sequences. Thus, MHC monomers are subject to substantial natural modification, yet are still capable of retaining their functions.
  • Modified protein chains can also be readily designed and manufactured utilizing various recombinant DNA techniques well known to those skilled in the art and described in detail, below. For example, the chains can vary from the naturally occurring sequence at the primary structure level by amino acid substitutions, additions, deletions, and the like. These modifications can be used in a number of combinations to produce the final modified protein chain.
  • modifications of the genes encoding the MHC monomer may be readily accomplished by a variety of well-known techniques, such as site-directed mutagenesis.
  • the effect of any particular modification can be evaluated by routine screening in a suitable assay for the desired characteristic. For instance, a change in the immunological character of the subunit can be detected by competitive immunoassay with an appropriate antibody.
  • the effect of a modification on the ability of the monomer to activate T cells can be tested using standard in vitro cellular assays or the methods described in the example section, below. Modifications of other properties such as redox or thermal stability, hydrophobicity, susceptibility to proteolysis, or the tendency to aggregate are all assayed according to standard techniques.
  • This invention provides amino acid sequence modification of MHC monomers prepared with various objectives in mind, including increasing the affinity of the subunit for antigenic peptides and/or T cell receptors, facilitating the stability, purification and preparation of the subunits.
  • the monomers may also be modified to modify plasma half life, improve therapeutic efficacy, or to lessen the severity or occurrence of side effects during therapeutic use of complexes of the present invention.
  • the amino acid sequence modifications of the subunits are usually predetermined variants not found in nature or naturally occurring alleles. The variants typically exhibit the same biological activity (for example, MHC-peptide binding) as the naturally occurring analogue.
  • Insertional modifications of the present invention are those in which one or more amino acid residues are introduced into a predetermined site in the MHC monomer and which displace the preexisting residues.
  • insertional modifications can be fusions of heterologous proteins or polypeptides to the amino or carboxyl terminus of the subunits.
  • modifications include fusions of the monomer with a heterologous signal sequence and fusions of the monomer to polypeptides having enhanced plasma half life (ordinarily>about 20 hours) such as immunoglobulin chains or fragments thereof as is known in the art.
  • Substitutional modifications are those in which at least one residue has been removed and a different residue inserted in its place.
  • Nonnatural amino acid i.e., amino acids not normally found in native proteins
  • isosteric analogs amino acid or otherwise
  • Substantial changes in function or immunological identity are made by selecting substituting residues that differ in their effect on maintaining the structure of the polypeqptide backbone (e.g., as a sheet or helical conformation), the charge or hydrophobicity of the molecule at the target site, or the bulk of the side chain.
  • substitutions which in general are expected to produce the greatest changes in function will be those in which (a) a hydrophilic residue, e.g., serine or threonine, is substituted for (or by) a hydrophobic residue, e.g.
  • electropositive side chain e.g., lysine, arginine, or histidine
  • an electronegative residue e.g., glutamine or aspartine
  • a residue having a bulky side chain e.g., phenylalanine
  • Substitutional modifications of the monomers also include those where functionally homologous (having at least about 70% homology) domains of other proteins are substituted by routine methods for one or more of the MHC subunit domains.
  • Particularly preferred proteins for this purpose are domains from other species, such as murine species as illustrated in FIG. 9 herein.
  • deletional modifications are characterized by the removal of one or more amino acid residues from the MHC monomer sequence. Typically, the transmembrane and cytoplasmic domains are deleted. Deletions of cysteine or other labile residues also may be desirable, for example in increasing the oxidative stability of the MHC complex. Deletion or substitutions of potential proteolysis sites, e.g., ArgArg, is accomplished by deleting one of the basic residues or substituting one by glutaminyl or histidyl residues.
  • a preferred class of substitutional or deletional modifications comprises those involving the transmembrane region of the subunit.
  • Transmembrane regions of MHC monomers are highly hydrophobic or lipophilic domains that are the proper size to span the lipid bilayer of the cellular membrane. They are believed to anchor the MHC molecule in the cell membrane.
  • Inactivation of the transmembrane domain typically by deletion or substitution of transmembrane domain hydroxylation residues, will facilitate recovery and formulation by reducing its cellular or membrane lipid affinity and improving its aqueous solubility.
  • the transmembrane and cytoplasmic domains can be deleted to avoid the introduction of potentially immunogenic epitopes. Inactivation of the membrane binding function is accomplished by deletion of sufficient residues to produce a substantially hydrophilic hydropathy profile at this site or by substitution with heterologous residues which accomplish the same result.
  • transmembrane-inactivated MHC monomer A principal advantage of the transmembrane-inactivated MHC monomer is that it may be secreted into the culture medium of recombinant hosts. This variant is soluble in body fluids such as blood and does not have an appreciable affinity for cell membrane lipids, thus considerably simplifying its recovery from recombinant cell culture.
  • modified MHC monomers of this invention will not have a functional transmembrane domain and preferably will not have a functional cytoplasmic sequence.
  • modified MHC monomers will consist essentially of the effective portion of the extracellular domain of the MHC monomer. In some circumstances, the monomer comprises sequences from the transmembrane region (up to about 10 amino acids), so long as solubility is not significantly affected.
  • the transmembrane domain may be substituted by any amino acid sequence, e.g., a random or predetermined sequence of about 5 to 50 serine, threonine, lysine, arginine, glutamine, aspartic acid and like hydrophilic residues, which altogether exhibit a hydrophilic hydropathy profile.
  • a random or predetermined sequence of about 5 to 50 serine, threonine, lysine, arginine, glutamine, aspartic acid and like hydrophilic residues, which altogether exhibit a hydrophilic hydropathy profile.
  • these monomers are secreted into the culture medium of recombinant hosts.
  • Glycosylation variants are included within the scope of this invention. They include variants completely lacking in glycosylation (unglycosylated) and variants having at least one less glycosylated site than the native form (deglycosylated) as well as variants in which the glycosylation has been changed. Included are deglycosylated and unglycosylated amino acid sequence variants, deglycosylated and unglycosylated subunits having the native, unmodified amino acid sequence.
  • substitutional or deletional mutagenesis is employed to eliminate the N- or O-linked glycosylation sites of the subunit, e.g., the asparagine residue is deleted or substituted for by another basic residue such as lysine or histidine.
  • flanking residues making up the glycosylation site are substituted or deleted, even though the asparagine residues remain unchanged, in order to prevent glycosylation by eliminating the glycosylation recognition site.
  • unglycosylated MHC monomers which have the amino acid sequence of the native monomers are produced in recombinant prokaryotic cell culture because prokaryotes are incapable of introducing glycosylation into polypeptides.
  • Glycosylation variants are conveniently produced by selecting appropriate host cells or by in vitro methods.
  • Yeast for example, introduce glycosylation which varies significantly from that of mammalian systems.
  • mammalian cells having a different species e.g., hamster, murine, insect, porcine, bovine or ovine
  • tissue origin e.g., lung, liver, lymphoid, mesenchymal or epidermal
  • In vitro processing of the subunit typically is accomplished by enzymatic hydrolysis, e.g., neuraminidase digestion.
  • MHC glycoproteins suitable for use in the present invention have been isolated from a multiplicity of cells using a variety of techniques including solubilization by treatment with papain, by treatment with 3M KCl, and by treatment with detergent. For example, detergent extraction of Class I protein followed by affinity purification can be used. Detergent can then be removed by dialysis or selective binding beads. The molecules can be obtained by isolation from any MHC I bearing cell, for example from an individual suffering from a targeted cancer or viral disease.
  • Isolation of individual heavy chain from the isolated MHC glycoproteins is easily achieved using standard techniques known to those skilled in the art.
  • the heavy chain can be separated using SDS/PAGE and electroelution of the heavy chain from the gel (see, e.g., Dornimair et al., supra and Hunkapiller, et al., Methods in Enzymol. 91:227-236 (1983).
  • Separate subunits from MHC I molecules are also isolated using SDS/PAGE followed by electroelution as described in Gorga et al. J. Biol. Chem. 262:16087-16094 (1987) and Dommair et al. Cold Spring Harbor Symp. Quant. Biol. 54:409-416 (1989)
  • ion exchange chromatography size exclusion chromatography or affinity chromatography.
  • the amino acid sequences of a number of Class I proteins are known, and the genes have been cloned, therefore, the heavy chain monomers can be expressed using recombinant methods.
  • recombinant techniques provide methods for carboxy terminal truncation which deletes the hydrophobic transmembrane domain.
  • the carboxy termini can also be arbitrarily chosen to facilitate the conjugation of ligands or labels, for example, by introducing cysteine and/or lysine residues into the molecule.
  • the synthetic gene will typically include restriction sites to aid insertion into expression vectors and manipulation of the gene sequence.
  • the genes encoding the appropriate monomers are then inserted into expression vectors, expressed in an appropriate host, such as E. coli , yeast, insect, or other suitable cells, and the recombinant proteins are obtained.
  • a second generation of construction includes chimeric constructs, as illustrated in FIG. 9.
  • the ⁇ 1 , ⁇ 2 , ⁇ 3 , domains of the class I heavy chain are linked typically by the ⁇ 3 domain of Class I with beta-2 microglobulin and coexpressed to stabilize the complex.
  • the transmembrane and intracellular domains of the Class I gene can optionally also be included.
  • Expression can be in procaryotic or eucaryotic systems.
  • Suitable eucaryotic systems include yeast, plant and insect systems, such as the Drosophila expression vectors under an inducible promoter.
  • Procaryotes most frequently are represented by various strains of E. coli .
  • other microbial strains may also be used, such as bacilli, for example Bacillus subtilis , various species of Pseudomonas, or other bacterial strains.
  • plasmid vectors which contain replication sites and control sequences derived from a species compatible with the host are used.
  • E. coli is typically transformed using derivatives of pBR322, a plasmid derived from an E.
  • procaryotic control sequences which are defined herein to include promoters for transcription initiation, optionally with an operator, along with ribosome binding site sequences, including such commonly used promoters as the ⁇ -lactamase (penicillinase) and lactose (lac) promoter systems (Change et al., Nature (1977) 198:1056) and the tryptophan (trp) promoter system (Goeddel et al., Nucleic Acids Res.
  • promoters for transcription initiation optionally with an operator
  • ribosome binding site sequences including such commonly used promoters as the ⁇ -lactamase (penicillinase) and lactose (lac) promoter systems (Change et al., Nature (1977) 198:1056) and the tryptophan (trp) promoter system (Goeddel et al., Nucleic Acids Res.
  • the expression systems useful in the eucaryotic hosts comprise promoters derived from appropriate eucaryotic genes.
  • a class of promoters useful in yeast include promoters for synthesis of glycolytic enzymes, including those for 3-phosphoglycerate kinase (Hitzeman, et al., J. Biol. Chem. (1980) 255:2073).
  • Other promoters include, for example, those from the enolase gene (Holland, M. J., et al. J. Biol. Chem. (1981) 256:1385) or the Leu2 gene obtained from YEp13 (Broach, J., et al., Gene (1978) 8:121).
  • a Drosophila expression system under an inducible promoter can also be used.
  • Suitable mammalian promoters include the early and late promoters from SV40 (Fiers, et al., Nature (1978) 273:113) or other viral promoters such as those derived from polyoma, adenovirus II, bovine papilloma virus or avian sarcoma viruses. Suitable viral and mammalian enhancers are cited above.
  • the expression system is constructed from the foregoing control elements operably linked to the MHC sequences using standard methods, employing standard ligation and restriction techniques which are well understood in the art. Isolated plasmids, DNA sequences, or synthesized oligonucleotides are cleaved, tailored, and religated in the form desired.
  • Site-specific DNA cleavage is performed by treatment with the suitable restriction enzyme (or enzymes) under conditions which are generally understood in the art, and the particulars of which are specified by the manufacturer of these commercially available restriction enzymes.
  • suitable restriction enzyme or enzymes
  • about 1 ⁇ g of plasmid or DNA sequence is cleaved by one unit of enzyme in about 20 ⁇ l of buffer solution; an excess of restriction enzyme may be used to insure complete digestion of the DNA substrate.
  • protein is removed by extraction with phenol/chloroform, and may be followed by ether extraction, and the nucleic acid recovered from aqueous fractions by precipitation with ethanol followed by running over a Sephadex G-50 spin column. If desired, size separation of the cleaved fragments may be performed.
  • Restriction cleaved fragments may be blunt ended by treating with the large fragment of E. coli DNA polymerase I (Klenow) in the presence of the four deoxynucleotide triphosphates (dNTPs) After treatment with Klenow, the mixture is extracted with phenol/chloroform and ethanol precipitated followed by running over a Sephadex G-50 spin column.
  • Klenow E. coli DNA polymerase I
  • dNTPs deoxynucleotide triphosphates
  • Synthetic oligonucleotides are prepared using commercially available automated oligonucleotide synthesizers. In the proteins of the invention, however, a synthetic gene is conveniently employed.
  • the gene design can include restriction sites which permit easy manipulation of the gene to replace coding sequence portions with these encoding analogs.
  • the constructed vector is then transformed into a suitable host for production of the protein.
  • transformation is done using standard techniques appropriate to such cells.
  • the calcium treatment employing calcium chloride, as described by Cohen, S. N., Proc. Natl. Acad. Sci. USA (1972) 69:2110, or the RbCl method described in Maniatis, et al., Molecular Cloning: A Laboratory Manual (1982) Cold Spring Harbor Press, p. 254 is used for procaryotes or other cells which contain substantial cell wall barriers.
  • the calcium phosphate precipitation method of Graham and van der Eb, Virology (1978) 52:546 or electroporation is preferred.
  • Transformations into yeast are carried out according to the method of Van Solingen, P., et al., J. Bacter. (1977) 130:946 and Hsiao, C. L., et al., Proc. Natl. Acad. Sci. USA (1979) 76:3829.
  • the transformed cells are then cultured under conditions favoring expression of the MHC sequence and the recombinantly produced protein recovered from the culture.
  • MHC-binding peptides are thought to be about 8 to about 10, possibly about 8 to about 11, or about 8 to about 12 residues in length, and contain both the agretope (recognized by the MHC molecule) and the epitope (recognized by T cell receptor on the T cell).
  • the epitope is a contiguous or noncontiguous sequence of about 5-6 amino acids that is recognized by the antigen-specific T cell receptor.
  • the agretope is a continuous or noncontiguous sequence that is responsible for binding of the peptide with the MHC glycoproteins.
  • the invention provides systems, kits, and assays for evaluating putative MHC-binding peptides to determine whether such fragments can be incorporated into a ternary complex with an MHC monomer or modified MHC monomer.
  • the invention provides systems, kits and screening methods to be used in screening of candidate peptides for use in diagnostic assays, vaccines, and other treatment modalities.
  • Putative MHC-binding peptides for use in the invention methods can be made using any method known in the art, the most convenient being peptide synthesis for fragments of 8 to 12 amino acids in length.
  • the invention provides a system comprising a solid surface having attached thereto one or more MHC monomer or modified MHC monomer wherein the monomer denatures in a denaturing condition and reconstitutes to form a functional binding pocket containing a suitable MHC-binding peptide under reconstituting conditions.
  • a plurality of the monomers can be bound to a single surface.
  • the surface of the system can be any known or later discovered solid surface including, without any limitation, any solid, polymer, membrane, synthetic surface, and the like.
  • the solid surface of the invention system can be a microtiter plate, such as the wells of a microtiter plate, or a bead, such as an agarose A bead, an agarose G bead, and the like.
  • the solid surface of the invention system is suitable for use in a high throughput scanning system, e.g., the surface is compatible with the high throughput system or allows a system to work with the entities associated with the surface in a high throughput manner, such as fluorescence determined flow cytometry.
  • streptagII binding affinity
  • StrepTactin a short peptide sequence
  • the molecule d-biotin, which binds with higher affinity to strepTactin (Kd ⁇ 1 ⁇ 10 ⁇ 13M) effectively competes with the StrepTagII for the binding site.
  • Attachment of the MHC monomers to the solid surface can be accomplished by any method known in the art.
  • the solid surface can be coated with a first binding ligand, such as avidin, and the monomer is then provided with a second binding ligand, such as biotin, wherein the first ligand binds specifically with the second ligand.
  • the second binding ligand may optionally be attached to the monomers via a C-terminal end. Attachment of the one or more monomers to the solid surface is optionally reversible or cleavable.
  • cleavable binding complex is commercially available from Amersham Bioscience Bioscience (Orsay France) such as Factor Xa, PreScission Protease and thrombin. All of these proteases can be used with the GST affinity tag from proteins expressed using pGEX-T vectors
  • the invention system comprising a solid support with attached MHC monomers is preferably stored in a renatured state, by causing formation of a ternary complex with the MHC monomer containing a MHC binding peptide of 8 to 12, or about 9 to 11 amino acids in the binding pocket and a beta-2 microglobulin molecule bound thereto, as described herein.
  • renaturation Formation of the ternary complex containing a MHC heavy chainor modified MHC attached to a solid support is referred to herein as “renaturation” and is accomplished under renaturing conditions as is know in the art and described herein.
  • renaturing conditions typically include the presence of a suitable MHC binding peptide for the monomer, the presence of beta-2 microglobulin, and a suitable refolding buffer having a pH of from about 7 to about 8.5. Suitable refolding buffers are illustrated in the Examples herein and are known in the art.
  • the solid support with bound MHC monomer(s) can be dried while in a renatured state, for example by exposure to a buffer containing sugars.
  • the solid support and attached MHC monomers in ternary complex are exposed to denaturing conditions to cause dissociation and unfolding of the monomers.
  • denaturing conditions can comprise exposure of the solid support and bound monomers to a pH of about 2 to about 4 for sufficient time to cause dissociation of the ternary complexes without damage to the monomers.
  • the invention system may further comprise a monoclonal antibody, described in greater detail below, that binds specifically to a conformational epitope that is present in the ternary complex and absent in the dissociated components of such a complex.
  • a conformational epitope may be formed in the reconstituted MHC monomers or modified MHC monomers used in the system and absent in the denatured monomers.
  • the invention system may further contain a supply of beta-2 microglobulin.
  • the MHC monomer used in the invention systems and methods can be any MHC monomer or modified MHC monomer, i.e., class I heavy chain, capable of binding a peptide in the range of 8 to 11 amino acids, for example 8 to 10 amino acids under renaturing conditions.
  • the MHC monomer can be encoded by any partial or full-length modified or unmodified MHC gene sequence from any species or subtype, or a combination thereof, including without limitation human and murine species, and chimera thereof.
  • Preferred MHC encoding gene sequences are those encoding any HLA allele genotype and any variation or polymorphism thereof.
  • the MHC monomer utilized in the invention systems and methods can be any partial or full-length HLA heavy chain that binds an HLA-binding peptide under renaturing conditions, i.e., any subtype or allele of HLA-A, HLA-B, or HLA-C,
  • the MHC monomer is modified by truncation to include only the ⁇ 1 , ⁇ 2 and the ⁇ 3 domains of an HLA heavy chain.
  • the MHC monomer can be a chimeric, such as a fusion protein, containing these MHC domains and an anchor domain, wherein the MHC domain binds to a MHC-binding a peptide, as described herein, while the anchor domain is suitable for immobilizing the MHC monomer to a surface.
  • the anchor domain can be a polypeptide fused with the HLA domain to form a fusion protein or can be any entity coupled to the HLA domain through any suitable means known in the art, e.g., biotinylated MHC monomer.
  • the MHC monomer can be attached to the solid surface by any suitable means known in the art.
  • the MHC monomer can be immobilized to a surface either directly or indirectly, e.g., via an anchoring or connecting entity.
  • the solid surface of the invention system is coated with a first ligand entity, which binds to or interacts with a second ligand connected to or within the MHC monomer, e.g., via covalent or noncovalent bond.
  • the surface is coated with avidin or its derivatives, e.g., streptavidin, and the MHC monomer contains biotin or its derivatives as its anchor domain. Attachment of the MHC monomer to the solid surface, in one embodiment of the invention, is reversible or cleavable.
  • the MHC monomer coated or immobilized to a solid surface can be denatured, e.g., stripped or dissociated in a denaturing condition, and then renatured, e.g., refolded from a denatured form under a non-denaturing or renaturing condition so as to bind an appropriate MHC-binding peptide.
  • the surface coated with the MHC monomer provided by the present invention can be dried and stored for use at a later time.
  • the storage is at 4 degrees C.
  • the system of the invention can further include a monoclonal antibody and a peptide.
  • the peptide can be any peptide that binds to the HLA heavy chain monomers, e.g., MHC-binding peptides.
  • the peptide has high affinity to the MHC monomer, e.g., HBc high affinity peptide.
  • the monoclonal antibody used in the invention systems and methods can be any monoclonal antibody that specifically binds to a conformational epitope present only in a ternary complex of an MHC monomer and not present in dissociated components of the ternary complex.
  • the conformational epitope can be present in beta-2 microglobulin when incorporated into the ternary complex.
  • the monoclonal antibody can recognize a conformational epitope present in the MHC monomer or modified MHC monomer being used in a particular invention system or method.
  • the monoclonal antibody may be species-matched to the MHC monomers, for example, when the solid support has attached HLA class I monomers, the monoclonal antibody is a murine, human or humanized anti-MHC class I monoclonal antibody.
  • the modified MHC monomer is a chimera containing domains from more than one species, the anti-MHC monoclonal antibody can be selected to bind to a conformational epitope present in only one of the domains. For example, as illustrated in FIG.
  • a ternary complex containing modified MHC monomer that is a chimera containing alpha-1 and alpha 2 domains of HLA-A2 heavy chain and a murine alpha-3 domain of H-2 Kb can be detected by a murine monoclonal antibody that binds to a conformational domain in the murine alpha-3 domain.
  • the monoclonal antibody can be any anti-MHC class I monoclonal antibody that recognizes any subclass of HLA monomer in a ternary complex, i.e., HLA-A, HLA-B or HLA-C.
  • a preferred anti-MHC-class I monoclonal antibody for use in the invention systems and methods is a mouse IgG2a conformational dependent anti-HLA monoclonal antibody produced by hybridoma B9.12.1, which as been deposited under the provisions of the Budapest Treaty on the International Recognition of the Deposit of Microorganisms for the Purpose of Patent Procedure and the Regulations thereunder (Budapest Treaty) at Collection Nationale de Cultures de Microorganismes (CNCM), Institut Pasteur 25, Rue du Dotica Roux, F75724 Paris Cedex 15 France, under registration number CNCM I-2941. This assures maintenance of viable cultures for 30 years from the date of deposit.
  • the monoclonal antibody used in the invention systems and methods is provided with a detectable label, i.e., a label that produces a detectable signal as is known in the art.
  • Labels may be conjugated to the antibody using any of a variety of procedures known in the art.
  • the antibody can be produced to include a label, such as a radioactive amino acid.
  • Labels suitable for use in the invention systems, kits and methods include, but are not limited to, radioisotopes, fluorochromes, enzymes, biotin and electron dense molecules.
  • Binding of the monoclonal antibody indicates formation of a ternary complex by binding of a MHC-binding peptide to the monomer and can be easily detected and/or quantified by detecting the signals produced by the signal entity after washing away unbound antibody and other components of the system.
  • a detectable label presently preferred is a fluorescent label, e.g., FITC.
  • the binding of fluorescently labeled antibodies on the solid support can be readily detected using a fluorimeter or by fluorescence determined flow cytometry.
  • the invention system can be provided either as part of another system or as a kit.
  • microtiter plates coated with the MHC monomers or modified monomers e.g., in dried form
  • a kit which can optionally additionally include, in separate vials or containers, an anti-MHC monoclonal antibody or an anti-beta-2 microglobulin antibody, as described herein, and a control peptide that binds specifically to the monomers attached to the solid support.
  • the kit includes an instruction explaining the procedures for using the system to conduct immunoassays, e.g., the methods provided by the present invention.
  • the kit can optionally also include any or all of the following: denaturing or refolding buffers, controls for the MHC monomers, the peptide, or the monoclonal antibody.
  • the invention provides methods for determining binding between a MHC monomer or modified MHC monomer and a putative MHC-binding peptide to be tested for binding to the monomer(s).
  • a solid surface having attached thereto a plurality of MHC monomers or modified MHC monomers is incubated in the presence and absence of the putative MHC-binding peptide.
  • the solid surface is one belonging to an invention system or kit and is prepared as described herein.
  • the MHC monomers attached to the solid support at the start of the assay procedure are in a reconstituted form
  • the MHC monomers are prepared for the assay by exposure to denaturing conditions as described herein, for example by exposure to a pH in the range from about 2 to about 4, or exposure overnight to a temperature higher than about 37° C. After denaturation, unbound MHC-binding peptides are washed away.
  • the solid support with attached denatured MHC monomers or modified MHC monomers is incubated with a putative MHC-binding peptide under reconstituting conditions for a suitable period of time to allow for formation of ternary complexes.
  • the reconstituting conditions will include the presence of a sufficient amount of beta-2 microglobulin (or beta 2 microglobulin modified to increase binding or stabilize ternary complex formation) to saturate the MHC monomers.
  • beta 2-microglobulin may be modified by attachment thereto of a stabilizing molecule, such as a leucine zipper, or the like, to stabilize ternary complex formation.
  • the reconstituting conditions may also include a temperature in the range from about minus 18° C. to about 37° C., for example about 4° C. to about 8° C.
  • binding to the MHC monomers of the putative MHC-binding peptide is determined by contacting the MHC monomers on the solid support with a monoclonal antibody that binds to a conformational epitope present only in ternary complex, for example a conformational epitope present in the refolded MHC monomer of the ternary complex and not present in a denatured MHC monomer. Binding of the antibody with the ternary complex attached to the solid support indicates that the putative MHC-binding peptide is an MHC-binding peptide specific for the MHC monomers or modified MHC monomers used in the assay.
  • a parallel assay (e.g., under the same reconstituting conditions, same monomer, and in the presence of the same monoclonal antibody) may be conducted using the monomers. Binding of the monoclonal antibody in the parallel assay to the ternary complex containing the standard MHC-binding peptide can be compared to binding of the monoclonal antibody to the ternary complex in the test assay to aid in determining the binding efficiency of the putative MHC-binding peptide, using computational methods known in the art..
  • the invention provides methods for determining the degree of binding affinity of an MHC monomer or modified MHC monomer for a putative MHC-binding peptide.
  • at least one denatured MHC monomer or modified MHC monomer attached to a solid surface is incubated under reconstituting conditions with the putative MHC-binding peptide and a monoclonal antibody that specifically binds to a conformational epitope created by formation of a ternary complex containing a corresponding reconstituted MHC monomer that is not present in any of the dissociated components of the complex.
  • Binding of the monoclonal antibody to the ternary complex so formed is compared with binding of the monoclonal antibody to a corresponding ternary complex containing the same MHC monomer or modified MHC monomer and a known MHC-binding peptide.
  • the difference in the binding indicates the relative degree of binding affinity of the reconstituted MHC monomer or modified MHC monomer for the putative MHC-binding peptide.
  • the test is done in multiples using different peptide concentrations in each parallel test.
  • the MHC monomers may belong to any species for which determination of appropriate class I binding peptides is desired, including, without limitation, murine and human or a chimera containing monomer subunits from a combination of species or subtypes.
  • Various readily available means can be used to determine the specific binding of the monoclonal antibody to the ternary complex containing the reconstituted MHC monomer.
  • the binding can be detected by directly labeling the monoclonal antibody with a detectable label, i.e., one that produces a detectable signal, and detecting the signal or via a secondary antibody which is detectably labeled and recognizes the monoclonal antibody that binds to the ternary complex containing the MHC monomer used in the assay.
  • Suitable detectable labels that can be used for this purpose are well known in the art and include labels selected from the group consisting of radioisotopes, fluorochromes, enzymes, biotin, electron dense molecules, and the like.
  • Fluorochromes or fluorescent labels are currently preferred since binding can readily be detected by subjecting the solid support to a fluorimeter.
  • the solid support is a plate, such as a 96 well microtiter plate, or beads, such as agarose A or agarose G beads
  • the assay can take advantage of high through-put florescence scanning using any of the methods known in the art.
  • HLA-A2m monomer in ternary complex with binding peptide Mart-1 26-35L was incubated at various concentrations with an anti-HLA-ABC-FITC or anti-HLA-FITC monoclonal antibody at concentrations of 0, 0.25, 0.5, 1, 2, and 4 ⁇ g/ml. Specifically, for each antibody concentration, the HLA monomer was added at concentrations of 0, 0.0078, 0.0156, 0.03125, 0.0625, 0.125, 0.25, and 0.5 ⁇ g/ml.
  • Buffer 1 Tris, Arginine, EDTA, GSH, GSSG and BSA
  • Buffer 2 Tris, NaCl, EDTA, NaN 3 , BSA and 0.05% TWEEN 20® detergent
  • Buffer 3 Tris, NaCl, EDTA, NaN 3 , BSA and 0.05% NONIDET® P40 detergent.
  • Renaturation of the HLA monomers was tested after 24 hours and 48 hours of incubation with 2 ⁇ g/ml of anti-HLA-class I-FITC conjugate. As shown in FIG. 5, the FITC signal increased as a function of the peptide concentration. This result shows that the HLA monomer renaturated by incorporation into a ternary complex and that renaturation of MHC monomers can be effectively detected with an anti-HLA-class I-FITC antibody. It was found that the best renaturation buffer was the Buffer 2 containing TWEEN 20®. Interestingly no refolding was measured with Buffer 1.
  • the best temperature for the antibody binding assay was 4° C.-8° C. and the best incubation period to allow renaturation was 24 hours.
  • Each well of white 96-well microtiter plates were coated with 200 ⁇ l of a 5 ⁇ g/ml biotinylated BSA solution in PBS and the plates were incubated for 16 hours at 4° C. The plates were washed and then 200 ⁇ l/well of avidin solution at 5 ⁇ g/ml was added. The plates were then incubated for another 16 hours at 4° C.
  • the assay procedure was as follows. Each sample 200 ⁇ l/well containing the HLA monomer in ternary complex at 0.25 ⁇ g/ml and diluted in Tris 10 mM, NaCl 150 mM, EDTA 0.5 mM, NaN3 0.1%, BSA 0.2%, was loaded into wells of the avidin-coated plate and incubated for 1 hour at room temperature on an orbital shaker in the dark. The wells were then rinsed three times with an automatic washer (SLT, Salzburg, Austria) using 300 ⁇ l of a 9 g/l NaCl solution containing 0.05% TWEEN 80®.
  • FITC-conjugated anti-HLA-class I antibody at 2 ⁇ g/ml were added.
  • the plates were incubated for 45 min at room temperature on an orbital shaker in the dark, washed three times, and 200 ⁇ l/well of Tris 10 mM, NaCl 150 mM, EDTA 0.5mM, NaN3 0.1%, BSA 0.2% were added.
  • the FITC fluorescence was measured with a Perkin Elmer LS-50B fluorimeter following these parameters:
  • Step 1 Step 2 Step 3 Step 4
  • HLA Incubate 200 ⁇ l/well Three washes Three washes heavy of each sample in Add 200 ⁇ l/well of Add 200 ⁇ l of chain and the 96-well anti-HLA-class I buffer Fluores- streptavidin streptavidin coated mAb at 2 ⁇ g/ml cence deter- PE white plates.
  • Incubate 45 min at mination Incubate 1 hour at room temperature in room temperature in the dark under the dark under agitation agitation
  • HLA-A*0201/Mart1 reconstituted monomers in various concentrations was incubated with various concentrations of the anti-HLA-class I-FITC mAb. As shown in FIG. 6, a plateau was reached with concentrations of anti-HLA-class I mAb at 1 to 2 ⁇ g/ml for all HLA heavy chain monomer concentrations.
  • a dose response curve at various concentration of reconstituted monomers was plotted using using 2 ⁇ g/ml of anti-HLA-ABC mAb. As shown in FIGS. 7A and 7B, the signal remained linear with increasing concentrations until 0.5 ⁇ g/ml of reconstituted HLA monomer was used. Concentrations of the reconstituted HLA monomer higher than 0.5 ⁇ g/ml provided signals that were very close to a plateau.
  • the data summarized in FIGS. 7A and 7B demonstrate that for the best result, the assay conditions should include 0.25 ⁇ g/ml of reconstituted HLA monomer and 2 ⁇ g/ml of anti-HLA-class I FITC mAb. These data also indicated that the sensitivity of the assay is about 4 to 6 ng/ml of the reconstituted HLA monomer.
  • class I MHC molecules For effective CD8+ T cell responses, class I MHC molecules must bind many peptides of diverse sequence in sufficient abundance for a long period of time. Many tumor cells appear to escape the immune response because antigenic peptides do not bind well to class I MHC molecules that present them. If a peptide does not bind efficiently to the MHC molecule, circulating T cells will not recognize the MHC ternary complex, and cells presenting them will not be eliminated.
  • Typical half-lives of immunodominant peptides are greater than 20 hours at 37° C. (Stuber, et al., (1994) Eur. J. immunol. 24, 765-768, and Pogue, et al., (1995) Proc. Natl. Acad. Sci. US 92, 8166-8170). From this evidence, a test was developed to use the invention solid phase assay to determine the stability of various complexes at different temperatures, and thus calculate the off rate of the peptides. This parameter is very valuable to know when peptides are used in vaccination for the purpose of eliciting an immune response.
  • B0 is the fluorescence determined at time zero. The time zero corresponds to the moment when the plates were washed once the monomer was reconstituted and the plates were placed at different temperatures. B is the fluorescence obtained at each time. After the Ln (Fluorescence Emission) as a function of the time was plotted.
  • FIG. 10 shows graphs of the dissociation curves for renatured peptides.
  • FIGS. 10 E-H shows graphs of the off rates for peptides.
  • FIG. 10I shows the effect of temperature on monomer dissociation).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Urology & Nephrology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
US10/269,473 2002-10-11 2002-10-11 Methods and systems for detecting MHC class I binding peptides Abandoned US20040072262A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
US10/269,473 US20040072262A1 (en) 2002-10-11 2002-10-11 Methods and systems for detecting MHC class I binding peptides
US10/684,268 US20040137537A1 (en) 2002-10-11 2003-10-10 Methods and systems for detecting MHC class I and II binding peptides
AU2003284113A AU2003284113B2 (en) 2002-10-11 2003-10-10 Methods and systems for detecting MHC class I and class II binding peptides
CA2501864A CA2501864C (en) 2002-10-11 2003-10-10 Methods and systems for detecting mhc class i and class ii binding peptides
PCT/US2003/032370 WO2004034033A2 (en) 2002-10-11 2003-10-10 Methods and systems for detecting mhc class i and class ii binding peptides
JP2004543745A JP4607590B2 (ja) 2002-10-11 2003-10-10 Mhcクラスiおよびクラスii結合ペプチドを検出するための方法およびシステム
EP03776344A EP1549952B1 (en) 2002-10-11 2003-10-10 Methods and systems for detecting mhc class i and class ii binding peptides
CN2003801010208A CN1703624B (zh) 2002-10-11 2003-10-10 用于检测mhc-ⅰ类和mhc-ⅱ类结合肽的方法和体系
US13/029,902 US8815528B2 (en) 2002-10-11 2011-02-17 Methods and systems for detecting MHC class I binding peptides
US14/336,944 US20150072886A1 (en) 2002-10-11 2014-07-21 Methods and Systems for Detecting MHC Class I Binding Peptides

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US10/269,473 US20040072262A1 (en) 2002-10-11 2002-10-11 Methods and systems for detecting MHC class I binding peptides

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10/684,268 Continuation-In-Part US20040137537A1 (en) 2002-10-11 2003-10-10 Methods and systems for detecting MHC class I and II binding peptides
US13/029,902 Continuation US8815528B2 (en) 2002-10-11 2011-02-17 Methods and systems for detecting MHC class I binding peptides

Publications (1)

Publication Number Publication Date
US20040072262A1 true US20040072262A1 (en) 2004-04-15

Family

ID=32068790

Family Applications (4)

Application Number Title Priority Date Filing Date
US10/269,473 Abandoned US20040072262A1 (en) 2002-10-11 2002-10-11 Methods and systems for detecting MHC class I binding peptides
US10/684,268 Abandoned US20040137537A1 (en) 2002-10-11 2003-10-10 Methods and systems for detecting MHC class I and II binding peptides
US13/029,902 Expired - Lifetime US8815528B2 (en) 2002-10-11 2011-02-17 Methods and systems for detecting MHC class I binding peptides
US14/336,944 Abandoned US20150072886A1 (en) 2002-10-11 2014-07-21 Methods and Systems for Detecting MHC Class I Binding Peptides

Family Applications After (3)

Application Number Title Priority Date Filing Date
US10/684,268 Abandoned US20040137537A1 (en) 2002-10-11 2003-10-10 Methods and systems for detecting MHC class I and II binding peptides
US13/029,902 Expired - Lifetime US8815528B2 (en) 2002-10-11 2011-02-17 Methods and systems for detecting MHC class I binding peptides
US14/336,944 Abandoned US20150072886A1 (en) 2002-10-11 2014-07-21 Methods and Systems for Detecting MHC Class I Binding Peptides

Country Status (7)

Country Link
US (4) US20040072262A1 (sl)
EP (1) EP1549952B1 (sl)
JP (1) JP4607590B2 (sl)
CN (1) CN1703624B (sl)
AU (1) AU2003284113B2 (sl)
CA (1) CA2501864C (sl)
WO (1) WO2004034033A2 (sl)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040137537A1 (en) * 2002-10-11 2004-07-15 Beckman Coulter, Inc. Methods and systems for detecting MHC class I and II binding peptides
US20050095655A1 (en) * 2003-11-03 2005-05-05 Montero-Julian Felix A. Solution-based methods for detecting MHC-binding peptides
US20050287611A1 (en) * 2004-05-07 2005-12-29 Nugent C T Iv MHC bridging system for detecting CTL-mediated lysis of antigen presenting cells
US20060040332A1 (en) * 2004-06-17 2006-02-23 Beckman Coulter Mycobacterium tuberculosis epitopes and methods of use thereof
WO2006031883A2 (en) * 2004-09-13 2006-03-23 Xencor, Inc. Analysis of mhc-peptide binding interactions
WO2007034151A2 (en) * 2005-09-19 2007-03-29 Proimmune Limited A method of screening mhc molecules
US20080064060A1 (en) * 2004-05-06 2008-03-13 Rainer Blasczyk Method For Productoin Of Soluble Mhc Proteins
WO2008057366A2 (en) 2006-11-01 2008-05-15 Beckman Coulter, Inc. Binding surfaces for affinity assays
US20090061478A1 (en) * 2006-01-30 2009-03-05 Lene Have Poulsen High-Speed Quantification of Antigen Specific T-Cells in Whole Blood by Flow Cytometry
EP2232257A1 (en) * 2008-01-08 2010-09-29 Siemens Healthcare Diagnostics Inc. Stabilization of solid support assay reagents
US20100248257A1 (en) * 2007-07-03 2010-09-30 Dako Denmark A/S Compiled Methods for Analysing and Sorting Samples
US20110195435A1 (en) * 2008-09-20 2011-08-11 Andrew Kelvin Sewell Use of a protein kinase inhibitor to detect immune cells, such as t cells
US20110212090A1 (en) * 2008-07-23 2011-09-01 Dako Denmark A/S Combinatorial Analysis and Repair
WO2012022975A1 (en) * 2010-08-17 2012-02-23 Isis Innovation Limited Identification of ligands and their use
US8268964B2 (en) 2007-03-26 2012-09-18 Dako Denmark A/S MHC peptide complexes and uses thereof in infectious diseases
US10351847B2 (en) 2013-07-26 2019-07-16 Oxford University Innovation Limited Identification and display of peptide ligands
US10369204B2 (en) 2008-10-02 2019-08-06 Dako Denmark A/S Molecular vaccines for infectious disease
WO2019211864A1 (en) * 2018-04-29 2019-11-07 Chimera Translational Research Fraternity Private Limited Molecular fluorescent multiplex assay to identify allo-antibodies against mhc antigens
US10611818B2 (en) 2007-09-27 2020-04-07 Agilent Technologies, Inc. MHC multimers in tuberculosis diagnostics, vaccine and therapeutics
US10968269B1 (en) 2008-02-28 2021-04-06 Agilent Technologies, Inc. MHC multimers in borrelia diagnostics and disease
US11992518B2 (en) 2008-10-02 2024-05-28 Agilent Technologies, Inc. Molecular vaccines for infectious disease

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8396671B2 (en) * 2006-02-16 2013-03-12 Microsoft Corporation Cluster modeling, and learning cluster specific parameters of an adaptive double threading model
US20070192033A1 (en) * 2006-02-16 2007-08-16 Microsoft Corporation Molecular interaction predictors
US8706421B2 (en) * 2006-02-16 2014-04-22 Microsoft Corporation Shift-invariant predictions
US8121797B2 (en) 2007-01-12 2012-02-21 Microsoft Corporation T-cell epitope prediction
US7972804B2 (en) * 2007-07-03 2011-07-05 One Lambda Methods of detecting antibodies specific for denatured HLA antigens
US8628932B2 (en) 2007-07-03 2014-01-14 One Lambda Methods of detecting antibodies specific for denatured HLA antigens
US8927462B2 (en) 2010-04-12 2015-01-06 Academia Sinica Method for identifying HLA complexes associated with adverse drug reactions
CA2852962C (en) 2011-10-28 2022-05-03 Regeneron Pharmaceuticals, Inc. Genetically modified mice expressing chimeric major histocompatibility complex (mhc) ii molecules
KR102211267B1 (ko) 2013-02-22 2021-02-04 리제너론 파아마슈티컬스, 인크. 사람화된 주요 조직적합성 복합체를 발현하는 마우스
CA2959428A1 (en) 2014-09-19 2016-03-24 Regeneron Pharmaceuticals, Inc. Chimeric antigen receptors
MX2017012829A (es) 2015-04-06 2018-02-23 Regeneron Pharma Respuestas inmunitarias mediadas por células t humanizadas en animales no humanos.
US20170205404A1 (en) * 2016-01-19 2017-07-20 General Electric Company Multifunctional beads and methods of use for capturing rare cells
RU2653017C1 (ru) * 2017-06-22 2018-05-04 Общество с ограниченной ответственностью "Лидер Гласс" Способ переработки специальных видов стекла
WO2019054409A1 (ja) * 2017-09-12 2019-03-21 国立大学法人北海道大学 Hlaタンパク質に相互作用する物質のスクリーニング方法及びスクリーニング用キット

Citations (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4017597A (en) * 1974-10-30 1977-04-12 Monsanto Company Unitized solid phase immunoassay kit and method
US4048298A (en) * 1975-02-25 1977-09-13 Rohm And Haas Company Solid phase double-antibody radioimmunoassay procedure
US4120945A (en) * 1976-07-06 1978-10-17 Becton, Dickinson & Company Substrate coated with receptor and labeled ligand for assays
US4208479A (en) * 1977-07-14 1980-06-17 Syva Company Label modified immunoassays
US4228237A (en) * 1978-09-21 1980-10-14 Calbiochem-Behring Corp. Methods for the detection and determination of ligands
US4478946A (en) * 1981-07-02 1984-10-23 South African Inventions Development Corporation Carrier bound immunosorbent
US4912030A (en) * 1985-01-15 1990-03-27 Institute Of Cancer Research Viral isolates and their use in diagnosis
US5187065A (en) * 1989-12-22 1993-02-16 Schutzer Steven E Method and materials for detecting lyme disease
US5514557A (en) * 1994-06-06 1996-05-07 Genetic Testing Institute Inc. Method and kit for detecting antibodies specific for HLA and/or platelet glycoproteins
US5534416A (en) * 1993-04-13 1996-07-09 Molecular Probes, Inc. Fluorescent viability assay using cyclic-substituted unsymmetrical cyanine dyes
US5583031A (en) * 1992-02-06 1996-12-10 President And Fellows Of Harvard College Empty major histocompatibility class II heterodimers
US5599720A (en) * 1982-08-27 1997-02-04 Multilyte Limited Measurement of analyte concentration
US5635363A (en) * 1995-02-28 1997-06-03 The Board Of Trustees Of The Leland Stanford Junior University Compositions and methods for the detection, quantitation and purification of antigen-specific T cells
US5734023A (en) * 1991-11-19 1998-03-31 Anergen Inc. MHC class II β chain/peptide complexes useful in ameliorating deleterious immune responses
US5759774A (en) * 1988-05-18 1998-06-02 Cobe Laboratories, Inc. Method of detecting circulating antibody types using dried or lyophilized cells
US5919639A (en) * 1996-06-26 1999-07-06 University Of Massachusetts Ii peptide therapeutics to enhance antigen presentation
US5965532A (en) * 1996-06-28 1999-10-12 Trustees Of Tufts College Multivalent compounds for crosslinking receptors and uses thereof
US6037135A (en) * 1992-08-07 2000-03-14 Epimmune Inc. Methods for making HLA binding peptides and their uses
US6046013A (en) * 1997-08-01 2000-04-04 Gti Process for identifying specific antibodies associated with HLA
US6225042B1 (en) * 1995-03-08 2001-05-01 The Scripps Research Institute Antigen presenting system and methods for activation of T-cells
US6270772B1 (en) * 1997-09-16 2001-08-07 Oregon Health Sciences University Recombinant MHC molecules useful for manipulation of antigen-specific T-cells
US6306636B1 (en) * 1997-09-19 2001-10-23 Arch Development Corporation Nucleic acid segments encoding wheat acetyl-CoA carboxylase
US20020006903A1 (en) * 1997-03-28 2002-01-17 Jonathan Schneck Use of multivalent chimeric peptide-loaded, MHC/Ig molecules to detect, activate or suppress antigen-specific T cell-dependent immune responses
US6355479B1 (en) * 1996-05-23 2002-03-12 The Scripps Research Institute MHC class II antigen-presenting systems and methods for activating CD4+ T cells
US6413517B1 (en) * 1997-01-23 2002-07-02 Epimmune, Inc. Identification of broadly reactive DR restricted epitopes
US6419931B1 (en) * 1991-08-26 2002-07-16 Epimmune Inc. Compositions and methods for eliciting CTL immunity
US20020106708A1 (en) * 2000-11-13 2002-08-08 Sigma-Aldrich Co. Assays reagents and kits for detecting or determining the concentration of analytes
US20020146746A1 (en) * 2001-04-05 2002-10-10 Douglas Nixon Methods of detecting specific cell lysis
US6485913B1 (en) * 1999-03-10 2002-11-26 Sequenom, Inc. Systems and methods for performing reactions in an unsealed environment
US20030044389A1 (en) * 2001-07-02 2003-03-06 Brown Patrick O. Microarrays for cell phenotyping and manipulation
US20030044415A1 (en) * 1998-06-05 2003-03-06 Savage Philip Michael Method for producing or enhancing a T-cell response against a target cell using a complex comprising an HLA class I molecule and an attaching means
US20030124513A1 (en) * 2001-05-29 2003-07-03 Mcswiggen James Enzymatic nucleic acid treatment of diseases or conditions related to levels of HIV
US20030124613A1 (en) * 2001-03-09 2003-07-03 Hildebrand William H. Epitope testing using soluble HLA
US20030166057A1 (en) * 1999-12-17 2003-09-04 Hildebrand William H. Method and apparatus for the production of soluble MHC antigens and uses thereof
US6727093B2 (en) * 1996-11-12 2004-04-27 City Of Hope HCMV-reactive T cells and uses therefor
US20040137537A1 (en) * 2002-10-11 2004-07-15 Beckman Coulter, Inc. Methods and systems for detecting MHC class I and II binding peptides
US20040253632A1 (en) * 2000-05-25 2004-12-16 Sunol Molecular Corporation Modulation of T -cell receptor interactions
US20050059107A1 (en) * 2001-10-17 2005-03-17 Bernard Maillere Method of selecting hla-dp4 ligands and the applications thereof
US20050095655A1 (en) * 2003-11-03 2005-05-05 Montero-Julian Felix A. Solution-based methods for detecting MHC-binding peptides
US20050287611A1 (en) * 2004-05-07 2005-12-29 Nugent C T Iv MHC bridging system for detecting CTL-mediated lysis of antigen presenting cells
US6992176B2 (en) * 2002-02-13 2006-01-31 Technion Research & Development Foundation Ltd. Antibody having a T-cell receptor-like specificity, yet higher affinity, and the use of same in the detection and treatment of cancer, viral infection and autoimmune disease
US20060040332A1 (en) * 2004-06-17 2006-02-23 Beckman Coulter Mycobacterium tuberculosis epitopes and methods of use thereof

Family Cites Families (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4444879A (en) * 1981-01-29 1984-04-24 Science Research Center, Inc. Immunoassay with article having support film and immunological counterpart of analyte
GB8508340D0 (en) * 1985-03-29 1985-05-09 Creighton T E Production of protein
FR2658197B1 (fr) * 1990-02-14 1992-05-22 Inst Nat Sante Rech Med Anticorps monoclonaux restreints reconnaissant un peptide associe a un antigene du complexe majeur d'histocompatibilite - applications au diagnostic et au traitement.
EP0555418A4 (en) 1990-10-30 1994-06-01 Immulogic Pharma Corp Peptide binding assays with mhc antigens
DE69332268T2 (de) 1992-10-15 2003-08-14 Toray Industries Verfahren zur herstellung von rekombinant mhcii protein in mikroorganismen
DE4238416A1 (de) 1992-11-13 1994-05-19 Max Planck Gesellschaft Bestimmung von Peptidmotiven auf MHC-Molekülen
NZ261571A (en) * 1993-02-04 1997-03-24 Denzyme Aps Producing correctly folded proteins using multiple denaturation and renaturation cycles
WO1995004817A1 (en) 1993-08-06 1995-02-16 Cytel Corporation Methods for ex vivo therapy using peptide-loaded antigen presenting cells for the activation of ctl
US6120992A (en) * 1993-11-04 2000-09-19 Valigene Corporation Use of immobilized mismatch binding protein for detection of mutations and polymorphisms, and allele identification in a diseased human
WO1997000067A1 (en) 1995-06-15 1997-01-03 University Of Victoria Innovation And Development Mycobacterium tuberculosis dna sequences encoding immunostimulatory peptides
DE19525784A1 (de) 1995-07-14 1997-01-16 Boehringer Mannheim Gmbh Autoreaktive Peptide aus der humanen Glutamin-Decarboxylase (GAD)
WO1997009429A2 (en) 1995-09-01 1997-03-13 Corixa Corporation Compounds and methods for diagnosis of tuberculosis
EP0958834B1 (en) 1996-11-13 2007-07-04 Meiji Dairies Corporation Use of peptide-based immunotherapeutic agent
BR9713145A (pt) 1996-11-27 2000-02-08 Eastman Chem Co Processo, composição absorvente de luz e compostos diácidos.
GB9725764D0 (en) 1997-12-04 1998-02-04 Isis Innovation HLA-E binding
AU3365499A (en) 1998-03-27 1999-10-18 Chancellor, Masters And Scholars Of The University Of Oxford, The Isolated multimeric complexes useful in analysis of t cells, peptides useful in making the complexes, and uses thereof
CA2325567A1 (en) * 1998-05-01 1999-11-11 Maxygen, Inc. Optimization of pest resistance genes using dna shuffling
WO2000015665A2 (en) 1998-09-14 2000-03-23 Pedersen Lars Oestergaard A method of producing a functional immunoglobulin superfamily protein
DE69934426T2 (de) * 1998-10-29 2007-10-04 Whitehead Institute For Biomedical Research, Cambridge KREBSIMMUNTHERAPIE UND KREBSDIAGNOSE UNTER VERWENDUNG UNIVERSELLER TUMORASSOZIIERTER ANTIGENE EINSCHLIESSLICH hTERT
ATE421976T1 (de) * 1999-05-18 2009-02-15 Dyax Corp Fab fragmentbibliotheken und verfahren für deren verwendung
US20060276629A9 (en) * 1999-12-17 2006-12-07 Hildebrand William H Purification and characterization of soluble human HLA proteins
US6649419B1 (en) * 2000-11-28 2003-11-18 Large Scale Proteomics Corp. Method and apparatus for protein manipulation
CA2440773A1 (en) * 2001-03-14 2002-09-19 Dakocytomation Denmark A/S Novel mhc molecule constructs, and methods of employing these constructs for diagnosis and therapy, and uses of mhc molecules
US20040248205A1 (en) 2003-04-16 2004-12-09 Stern Lawrence J. Major histocompatibility complex (MHC)-peptide arrays
DE602004030125D1 (de) 2003-07-22 2010-12-30 Beckman Coulter Inc Verfahren zum nachweis der aktivierung von t-zellen durch mhc-bindende peptide
JP4505401B2 (ja) 2005-10-28 2010-07-21 学校法人同志社 受光素子

Patent Citations (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4017597A (en) * 1974-10-30 1977-04-12 Monsanto Company Unitized solid phase immunoassay kit and method
US4048298A (en) * 1975-02-25 1977-09-13 Rohm And Haas Company Solid phase double-antibody radioimmunoassay procedure
US4120945A (en) * 1976-07-06 1978-10-17 Becton, Dickinson & Company Substrate coated with receptor and labeled ligand for assays
US4208479A (en) * 1977-07-14 1980-06-17 Syva Company Label modified immunoassays
US4228237A (en) * 1978-09-21 1980-10-14 Calbiochem-Behring Corp. Methods for the detection and determination of ligands
US4478946A (en) * 1981-07-02 1984-10-23 South African Inventions Development Corporation Carrier bound immunosorbent
US5599720A (en) * 1982-08-27 1997-02-04 Multilyte Limited Measurement of analyte concentration
US4912030A (en) * 1985-01-15 1990-03-27 Institute Of Cancer Research Viral isolates and their use in diagnosis
US5759774A (en) * 1988-05-18 1998-06-02 Cobe Laboratories, Inc. Method of detecting circulating antibody types using dried or lyophilized cells
US5187065A (en) * 1989-12-22 1993-02-16 Schutzer Steven E Method and materials for detecting lyme disease
US6419931B1 (en) * 1991-08-26 2002-07-16 Epimmune Inc. Compositions and methods for eliciting CTL immunity
US5734023A (en) * 1991-11-19 1998-03-31 Anergen Inc. MHC class II β chain/peptide complexes useful in ameliorating deleterious immune responses
US5583031A (en) * 1992-02-06 1996-12-10 President And Fellows Of Harvard College Empty major histocompatibility class II heterodimers
US6037135A (en) * 1992-08-07 2000-03-14 Epimmune Inc. Methods for making HLA binding peptides and their uses
US5534416A (en) * 1993-04-13 1996-07-09 Molecular Probes, Inc. Fluorescent viability assay using cyclic-substituted unsymmetrical cyanine dyes
US5514557A (en) * 1994-06-06 1996-05-07 Genetic Testing Institute Inc. Method and kit for detecting antibodies specific for HLA and/or platelet glycoproteins
US5635363A (en) * 1995-02-28 1997-06-03 The Board Of Trustees Of The Leland Stanford Junior University Compositions and methods for the detection, quantitation and purification of antigen-specific T cells
US6225042B1 (en) * 1995-03-08 2001-05-01 The Scripps Research Institute Antigen presenting system and methods for activation of T-cells
US6355479B1 (en) * 1996-05-23 2002-03-12 The Scripps Research Institute MHC class II antigen-presenting systems and methods for activating CD4+ T cells
US5919639A (en) * 1996-06-26 1999-07-06 University Of Massachusetts Ii peptide therapeutics to enhance antigen presentation
US5965532A (en) * 1996-06-28 1999-10-12 Trustees Of Tufts College Multivalent compounds for crosslinking receptors and uses thereof
US6727093B2 (en) * 1996-11-12 2004-04-27 City Of Hope HCMV-reactive T cells and uses therefor
US6413517B1 (en) * 1997-01-23 2002-07-02 Epimmune, Inc. Identification of broadly reactive DR restricted epitopes
US20020006903A1 (en) * 1997-03-28 2002-01-17 Jonathan Schneck Use of multivalent chimeric peptide-loaded, MHC/Ig molecules to detect, activate or suppress antigen-specific T cell-dependent immune responses
US6046013A (en) * 1997-08-01 2000-04-04 Gti Process for identifying specific antibodies associated with HLA
US6270772B1 (en) * 1997-09-16 2001-08-07 Oregon Health Sciences University Recombinant MHC molecules useful for manipulation of antigen-specific T-cells
US6306636B1 (en) * 1997-09-19 2001-10-23 Arch Development Corporation Nucleic acid segments encoding wheat acetyl-CoA carboxylase
US20030044415A1 (en) * 1998-06-05 2003-03-06 Savage Philip Michael Method for producing or enhancing a T-cell response against a target cell using a complex comprising an HLA class I molecule and an attaching means
US6485913B1 (en) * 1999-03-10 2002-11-26 Sequenom, Inc. Systems and methods for performing reactions in an unsealed environment
US20030166057A1 (en) * 1999-12-17 2003-09-04 Hildebrand William H. Method and apparatus for the production of soluble MHC antigens and uses thereof
US20040253632A1 (en) * 2000-05-25 2004-12-16 Sunol Molecular Corporation Modulation of T -cell receptor interactions
US20020106708A1 (en) * 2000-11-13 2002-08-08 Sigma-Aldrich Co. Assays reagents and kits for detecting or determining the concentration of analytes
US20030124613A1 (en) * 2001-03-09 2003-07-03 Hildebrand William H. Epitope testing using soluble HLA
US20020146746A1 (en) * 2001-04-05 2002-10-10 Douglas Nixon Methods of detecting specific cell lysis
US20030124513A1 (en) * 2001-05-29 2003-07-03 Mcswiggen James Enzymatic nucleic acid treatment of diseases or conditions related to levels of HIV
US20030044389A1 (en) * 2001-07-02 2003-03-06 Brown Patrick O. Microarrays for cell phenotyping and manipulation
US20050059107A1 (en) * 2001-10-17 2005-03-17 Bernard Maillere Method of selecting hla-dp4 ligands and the applications thereof
US6992176B2 (en) * 2002-02-13 2006-01-31 Technion Research & Development Foundation Ltd. Antibody having a T-cell receptor-like specificity, yet higher affinity, and the use of same in the detection and treatment of cancer, viral infection and autoimmune disease
US20040137537A1 (en) * 2002-10-11 2004-07-15 Beckman Coulter, Inc. Methods and systems for detecting MHC class I and II binding peptides
US20050095655A1 (en) * 2003-11-03 2005-05-05 Montero-Julian Felix A. Solution-based methods for detecting MHC-binding peptides
US20050287611A1 (en) * 2004-05-07 2005-12-29 Nugent C T Iv MHC bridging system for detecting CTL-mediated lysis of antigen presenting cells
US20060040332A1 (en) * 2004-06-17 2006-02-23 Beckman Coulter Mycobacterium tuberculosis epitopes and methods of use thereof

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040137537A1 (en) * 2002-10-11 2004-07-15 Beckman Coulter, Inc. Methods and systems for detecting MHC class I and II binding peptides
US20050095655A1 (en) * 2003-11-03 2005-05-05 Montero-Julian Felix A. Solution-based methods for detecting MHC-binding peptides
US20080064060A1 (en) * 2004-05-06 2008-03-13 Rainer Blasczyk Method For Productoin Of Soluble Mhc Proteins
US20050287611A1 (en) * 2004-05-07 2005-12-29 Nugent C T Iv MHC bridging system for detecting CTL-mediated lysis of antigen presenting cells
US20060040332A1 (en) * 2004-06-17 2006-02-23 Beckman Coulter Mycobacterium tuberculosis epitopes and methods of use thereof
US7678379B2 (en) 2004-06-17 2010-03-16 Beckman Coulter, Inc. Mycobacterium tuberculosis epitopes and methods of use thereof
WO2006031883A3 (en) * 2004-09-13 2006-09-14 Xencor Inc Analysis of mhc-peptide binding interactions
WO2006031883A2 (en) * 2004-09-13 2006-03-23 Xencor, Inc. Analysis of mhc-peptide binding interactions
WO2007034151A2 (en) * 2005-09-19 2007-03-29 Proimmune Limited A method of screening mhc molecules
WO2007034151A3 (en) * 2005-09-19 2007-08-16 Proimmune Ltd A method of screening mhc molecules
US20080206789A1 (en) * 2005-09-19 2008-08-28 Proimmune Limited Method of Screening Mhc Molecules
US20090061478A1 (en) * 2006-01-30 2009-03-05 Lene Have Poulsen High-Speed Quantification of Antigen Specific T-Cells in Whole Blood by Flow Cytometry
US20080176340A1 (en) * 2006-11-01 2008-07-24 Beckman Coulter, Inc. Binding surfaces for affinity assays
EP2069790A2 (en) * 2006-11-01 2009-06-17 Beckman Coulter, Inc. Binding surfaces for affinity assays
EP2069790A4 (en) * 2006-11-01 2009-11-25 Beckman Coulter Inc BONDING SURFACES FOR AFFINITY TESTS
WO2008057366A2 (en) 2006-11-01 2008-05-15 Beckman Coulter, Inc. Binding surfaces for affinity assays
KR101457716B1 (ko) 2006-11-01 2014-11-03 베크만 컬터, 인코포레이티드 친화도 분석을 위한 결합 표면
US8268964B2 (en) 2007-03-26 2012-09-18 Dako Denmark A/S MHC peptide complexes and uses thereof in infectious diseases
US10336808B2 (en) 2007-03-26 2019-07-02 Dako Denmark A/S MHC peptide complexes and uses thereof in infectious diseases
US20100248257A1 (en) * 2007-07-03 2010-09-30 Dako Denmark A/S Compiled Methods for Analysing and Sorting Samples
US10030065B2 (en) 2007-07-03 2018-07-24 Dako Denmark A/S MHC multimers, methods for their generation, labeling and use
US10611818B2 (en) 2007-09-27 2020-04-07 Agilent Technologies, Inc. MHC multimers in tuberculosis diagnostics, vaccine and therapeutics
EP2232257A1 (en) * 2008-01-08 2010-09-29 Siemens Healthcare Diagnostics Inc. Stabilization of solid support assay reagents
EP2232257A4 (en) * 2008-01-08 2011-05-25 Siemens Healthcare Diagnostics Stabilization of test reagents on a solid surface
US10968269B1 (en) 2008-02-28 2021-04-06 Agilent Technologies, Inc. MHC multimers in borrelia diagnostics and disease
US20110212090A1 (en) * 2008-07-23 2011-09-01 Dako Denmark A/S Combinatorial Analysis and Repair
US10722562B2 (en) 2008-07-23 2020-07-28 Immudex Aps Combinatorial analysis and repair
US9404916B2 (en) 2008-09-20 2016-08-02 University College Cardiff Consultants Limited Use of a protein kinase inhibitor to detect immune cells, such as T cells
US20110195435A1 (en) * 2008-09-20 2011-08-11 Andrew Kelvin Sewell Use of a protein kinase inhibitor to detect immune cells, such as t cells
US10369204B2 (en) 2008-10-02 2019-08-06 Dako Denmark A/S Molecular vaccines for infectious disease
US11992518B2 (en) 2008-10-02 2024-05-28 Agilent Technologies, Inc. Molecular vaccines for infectious disease
WO2012022975A1 (en) * 2010-08-17 2012-02-23 Isis Innovation Limited Identification of ligands and their use
US10351847B2 (en) 2013-07-26 2019-07-16 Oxford University Innovation Limited Identification and display of peptide ligands
WO2019211864A1 (en) * 2018-04-29 2019-11-07 Chimera Translational Research Fraternity Private Limited Molecular fluorescent multiplex assay to identify allo-antibodies against mhc antigens

Also Published As

Publication number Publication date
EP1549952A2 (en) 2005-07-06
CA2501864C (en) 2012-12-18
CN1703624B (zh) 2012-07-04
JP4607590B2 (ja) 2011-01-05
WO2004034033A3 (en) 2004-09-10
US8815528B2 (en) 2014-08-26
EP1549952B1 (en) 2012-06-06
JP2006502416A (ja) 2006-01-19
US20040137537A1 (en) 2004-07-15
CA2501864A1 (en) 2004-04-22
US20150072886A1 (en) 2015-03-12
EP1549952A4 (en) 2007-04-25
CN1703624A (zh) 2005-11-30
AU2003284113A1 (en) 2004-05-04
AU2003284113B2 (en) 2010-02-18
US20110171752A1 (en) 2011-07-14
WO2004034033A2 (en) 2004-04-22

Similar Documents

Publication Publication Date Title
US8815528B2 (en) Methods and systems for detecting MHC class I binding peptides
US20050095655A1 (en) Solution-based methods for detecting MHC-binding peptides
US7678379B2 (en) Mycobacterium tuberculosis epitopes and methods of use thereof
Schirle et al. Combining computer algorithms with experimental approaches permits the rapid and accurate identification of T cell epitopes from defined antigens
US20060228758A1 (en) Analysis of MHC-peptide binding interactions
US20050287611A1 (en) MHC bridging system for detecting CTL-mediated lysis of antigen presenting cells
US20060084116A1 (en) Analysis of MHC-peptide binding interactions
US20040248205A1 (en) Major histocompatibility complex (MHC)-peptide arrays
US20060228759A1 (en) Analysis of MHC-peptide binding interactions
TWI795365B (zh) 製造和使用可溶性主要組織相容性複合體(mhc)分子類之方法
WO2022026921A1 (en) Identification and use of t cell epitopes in designing diagnostic and therapeutic approaches for covid-19
EP1648919B1 (en) Methods for detecting activation of t-cells by mhc binding peptides
EP4085069A1 (en) Systems and methods for identification of mhc-i peptide epitopes
WO2007032778A2 (en) Analysis of mhc-peptide binding interactions via population specific mhc-arrays
Delvig et al. CD4 T-cell epitope mapping
JP2019532920A (ja) Mhc結合ペプチドアレイおよびその使用方法

Legal Events

Date Code Title Description
AS Assignment

Owner name: BECKMAN COULTER INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MONTERO-JULIAN, FELIX A.;MONSEAUX, SYLVAIN;REEL/FRAME:013742/0414

Effective date: 20021105

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION