WO2022039604A1 - Dosage multiplex pour la maladie de johne et la grossesse - Google Patents

Dosage multiplex pour la maladie de johne et la grossesse Download PDF

Info

Publication number
WO2022039604A1
WO2022039604A1 PCT/NZ2021/050139 NZ2021050139W WO2022039604A1 WO 2022039604 A1 WO2022039604 A1 WO 2022039604A1 NZ 2021050139 W NZ2021050139 W NZ 2021050139W WO 2022039604 A1 WO2022039604 A1 WO 2022039604A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
substrate
pag
fragment
sample
Prior art date
Application number
PCT/NZ2021/050139
Other languages
English (en)
Inventor
Sandeep Kumar VASHIST
Jimena Maria TEJERINA
Original Assignee
Pictor Limited
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 Pictor Limited filed Critical Pictor Limited
Publication of WO2022039604A1 publication Critical patent/WO2022039604A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • 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
    • G01N33/02Food
    • G01N33/04Dairy products
    • 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
    • G01N33/5302Apparatus specially adapted for immunological test procedures
    • G01N33/5304Reaction vessels, e.g. agglutination plates
    • 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
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • G01N33/535Production of labelled immunochemicals with enzyme label or co-enzymes, co-factors, enzyme inhibitors or enzyme substrates
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54313Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals the carrier being characterised by its particulate form
    • G01N33/54326Magnetic particles
    • 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
    • G01N33/54353Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals with ligand attached to the carrier via a chemical coupling agent
    • 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
    • G01N33/56911Bacteria
    • G01N33/5695Mycobacteria
    • 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
    • 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
    • G01N33/689Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to pregnancy or the gonads
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • 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
    • G01N2333/195Assays involving biological materials from specific organisms or of a specific nature from bacteria
    • G01N2333/35Assays involving biological materials from specific organisms or of a specific nature from bacteria from Mycobacteriaceae (F)
    • 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
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • G01N2333/4701Details
    • G01N2333/471Pregnancy proteins, e.g. placenta proteins, alpha-feto-protein, pregnancy specific beta glycoprotein
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2469/00Immunoassays for the detection of microorganisms
    • G01N2469/20Detection of antibodies in sample from host which are directed against antigens from microorganisms
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/26Infectious diseases, e.g. generalised sepsis
    • 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
    • G01N33/54366Apparatus specially adapted for solid-phase testing
    • G01N33/54386Analytical elements
    • G01N33/54387Immunochromatographic test strips
    • G01N33/54388Immunochromatographic test strips based on lateral flow
    • 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
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • G01N33/582Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances with fluorescent label

Definitions

  • the present invention relates generally to multiplex immunoassays and specifically to a multiplex immunoassay (MIA) for Johne’s disease and pregnancy.
  • MIA multiplex immunoassay
  • Johne’s disease an infection caused by Mycobacterium avium paratuberculosis (MAP) is a major production limiting disease of dairy cows. There is no cure for this disease and the only way to mitigate losses is via whole herd testing for Johne’s disease and intervention. Further, the diagnosis of pregnancy is an essential component of sound reproductive management in the dairy industry, which is confirmed by the detection of pregnancy associated glycoproteins (PAGs) in milk and serum samples from cattle.
  • PAGs pregnancy associated glycoproteins
  • PAGs pregnancy related glycoproteins
  • Bovines have been found to express pregnancy related glycoproteins (PAGs) during pregnancy.
  • PAGs belong to a large family of inactive aspartic proteinases expressed by the placenta of domestic ruminants including cows, ewes, and goats.
  • the PAG gene family comprises at least 22 transcribed genes as well as some variants (Telugu et al., 2009).
  • Mean PAG concentrations in cattle increase from 15 to 35 days in gestation.
  • PAGs are produced by mono- and binucleate trophoblast cells in the ruminant placenta.
  • PAG appears in maternal blood and, from approximately 4 weeks after fertilization onward, may serve as a reliable means of diagnosing pregnancy.
  • a range of factors are said to affect plasma PAG concentrations, such as number and sex of fetus, mass of calf and placenta, level of milk production and genetic constitution.
  • the present invention is based on the seminal discovery of the use of a multiplex immunoassay for detection of Johne’s disease and pregnancy in bovines and other ruminants. Specifically, the invention provides immunoassays that simultaneously detect bovine pregnancy associated glycoproteins (PAGs) and antibodies produced in response to infection by Mycabactenuni avium subspecies paratuberculosis (MAP).
  • PAGs bovine pregnancy associated glycoproteins
  • MAP Mycabactenuni avium subspecies paratuberculosis
  • the present invention provides a substrate with at least two capture elements specific for Johne’s disease and/or pregnancy on the substrate, each capture element corresponding to and being able to bind a target analyte, the substrate further optionally has a plurality of control elements including at least one fiduciary marker, at least one negative control to monitor background signal, at least one negative control to monitor assay specificity, at least one positive colorimetric control, at least one positive control to monitor assay performance and any combination thereof.
  • the capture elements bind target analytes, wherein the target analytes are indicative of a Johne’s disease infection and/or pregnancy.
  • the target analyte is a protein, a protein fragment, a peptide, a polypeptide, a polypeptide fragment, an antibody, an antibody fragment, an antibody binding domain, an antigen, an antigen fragment, an antigenic determinant, an epitope, a hapten, an immunogen, an immunogen fragment, epitope, or any combination thereof.
  • the target analyte is a bovine pregnancy associated glycoprotein (PAG) or epitope thereof.
  • the PAG is selected from PAG-4, PAG-6, PAG-9, PAG-20, PAG-21 or any combination thereof.
  • the target analyte is a Mycobacterium avium subspecies paratuberculosis (MAP) antibody, fragment or binding domain thereof.
  • the capture element is a protein, a protein fragment, a binding protein (BP), a binding protein fragment, an antibody, an antibody fragment, an antibody heavy chain, an antibody light chain, a single chain antibody, a single-domain antibody, a Fab antibody fragment, an Fc antibody fragment, an Fv antibody fragment, a F(ab')2 antibody fragment, a Fab' antibody fragment, a single-chain Fv (scFv) antibody fragment, an antibody binding domain, an antigen, an antigenic determinant, an epitope, a hapten, an immunogen, an immunogen fragment, or any combination thereof.
  • capture element is a Mycobacterium avium subspecies paratuberculosis (MAP) antigen.
  • the capture element is a bovine pregnancy associated glycoprotein (PAG) or epitope thereof, antibody, fragment or binding domain thereof.
  • the PAG is selected from PAG-4, PAG-6, PAG-9, P AG-20, P AG-21 or any combination thereof.
  • the substrate is a solid or porous substrate.
  • the solid substrate is a paramagnetic bead, microtiter plate, microparticle, or a magnetic bead.
  • the present invention provides a kit for detecting a plurality of target analytes in a sample, including a substrate and optionally one or both of a background reducing reagent, and a colorimetric detection system.
  • the antibodies for detection comprise antibody-binding protein (BP) conjugates, antibody- enzyme label conjugates, or any combination thereof.
  • the sample is a milk or a blood sample.
  • blood sample is serum or plasma.
  • the substrate is a solid.
  • the solid substrate is a paramagnetic bead, microtiter plate, or microparticle.
  • the background reducing reagent is, Mycobacterium phlei.
  • the colorimetric detection system comprises HRP-labelled anti-PAGs Ab and HRP-labelled anti-bovine IgG Ab.
  • the secondary antibody comprises at least one bovine anti-PAG antibody.
  • the present invention provides a method for processing a microarray by providing a substrate, adding at least one sample to the substrate; and processing the substrate such that a detectable result is given by two or more of at least one fiduciary marker, at least one positive colorimetric control, and at least one positive control to monitor assay performance.
  • the present invention provides a method for detecting an analyte in a sample comprising providing a substrate, adding at least one sample to the substrate, and processing the substrate such that a detectable result is provided.
  • the detectable result includes two or more of at least one fiduciary marker, at least one positive colorimetric control, and at least one positive control to detect an analyte in the sample.
  • the present invention provides an isolated peptide having the amino acid sequence DTVRIGDLVSTDQ (SEQ ID NO: 1). In an additional embodiment, the present invention provides an isolated peptide having the amino acid sequence GSWMFGGVDHRYYKGELNW (SEQ ID NO:2). In a further embodiment, the present invention provides an isolated peptide having the amino acid sequence ITIGTPPQEFQV (SEQ ID NO:3).
  • the present invention provides an antibody that binds a peptide having the amino acid sequence DTVRIGDLVSTDQ (SEQ ID NO: 1).
  • the antibody is monoclonal.
  • the present invention provides an antibody that binds a peptide having the amino acid sequence GSVVMFGGVDHRYYKGELNW (SEQ ID NO:2).
  • the antibody is monoclonal.
  • the present provides an antibody that binds a peptide having the amino acid sequence ITIGTPPQEFQV (SEQ ID NO:3).
  • the antibody is monoclonal.
  • FIG. 1 shows assay formats used for the detection of anti-MAP antibody and PAGs in the membrane-free multiplex immunoassay (MIA) for the diagnosis of Johne’s disease and pregnancy, respectively.
  • the anti-MAP Ab in Mycobacterium paratuberculosis infected cow’ s milk sample in Johne’s disease are detected via an indirect ELISA (left) while PAGs in pregnant cow’s milk sample are detected by a sandwich ELISA (right).
  • Figures 2A-E show a membrane-free MIA procedure.
  • Fig 2A Printing of MAP Ag and anti-PAGs Ab on the assay surface.
  • Fig. 2B Blocking of assay surface after protein/ Ab printing.
  • Detection of target analytes in the cow’s milk sample.
  • Fig. 2D Detection of specifically bound analytes by binding with HRP-labelled detection Ab against the analytes.
  • Fig. 2E Generation of colorimetric array spots by the addition of HRP substrate (TMB).
  • Figures 3 shows a membrane-free MIA.
  • the desired biomarkers i.e. MAP Ag and anti- PAGs Ab, are printed as duplicate spots in each well of detachable 8-wells modules of a 96-well MTP.
  • FIG 4 shows leach-proof immobilization of biomolecules (antibody (Ab) or antigen (Ag)) to the solid substrate of MTP’s well using 3-aminopropyltriethoxysilane (APTES).
  • biomolecules antibody (Ab) or antigen (Ag)
  • APTES 3-aminopropyltriethoxysilane
  • FIG. 5 shows the steps involved in the MIA.
  • the initial step involves the addition of the sample and the detection of target analytes (anti-MAP Abs and PAGs) within it.
  • anti-PAG secondary antibody is added forming the sandwich reaction.
  • HRP-labeled detection antibody is added, which binds to the bovine IgG of both assays.
  • TMB HRP substrate
  • Figure 6 shows the capture Ab against PAG printed in duplicate in each well of a strip while MAP antigen is printed in duplicate in each well of another strip. The detection addition and other internal control spots are printed in all the wells. The white circles signify that nothing has been printed at that specific position.
  • Figures 7A-B show alignments of bovine pregnancy associate glycoproteins (PAGs) and a surface model of PAG-20.
  • Fig. 7A Sequence alignment of PAG-20, PAG-21, PAG-9, PAG- 6, and PAG-4. The three antigenic peptides that are common to all PAGs are highlighted. Three of the four antigenic sites, framed on squares, are the sequences selected to synthesize the peptides required for the Phage display technology based animal-free production of anti-PAGs Ab.
  • Fig. 7B An example of surface model of PAG-20 and its antigenic sites.
  • the present invention is based on the seminal discovery of the use of a multiplex immunoassay for detection of Johne’s disease and pregnancy in bovines and other ruminants. Specifically, the invention provides immunoassays that simultaneously detect bovine pregnancy associated glycoproteins (PAGs) and antibodies produced in response to infection by Mycobacterium avium subspecies paratuberculosis (MAP).
  • PAGs bovine pregnancy associated glycoproteins
  • MAP Mycobacterium avium subspecies paratuberculosis
  • Johne’s disease an infection caused by Mycobacterium avium paratuberculosis (MAP), is a major production limiting disease of dairy cows. There is no cure for this disease and the only way to mitigate losses is via whole herd testing for Johne's disease and intervention. Further, the diagnosis of pregnancy is an essential component of sound reproductive management in the dairy industry, which is confirmed by the detection of pregnancy associated glycoproteins (PAGs) in milk and serum samples from cattle. Johne’s disease and pregnancy diagnosis are currently performed during the same season by two separate ELISAs using milk samples from cattle.
  • the present invention provides a multiplex immunoassay (MIA) procedure that detects both the antigens (Ags) and antibodies (Abs) in a single MIA.
  • MIA multiplex immunoassay
  • the anti-PAGs Abs of the invention are custom-produced via an animal-free antibody production technology using the custom- synthesized multiple-epitope bearing PAGs peptides.
  • Three epitopes have been identified that are present in PAG-4, PAG-6, PAG-9, PAG-20, and PAG-21 and thus, enable the early diagnosis of pregnancy in cattle.
  • the invention describes the multiplex immunoassay (MIA) procedures for the simultaneous detection of multiple analytes for the diagnosis of Johne’s disease and pregnancy using the milk samples from cattle and other farm animals. It involves the detection of two distinct bovine analytes, PAGs and IgG antibodies (Abs) towards MAP, in milk or serum via a multiplex immunoassay.
  • MIA multiplex immunoassay
  • the present invention provides a substrate with at least two capture elements specific for Johne’s disease on the substrate, each capture element corresponding to and being able to bind a target analyte, the substrate further optionally has a plurality of control elements including at least one fiduciary marker, at least one negative control to monitor background signal, at least one negative control to monitor assay specificity, at least one positive colorimetric control, at least one positive control to monitor assay performance and any combination thereof.
  • the capture elements bind target analytes, wherein the target analytes are indicative of a Johne’s disease infection.
  • the target analyte is a protein, a protein fragment, a peptide, a polypeptide, a polypeptide fragment, an antibody, an antibody fragment, an antibody binding domain, an antigen, an antigen fragment, an antigenic determinant, an epitope, a hapten, an immunogen, an immunogen fragment, epitope, or any combination thereof.
  • the target analyte is a bovine pregnancy associated glycoprotein (PAG) or epitope thereof.
  • the PAG is selected from PAG-4, PAG- 6, PAG-9, PAG-20, PAG-21 or any combination thereof.
  • the target analyte is a Mycobacterium avium subspecies paratuberculosis (MAP) antibody, fragment or binding domain thereof.
  • the capture element is a protein, a protein fragment, a binding protein (BP), a binding protein fragment, an antibody, an antibody fragment, an antibody heavy chain, an antibody light chain, a single chain antibody, a single-domain antibody, a Fab antibody fragment, an Fc antibody fragment, an Fv antibody fragment, a F(ab')2 antibody fragment, a Fab' antibody fragment, a single-chain Fv (scFv) antibody fragment, an antibody binding domain, an antigen, an antigenic determinant, an epitope, a hapten, an immunogen, an immunogen fragment, or any combination thereof.
  • capture element is a Mycobacterium avium subspecies parattihecculosis (MAP) antigen or epitope thereof.
  • the capture element is a bovine pregnancy associated glycoprotein (PAG) antibody, fragment or binding domain thereof.
  • the PAG is selected from PAG-4, PAG-6, PAG-9, PAG-20, PAG-21 or any combination thereof.
  • the substrate is a solid or porous substrate.
  • the solid substrate is a paramagnetic bead, microtiter plate, microparticle, or a magnetic bead.
  • substrate is any surface that supports an immunoassay.
  • the substrate of the invention may be a solid substrate or a porous substrate, for example.
  • the substrate is a solid substrate.
  • solid substrates include, but are not limited to, 96 well microtiter plate, glass, microbeads, nano/micro- particles and magnetic beads.
  • a 96 well microtiter plate is polystyrene, polydimethylsiloxane (PDMS), poly (methyl methacrylate) (PMMA), polycarbonate, cyclic polyolefins, Zeonor, Zeonex, or cellulose acetate.
  • the solid substrate maybe glass beads, nano- /microparticles, magnetic beads or paramagnetic beads.
  • the assay elements are placed on the substrate surface, with or without an adapter molecule between the substrate and the element.
  • the assay elements bind to the substrate by covalent or non-covalent interaction.
  • methods of placing assay elements on the substrate include printing, spotting or other techniques known in the art.
  • printing can be used to include any of the methods for placing the assay elements on a membrane.
  • array or “microarray” as used herein refer to a collection of multiple assay elements on a substrate. Specifically, an array is a collection of capture elements and/or control elements on a substrate.
  • the elements on the array are placed on the substrate in discrete areas of between 100 pm to 500 pm in diameter. More preferably, the discrete areas are between 350pm to 400pm in diameter. In certain aspects, the discrete areas of the array are placed in a 5x5 grid. In one aspect, the array comprises up to nine control elements and two replicates of each of eight different capture elements. In one aspect, the capture elements are printed in two or more replicates of four different capture elements and multiples thereof. [0037] As used herein, the term “assay element” refers to any of a number of different elements for use in an array of the invention. Exemplary assay elements include, but are not limited to, capture elements and control elements.
  • capture element refers to a molecule that is able to bind to a target analyte.
  • useful capture elements include proteins, protein fragments, polypeptides, polypeptide fragments, binding proteins, binding protein fragments, antibodies (polyclonal, monoclonal, or chimeric), antibody fragments, antibody heavy chains, antibody light chains, single chain antibodies, single-domain antibodies (a VHH for example), Fab antibody fragments, Fc antibody fragments, Fv antibody fragments, F(ab')2 antibody fragments, Fab' antibody fragments, single-chain Fv (scFv) antibody fragments, antibody binding domains, antigens, antigenic determinants, epitopes, haptens, immunogens, immunogen fragments, and binding domains.
  • Useful capture elements will correspond to and be able to bind a specific target analyte, such as a molecule or class of molecules that are present in a sample to be tested.
  • the capture element is selected from a protein, a protein fragment, a binding protein, a binding protein fragment, an antibody, an antibody fragment, an antibody heavy chain, an antibody light chain, a single chain antibody, a single-domain antibody (a VHH for example), a Fab antibody fragment, an Fc antibody fragment, an Fv antibody fragment, a F(ab')2 antibody fragment, a Fab' antibody fragment, a single-chain Fv (scFv) antibody fragment, an antibody binding domain, an antigen, an antigenic determinant, an epitope, a hapten, an immunogen, an immunogen fragment, and a binding domain.
  • the capture elements may comprise antibodies or fragments thereof that are immobilized on the substrate surface and are specific for different antigens or ligands that may be present in a sample.
  • the capture elements may comprise antigens or ligands and the assay involves the detection of specific antibodies that may be present in a sample.
  • the capture elements may comprise of a receptor or a subunit of a receptor that binds a specific ligand.
  • the capture element can be Mycobacterium avium subspecies paratuberculosis (MAP) antigen or epitope thereof or antibodies to PAGs including PAG-4, PAG- 6, PAG-9, PAG-20, P AG-21 or any combination thereof.
  • MAP Mycobacterium avium subspecies paratuberculosis
  • biomarker refers to any substance used as an indicator of a biologic state.
  • a biomarker can be any substance whose detection indicates a particular disease state (for example, the presence of an antibody may indicate an infection).
  • a biomarker can be indicative of a change in expression or state of a protein that correlates with the risk or progression of a disease, or with the susceptibility of the disease to a given treatment.
  • a biomarker may be used as a surrogate for a natural endpoint such as survival or irreversible morbidity. If a treatment alters the biomarker, which has a direct connection to improved health, the biomarker serves as a “surrogate endpoint” for evaluating clinical benefit.
  • the target analyte is a biomarker.
  • the target analyte is selected from a protein, a protein fragment, a peptide, a polypeptide, a polypeptide fragment, an antibody, an antibody fragment, an antibody binding domain, an antigen, an antigen fragment, an antigenic determinant, an epitope, a hapten, an immunogen, an immunogen fragment, or any combination of any two or more thereof.
  • the target analyte is PAG-4, PAG-6, PAG-9, PAG-20, PAG-21 or any combination thereof.
  • the target analyte may also be a MAP antibody, antibody fragment or binding domain thereof.
  • Capture elements specific for a target analyte are used to detect the presence or absence of the analyte in a sample.
  • a wide range of complementary binding or coupling partners are known, with the choice of capture elements determined by the analytes to be detected, the requirement for adapter molecules and the level of specificity required for the assay.
  • the capture elements are specific for binding/ detecting PAGs (e.g. PAG-4, PAG-6, PAG-9, PAG-20, PAG-21 or any combination thereof.) and IgG antibodies produced by a Johne’s disease infection (e.g. antibodies to Mycobacterium avium subspeciesparatuberculosis (MAP)).
  • MAP Mycobacterium avium subspeciesparatuberculosis
  • control element refers to an element that is used to provide information on the function of the assay, for example binding specificity, the level of non-specific background binding, the degree of binding cross-reactivity, and the performance of assay reagents and the detection system.
  • Preferred controls useful herein include at least one negative control to monitor background signal, at least one negative control to monitor assay specificity, at least one positive colorimetric control, and at least one positive control to monitor assay performance.
  • the substrate of the invention comprises at least one fiduciary marker that will always be detectable on the substrate, preferably detectable irrespective of the performance of the assay or processing of the substrate.
  • fiduciary marker refers to a colored marker or label that will always be detectable on the substrate, preferably irrespective of the performance of the assay or processing of the substrate.
  • the use of at least one fiduciary marker will obviate the necessity of this element being detected based on successful array processing, in comparison to the positive colorimetric controls.
  • the fiduciary marker is therefore a “true” positive control that would always be detectable regardless of array processing, and can be used to orient and help to grid the array.
  • the fiduciary marker is a dye, dye-conjugated protein or a chromogenic protein such as hemoglobin.
  • the term “negative control” refers to an element comprising print buffer or an unrelated protein to which no complementary binding partner is intended to be present in the assay. Any detectable signal from the negative control can be used to determine the background threshold of the assay and the accuracy of any positive results.
  • the negative control to monitor background signal is print buffer.
  • the print buffer is a solution used to carry and print the capture elements and control elements onto the substrate and may comprise buffered saline, APTES, glycerol and a surfactant, preferably a polysorbate surfactant such as Tween 20.
  • the blocking solution is used to reduce non-specific protein binding to the substrate surface and preferably comprises skim milk, casein, bovine serum albumin, gelatins from fish, pigs or other species, dextran or any mixture of any two or more thereof, preferably in a solution of phosphate buffered saline and a surfactant such as Tween 20.
  • control capture element refers to a capture element that functions as a control, either a negative control that should not bind any analyte or a positive control that will bind a non-target analyte.
  • the substrate of the invention also comprises at least one control to monitor assay performance.
  • the control is intended to provide information of the efficiency of the complementary binding interactions or the quality or performance of the reagents used.
  • control to monitor assay performance refers to an element that forms one part of a complementary binding interaction during an assay and is intended to provide information on the accuracy of the assay result.
  • the positive control to monitor assay performance comprises one binding partner of a complementary binding pair, where the other binding partner is a sample component or an assay reagent.
  • the assay performance control is preferably selected from a target analyte, a binding partner corresponding to and able to bind a non-target analyte that will be present in the sample, a binding partner corresponding to and able to bind an assay reagent, and a colorimetric enzyme label, or any combination of any two or more thereof.
  • binding partner corresponding to and able to bind a non-target analyte that will be present in the sample is an anti-Ig antibody that will bind an immunoglobulin present in a serum sample, therefore confirming a sample has been added.
  • An example of a binding partner corresponding to and able to bind an assay reagent is an anti-Ig antibody that will bind a secondary immunoglobulin that is used to process the assay, such as biotinylated anti-target analyte antibody.
  • Another example of a binding partner corresponding to and able to bind an assay reagent is a biotinylated antibody that will bind a streptavidin-per oxidase conjugate that is used to process the assay.
  • the assay performance control comprises one binding partner of a complementary binding pair, wherein the other binding partner is an assay reagent.
  • the assay performance control is preferably selected from the list comprising the target analyte, a nonspecific binding partner or a colorimetric enzyme label.
  • the complementary binding partners comprise antibody-antigen interactions or antibody-ligand interactions.
  • the substrate of the invention also comprises at least one control to monitor assay specificity.
  • the control is intended to provide information of the specificity of binding between the capture element and the target analyte, or between the binding partners of the assay detection steps.
  • control to monitor assay specificity refers to an element that is closely related to at least one binding partner of a complementary binding pair present in the assay and is intended to provide information of the specificity of the complementary binding.
  • This control is a negative control that is not expected to generate a detectable result during normal assay processing.
  • the assay specificity control would comprise an antibody that should not bind any antigen in the sample.
  • the assay specificity control would comprise an antigen that should not bind any antibody in the sample.
  • the assay specificity control is Mycobacterium phlei protein extract.
  • the assay specificity control Mycobacterium tuberculosis antigen.
  • positive colorimetric control refers to an enzyme or enzyme conjugate that provides a detectable signal upon addition of the enzyme substrate.
  • the positive colorimetric control is an enzyme label conjugate capable of reacting with a colorimetric substrate, comprising an enzyme selected from the list comprising horseradish peroxidase, alkaline phosphatases, P-D-galactosidase or glucose oxidase.
  • an enzyme selected from the list comprising horseradish peroxidase, alkaline phosphatases, P-D-galactosidase or glucose oxidase.
  • the identity of the assay controls will be dependent on the type of array, the identity of the target analyte, and the type of sample to be analyzed.
  • anti-bovine IgG-HRP or specific monoclonal IgG-HRP may be used in arrays printed with antigens and antibodies, respectively.
  • the final detection antibody in antigen arrays will often be anti-bovine IgG-HRP, while for antibody arrays it will often be a HRP conjugated IgG antibody specific for the targeted analyte or a bovine IgG specific for the target analyte that is detected later on the next step with anti-bovine IgG-HRP.
  • HRP conjugated IgG antibody specific for the targeted analyte or a bovine IgG specific for the target analyte that is detected later on the next step with anti-bovine IgG-HRP.
  • the target protein or antigen and mouse IgG, bovine IgG and anti-bovine IgG present on antigen or antibody arrays can act either as positive or negative controls depending on the array format, in addition to providing information of assay specificity.
  • antigen or mouse IgG spots should provide the positive signal in antibody arrays, while the latter two should provide a positive signal in antigen arrays.
  • These controls can also serve as controls for overall assay performance.
  • sample and “specimen” as used herein are used in their broadest sense to include any composition that is obtained and/or derived from biological or environmental source, as well as sampling devices (e.g., swabs) which are brought into contact with biological or environmental samples.
  • Biological samples include body fluids such as milk, urine, blood, plasma, fecal matter, cerebrospinal fluid (CSF), and saliva.
  • the biological sample is fluid obtained from a mammal, including milk, blood, plasma, serum, and stool. These examples are illustrative, and are not to be construed as limiting the sample types applicable to the present invention.
  • the sample is a milk or blood sample, including a plasma or serum sample.
  • the assay techniques used in conjunction with the substrates of the present invention include any of a number of well-known colorimetric enzyme-linked assays. Examples of such systems are well known in the art.
  • the assay techniques are based upon the formation of a complex between a complementary binding pair, followed by detection with a colorimetric detection system comprising an enzyme-conjugate label and a colorimetric substrate.
  • the detection system will be described with reference to enzyme-linked immunosorbent assays (ELISA), though a skilled person would appreciate that such techniques are not restricted to the use of antibodies but are equally applicable to any colorimetric assay.
  • the ELISA is in the “sandwich” assay format. In this format, the target analyte to be measured is bound between two antibodies — the capture antibody and the detection antibody. In another embodiment, the ELISA is a non-competitive assay, in which an antibody binds to the capture antigen and the amount of bound antibody is determined by a secondary detection antibody.
  • Either monoclonal or polyclonal antibodies may be used as the capture and detection antibodies in sandwich ELISA systems.
  • Monoclonal antibodies have an inherent monospecificity toward a single epitope that allows fine detection and quantitation of small differences in antigen.
  • a polyclonal antibody can also be used as the capture antibody to bind as much of the antigen as possible, followed by the use of a monoclonal antibody as the detecting antibody in the sandwich assay to provide improved specificity.
  • a monoclonal antibody can also be used as the capture antibody to provide specific analyte capture, followed by the use of a polyclonal antibody as the detection antibody in the sandwich assay. Additionally, both the capture and the detection antibodies could be monoclonal.
  • antibody as used herein includes naturally occurring antibodies as well as non-naturally occurring antibodies, including, for example, single chain antibodies, chimeric, bifunctional and humanized antibodies, as well as antigen-binding fragments thereof.
  • non-naturally occurring antibodies can be constructed using solid phase peptide synthesis, can be produced recombinantly or can be obtained, for example, by screening combinatorial libraries consisting of variable heavy chains and variable light chains (see Huse et al., Science 246: 1275- 1281, 1989, which is incorporated herein by reference).
  • These and other methods of making, for example, chimeric, humanized, CDR-grafted, single chain, and bifunctional antibodies are well known (Winter and Harris, Immunol.
  • modified or derivatized antibodies, or antigen binding fragments of antibodies can be useful for the present methods.
  • pegylated antibodies can be useful for the present methods.
  • Fab, F(ab')2, Fd and Fv fragments of an antibody that retain specific binding activity are included within the definition of an antibody.
  • second antibody refers to an antibody that will bind a target analyte and that is conjugated with either an adaptor molecule such as biotin or an enzyme label such as horseradish peroxidase (HRP).
  • Antibody-adaptor conjugates are processed to give a detectable result by contacting the antibody-adaptor conjugate with an adaptor- enzyme conjugate and then the enzyme substrate; for example, antibody-biotin conjugates will bind streptavidin-HRP conjugates.
  • Antibody-enzyme label conjugates include antibody-HRP conjugates.
  • binding specifically or “specific binding activity” or the like, means that two molecules form a complex that is relatively stable under physiologic conditions.
  • an antigen-binding domain is specific for a particular epitope, which is carried by a number of antigens, in which case the antibody carrying the antigen-binding domain will be able to bind to the various antigens carrying the epitope.
  • Specific binding is characterized by a high affinity and a low to moderate capacity.
  • the binding is considered specific when the affinity constant is about U I CF 6 M, generally at least about U 1 CT 7 M, usually at least about UK) -8 M, and particularly at least about 1 xlO -9 M or less.
  • Blockers serve to decrease or at best eliminate nonspecific protein binding from the sample on the substrate surface, thereby decreasing overall background signal. This increases the ratio of signal to noise, thereby increasing the overall sensitivity of the assay. Blockers play no active part in the subsequent reactions between the sample and other assay reagents and the immobilized proteins on the substrate.
  • blockers include, but are not limited to, bovine serum albumin, casein, non-fat dry milk, gelatin derived from fish, pigs and other sources, dextran, serum derived from sources other than the sample being analyzed such as from steelhead salmon, guinea pigs, hamsters, rabbit and other sources, polyethylene glycol, polyvinyl pyrrollidone, and commercial preparations including HeteroBlock (Omega Biologicals, Bozeman, Mont.), SuperBlock, StartingBlock, SEA BLOCK (Pierce, Rockford, Ill.).
  • blockers are made up in buffer solutions such as, for example, phosphate buffer, phosphate buffered saline, Tris buffer, acetate buffer and others.
  • the blockers may also be supplemented with detergents such as, for example, Tween 20, Tween 80, Nonidet P40, sodium dodecyl sulfate and others.
  • the enzyme-conjugate label comprising an enzyme selected from the list comprising horseradish peroxidase, alkaline phosphatase, P-D-galactosidase or glucose oxidase.
  • the enzyme label may be conjugated directly to a primary antibody or introduced through a secondary antibody that recognizes the primary antibody. It may also be conjugated to a protein such as streptavidin if the primary antibody is biotin labelled.
  • the assay detection system comprises a detection colorimetric substrate selected from the list comprising 3,3', 5,5'-tetramethylbenzidine, diaminobenzidine, metal-enhanced diaminobenzidine, 4-chl oro-1 -naphthol, colloidal gold, nitro-blue tetrazolium chloride, 5-bromo-4-chloro-3'-indolylphosphate p-toluidine salt and naphthol AS-MX phosphate+Fast Red TR Salt.
  • a detection colorimetric substrate selected from the list comprising 3,3', 5,5'-tetramethylbenzidine, diaminobenzidine, metal-enhanced diaminobenzidine, 4-chl oro-1 -naphthol, colloidal gold, nitro-blue tetrazolium chloride, 5-bromo-4-chloro-3'-indolylphosphate p-toluidine salt and naphthol AS-MX
  • the colorimetric reaction can be detected and optionally quantified and analyzed using an image capture device such as a digital camera or a desktop scanner attached to a computer.
  • an image capture device such as a digital camera or a desktop scanner attached to a computer.
  • concentration values of known standard elements can be used to generate standard curves.
  • Concentration values for unknown analytes can be analyzed using the standard curve for each analyte to calculate actual concentrations. Values for each analyte can be identified based on the spotting position of each capture element within the array.
  • the present invention provides methods of in vitro diagnostic applications for the detection of Johne’s disease or pregnancy such as manual multiplex immune assay, automated multiplex immune assay (MIA), Automated MIA (e.g., PictArrayTM, U.S. Patent No.
  • MIA automated multiplex immune assay
  • MIA Automated MIA
  • PictArrayTM U.S. Patent No.
  • LFIA multiple lateral flow immunoassay
  • CLIA automated chemiluminescent immune assay
  • CLIA wash-free immune assays (manual and automated), automated centrifugal microfluidics-based immunoassay, lab-on-a-chip based immunoassay, paramagnetic bead-based manual ELISA, point-of-care (POC) immunoassays, smart phone based immunoassay and other immunoassay formats.
  • detection is by colorimetric imaging (e.g., PictArrayTM, U.S. Patent No.
  • absorbance e.g., manual ELISA
  • chemiluminescence e.g., automated CLIA, CRET
  • florescence e.g., manual immunoassays, ELISA, FRET
  • naked eye e.g., lateral flow immunoassays
  • MIA manual multiplex immune assay
  • the PAGs are detected by sandwich immunoassay (IA) while anti-MAP antibodies (Abs) are determined via an indirect IA.
  • IA sandwich immunoassay
  • Abs anti-MAP antibodies
  • Both the analytes would be simultaneously detected in the same well for the diagnosis of Johne’s disease and pregnancy a membrane-free MIA procedure will be used for the printing of microarray spots (e.g., PictArrayTM, U.S. Patent No. 9,625,453) directly on the solid surface of 96-well microtiter plate (MTP).
  • MTP microtiter plate
  • biorecognition elements responsible for the binding of PAGs and anti-MAP Abs will be printed onto the surface of the MTP well using a leach-proof biomolecular immobilization procedure that involves the addition of 1% aminopropyltri ethoxy silane (APTES) in the printing buffer.
  • APTES aminopropyltri ethoxy silane
  • the automated MIA automates all the steps in the manual MIA and employs an integrated colorimetric reader and image analysis software.
  • the automated IAS are ideal for high- throughput tests for Johne’s disease and pregnancy in cattle.
  • LFIA multiplex lateral flow immunoassay
  • biomarkers are immobilized in two different lines on a nitrocellulose membrane strip.
  • the results from this assay can be interpreted by naked eye.
  • the rapid LFIA tests could be performed using previously described MIA formats including manual and automated MIAs.
  • the biorecognition elements responsible for the detection of PAGs and anti-MAP Abs would be immobilized in two control lines on a nitrocellulose membrane strip.
  • Manual singleplex ELISA could also be used to detect Johne’s disease and pregnancy and would use custom-synthesized anti -PAG Abs that would be specific for various PAGs (e.g. PAG-4, PAG-6, PAG-9, PAG-20, PAG-21).
  • Automated multiplex and singleplex CLIAs can also be developed for the diagnosis of Johne’s disease and pregnancy in cattle.
  • the MAP and anti -PAG Ab could be bound covalently to paramagnetic beads (micron-sub-micron size) and used for the detection of anti-MAP Ab and PAGs in sample.
  • Various formulations of paramagnetic beads could be coated with MAP and anti- PAG Ab and then mixed together for MIA.
  • the detection signal in case of automated CLIAs could be generated by conjugating the detection antibody with acridinium or other chemiluminescent labels and providing the appropriate trigger solutions for the generation of chemiluminescent signal.
  • the centrifugal microfluidics-based automated immunoassay could also be performed to detect Johne’s disease and pregnancy.
  • the MAP can be covalently bound to paramagnetic beads and used for the detection of anti-MAP antibodies.
  • the capture antibodies for PAGs can be coated covalently to the paramagnetic beads and used for the detection of PAGs via sandwich IA.
  • the magnetic beads are transferred from one chamber to another in the apparatus by a magnet.
  • the detection of analyte occurs in a reaction chamber, which is followed by washing the specific immune complexes formed on paramagnetic beads and their transfer to the detection chamber for the generation and reading of assay signal.
  • the assay signal most commonly used in is the chemiluminescent, however, absorbance and fluorescence could also be used. All the microfluidic operations could be performed in a compact benchtop centrifugal microfluidic analyzer.
  • a lab-on-a-chip (LOC)-based immunoassay (IA) could also be developed that could use MIA procedures similar to those previously described.
  • LOC-based IAS could use paramagnetic beads or solid surfaces for the covalent attachment of MAP and anti-PAGs Ab.
  • the detection signal could be chemiluminescent, fluorescent, absorbance, electrochemical or colorimetric.
  • Manual singleplex ELISA can be used to detect Johne’s disease and pregnancy, employing paramagnetic beads that could be bound covalently to MAP or anti-PAGs Ab.
  • a point of care (POC) IA such as that based on electrochemical detection using a disposable strip, can also be developed.
  • the IA would be a label-free immunoassay using an electrochemical reader or a smartphone-based reader.
  • Various smartphone-readout based IAs could also be developed where an optomechanical attachment would be developed enabling the readout of colorimetric detection signal via the smartphone back camera. Therefore, the smartphone will act as the POC colorimetric readout device.
  • the invented assay formats for the detection of Johne’s disease and pregnancy diagnosis could also be used for the development of novel and emerging IA formats, which are based on advances in complementary technologies.
  • Some examples of such prospective IA formats are wash-free IA based on FRET (fluorescence resonance energy transfer) or CRET (chemiluminescence resonance energy transfer); signal- enhanced IA based on the use of nanoparticle-based signal detection step or the use of micro- and subsmicro- beads for binding capture antibodies/antigens; rapid multiplex IAs based on Lab-in-a- tube technology, and vertical microfluidics.
  • kits for the detection of target analytes are particularly amenable to use in kits for the detection of target analytes.
  • kits may comprise the substrates together with instructions and any assay consumables required.
  • Different kits are envisaged for different target analytes and types of array.
  • the present invention provides a kit for detecting a plurality of target analytes in a sample, including a substrate and optionally one or both of a background reducing reagent, and a colorimetric detection system.
  • the kit further has one or more items selected from a wash solution, one or more antibodies for detection of antigens, ligands or antibodies bound to the capture elements or for detection of the positive controls, software for analyzing captured target analytes, a protocol for measuring the presence of target analytes in samples, a sample diluent, blotting TMB (3,3',5,5’-tetramethylbenzidine; MW :::: 240.4) and a secondary antibody.
  • the antibodies for detection comprise antibody-binding protein (BP) conjugates, antibody- enzyme label conjugates, or any combination thereof.
  • the sample is a milk or a blood sample.
  • blood sample is serum or plasma.
  • the substrate is a solid or porous substrate.
  • the solid substrate is a paramagnetic bead, microtiter plate, or microparticle.
  • the background reducing reagent is Mycobacterium phlei protein extract.
  • the colorimetric detection system comprises HRP-labelled anti-PAGs Ab and HRP-labelled anti- bovine IgG Ab.
  • the secondary antibody comprises at least one bovine anti-PAG antibody.
  • the invention also relates to a method of processing a substrate of the invention.
  • a method of processing a substrate of the invention comprises providing a substrate of the invention as described above, adding at least one sample to the substrate, and processing the substrate such that a detectable result is given by two or more of at least one fiduciary marker, at least one positive colorimetric control, and at least one positive control to monitor assay performance.
  • the present invention provides methods for processing a microarray by providing a substrate, adding at least one sample to the substrate, and processing the substrate such that a detectable result is given by two or more of at least one fiduciary marker, at least one positive colorimetric control, and at least one positive control to monitor assay performance.
  • the step of processing the substrate or microarray comprises a blocking step during which available protein-binding sites on the substrate or microarray are blocked with a blocker, an optional wash step, contacting the substrate or microarray with the sample containing the one or more analytes to be measured, a wash step to remove non-bound material from the substrate or microarray, contacting the substrate or microarray with one or more secondary antibodies that correspond to and will bind one or more target analytes and non-target analyte that is bound to an assay performance control, a wash step, and contacting the substrate or microarray with one or both of an enzyme conjugate or an enzyme substrate to generate a detectable result.
  • the present invention provides methods for detecting an analyte in a sample comprising providing a substrate, adding at least one sample to the substrate, and processing the substrate such that a detectable result is provided.
  • the detectable result includes two or more of at least one fiduciary marker, at least one positive colorimetric control, and at least one positive control to detect an analyte in the sample.
  • the substrate of the invention can be used for the simultaneous detection of at least one target analyte in a sample, and preferably a plurality of different target analytes in a sample, and have utility in diagnostic and screening assays.
  • the substrates of the invention provide the advantage that they can be adapted to high throughput (or ultra high throughput) analysis and, therefore, any number of samples (e.g., 96, 1024, 10,000, 100,000, or more) can be examined in parallel, depending on the particular support used.
  • a particular advantage of adapting the substrates to high throughput analysis is that an automated system can be used for adding or removing reagents from one or more of the samples at various times, for adding different reagents to particular samples, or for subjecting the samples to various heating cycles.
  • the automated system may consist of one or more temperature-controlled chambers and one or more robotic arms mounted on a deck that has platforms configured to hold 96-well plates.
  • the movement of the robotic arms and the temperature in the chambers are controlled by a central computer unit.
  • the array plates are stacked on the deck of the instrument.
  • the plates containing samples to be analyzed are stacked in a chamber with temperature of 4°C.
  • One robotic arm then sequentially transfers each individual array plate on one platform while the other arm sequentially transfers each individual sample plate on the second platform.
  • a nozzle containing 96 disposable tips then aspirates a predetermined volume of sample from each well of the sample plate and transfers the sample to the corresponding wells of the array plate.
  • the array plate containing the sample is then transferred to a chamber with temperature of 37°C. This process is repeated until sample has been added to all the array plates stacked on the deck.
  • the array plates are incubated for a predetermined time followed by transfer of each plate to the platform for addition of wash buffer with the nozzle containing 96 disposable tips.
  • the wash buffer is aspirated after a predetermined time and this wash process is repeated multiple (i.e., two or more) times.
  • Each array plate then receives the secondary antibody followed by transfer to a chamber with temperature of 37°C.
  • the array plates are incubated for a predetermined time followed by transfer of each plate to the platform for addition of wash buffer with the nozzle containing 96 disposable tips.
  • the wash buffer is aspirated after a predetermined time and this wash process is repeated multiple (i.e., two or more) times. Each array plate then receives the detection reagent followed by incubation for a predetermined time followed by transfer of each plate to the platform for addition of wash buffer with the nozzle containing 96 disposable tips.
  • the wash buffer is aspirated after a predetermined time and the plate transferred to the 37° C chamber for drying. The plates are transferred back to the deck after a predetermined period and manually processed for analyses of data.
  • such high throughput assays provide a means for examining duplicate, triplicate, or more aliquots of a single sample, thus increasing the validity of the results obtained, and for examining control samples under the same conditions as the test samples, thus providing an internal standard for comparing results from different assays.
  • the present invention provides a method for detecting an analyte in a sample comprising providing a substrate, adding at least one sample to the substrate, and processing the substrate such that a detectable result is provided.
  • the detectable result includes two or more of at least one fiduciary marker, at least one positive colorimetric control, and at least one positive control to detect an analyte in the sample.
  • the present invention also provides for antibodies to PAGs.
  • the antibodies bind to antigenic sites common to all PAGs.
  • the present invention provides an isolated peptide having the amino acid sequence DTVRIGDLVSTDQ (SEQ ID NO: 1). In an additional embodiment, the present invention provides an isolated peptide having the amino acid sequence GSWMFGGVDHRYYKGELNW (SEQ ID NO:2). In a further embodiment, the present invention provides an isolated peptide having the amino acid sequence ITIGTPPQEFQV (SEQ ID NO:3).
  • the present invention provides an antibody that binds a peptide having the amino acid sequence DTVRIGDLVSTDQ (SEQ ID NO: 1).
  • the antibody is monoclonal.
  • the present invention provides an antibody that binds a peptide having the amino acid sequence GSVVMFGGVDHRYYKGELNW (SEQ ID NO:2). In one aspect, the antibody is monoclonal. [0105] In a further embodiment, the present provides an antibody that binds a peptide having the amino acid sequence ITIGTPPQEFQV (SEQ ID NO:3). In one aspect, the antibody is monoclonal.
  • antibody refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that immunospecifically binds an antigen.
  • “Native antibodies” and “intact immunoglobulins”, or the like, are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. The light chains from any vertebrate species can be assigned to one of two clearly distinct types, called kappa (K) and lambda (X), based on the amino acid sequences of their constant domains.
  • immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgA, and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of immunoglobulins are called a, 5, s, y, and p, respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • antibodies can be cleaved with the proteolytic enzyme papain, which causes each of the heavy chains to break, producing three separate antibody fragments.
  • the two units that consist of a light chain and a fragment of the heavy chain approximately equal in mass to the light chain are called the Fab fragments (i.e., the "antigen binding" fragments).
  • the third unit, consisting of two equal segments of the heavy chain, is called the Fc fragment.
  • the Fc fragment is typically not involved in antigen-antibody binding, but is important in later processes involved in ridding the body of the antigen.
  • “Fv” is the minimum antibody fragment which contains a complete antigen-recognition and -binding site.
  • This region consists of a dimer of one heavy- and one light-chain variable domain in tight, non-covalent association. It is in this configuration that the three CDRs of each variable domain interact to define an antigen-binding site on the surface of the VH-VL dimer. Collectively, the six CDRs confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site. “Single-chain Fv” or “scFv” antibody fragments comprise the VH and VL domains of antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the scFv to form the desired structure for antigen binding.
  • a polypeptide linker between the VH and VL domains which enables the scFv to form the desired structure for antigen binding.
  • an “antigen” covers any substance that will elicit an immune response.
  • an “antigen” relates to any substance, preferably a peptide or protein, that reacts specifically with antibodies.
  • the term “antigen” comprises any molecule which comprises at least one epitope.
  • an antigen in the context of the present invention is a molecule which, optionally after processing, induces an immune reaction.
  • any suitable antigen may be used, which is a candidate for an immune reaction, wherein the immune reaction is preferably a cellular immune reaction.
  • An antigen is preferably a product which corresponds to or is derived from a naturally occurring antigen.
  • the membrane-free MIA format involves the simultaneous detection of anti-MAP Ab and desired PAGs (i.e., PAG-4, PAG-6, PAG-9, PAG-20, PAG-21) in a milk sample, which act as the diagnostic biomarkers of Johne’s disease and pregnancy in cattle.
  • PAG-4, PAG-6, PAG-9, PAG-20, PAG-21 desired PAGs
  • the MIA is a membrane-free format that employs directly the polystyrene solid substrate of the 96- well microtiter plate’s (MTP) well for the spotting of microarrays.
  • MTP microtiter plate
  • the raw materials to be used for the development of the assay to detect Johne’s disease have been identified and are shown in Table 1. Further, the selected raw materials for PAGs assay have also been identified.
  • the generalized assay format of membrane-free MIA is summarized in Fig. 1.
  • the antiMAP Abs would be detected by indirect immunoassay (IA) (Fig. 1A), while the PAGs would be detected by sandwich IA (Fig. IB).
  • the multi-specific capture Ab against PAGs and the MAP antigen will be printed as spots onto the assay surface using a microarray printer either within the same well or in separate wells depending on the assay format.
  • the subsequent steps in the MIA leads to the formation of colored spots if the analytes were present in the sample (Fig. 2).
  • the intensities of colored spots are directly proportional to the concentration of analytes present in the sample.
  • MIA The proof-of-concept of MIA was demonstrated on the detachable 8-well module of 96- well MTP, where the two bovine tests, i.e. Johne’s disease and pregnancy, were performed into a single well that also incorporated four IA controls for monitoring the performance of MIA (Fig. 3).
  • the MIA could detect up to ten analytes in duplicate in a single well.
  • a milk sample is the preferred sample for the analysis of Johne’s disease and pregnancy for non- invasive in vitro diagnosis.
  • the MIA could also be performed using serum and plasma samples.
  • the Johne’s disease antigen i.e. MAP, a protoplasmic cell extract of Mycobacterium ssp., was printed at a concentration of 5 mg/ml in 1% APTES using the carbonate buffer as the print buffer.
  • MAP a protoplasmic cell extract of Mycobacterium ssp.
  • the biomolecules i.e. MAP Ag and anti -P AGs Ab, were prepared at their specific optimized concentrations in 1% APTES solution in deionized water (DIW) using the carbonate buffer as the print buffer (Fig. 4).
  • the biomolecules were then printed onto the membrane-free substrate in the desired format using an array er, e.g. s PictArrayTM, U.S. Patent No. 9,625,453, as shown in Fig. 3.
  • the printed 8-well detachable strips of a 96- well MTP were stored overnight and then blocked by incubating with 200 ul of a 2.5% casein blocking solution for 1 hour at 37°C. The excess unbound reagents were then washed away in three consecutive washings.
  • Rehydrated product of lyophilized m. phlei whole cells M. phlei bacterial culture is heat inactivated and lyophilized into dry pellets. The dry pellets are rehydrated in 0.85% saline water to form an insoluble suspension, which is used as absorbent at a concentration of 0.2-2.5 mg/ml.
  • Soluble bacterial protein extracts Proteins are extracted from heat-killed M. phlei bacterial culture and mixed with homogenous buffer solution to form the absorbent for M.phlei.
  • Absorbent present in the sample dilution buffer Diluted samples are incubated for 15- 60 mins before the MIA. Thereafter, the clear supernatant is taken as sample for the MIA.
  • Absorbent-coated preincubation plate Samples would first be diluted in absorbent- coated preincubation plate and incubated for 15-60 mins. Thereafter, the sample is transferred to the assay plate for the MIA.
  • Absorbent-coated paramagnetic beads would be added to the sample and incubated for 5-15 mins. Thereafter, the clean sample is taken for the MIA while the paramagnetic beads are held together by a magnet at the bottom of the tube.
  • Sample The milk sample is diluted 1:20 using sample diluent (PBS-T). 100 ul of the diluted milk sample is added to each MTP’s well. The MTP is incubated at 37°C for 30 min and subsequently washed 3 times with 300 pl of IX washing solution.
  • PBS-T sample diluent
  • Detection solution The HRP-labelled secondary Abs are then provided to detect the formation of specific immune complexes on the printed spots. 100 ul of mixed anti-bovine IgG conjugate and anti-PAG conjugate is added to each MTP’s well. The MTP is incubated at 37°C for 30 min and then washed 3 times with 300 pl of IX washing solution.
  • TMB The precipitating TMB is used as a substrate for HRP to develop the colorimetric spots. 100 ul of TMB is added to the MTP’s well and incubated for 15 minutes at room temperature, where the MTP is covered to protect it from light. Afterwards, the liquid is discarded, and MTP is inverted and tapped onto an absorbent paper towel to eliminate any remaining liquid. [0128] Readout: MTP is placed into a commercial reader for the determination of colorimetric intensities of spots. The results of the MIA are generated by software
  • Sample The milk sample is diluted 1:20 using sample diluent (PBS-T). 100 ul of the diluted milk sample is added to each MTP’s well. The MTP is incubated at 37°C for 30 min and subsequently washed 3 times with 300 pl of IX washing solution.
  • Secondary antibody 100 ul of specific bovine anti-PAG Ab is added to each well of MTP. The MTP is incubated at 37°C for 30 min and then washed 3 times with 300 pl of IX washing solution.
  • Detection solution 100 ul of anti-bovine IgG antibody conjugated to HRP is added to each well of MTP. The plate is incubated at 37°C for 30 min and subsequently washed 3 times with 300 pl of IX washing solution.
  • TMB The precipitating TMB is used as a substrate for HRP to develop the colorimetric spots. 100 ul of TMB is added to the MTP’s well and incubated for 15 minutes at room temperature, where the MTP is covered to protect it from light Afterwards, the liquid is discarded, and MTP is inverted and tapped onto an absorbent paper towel to eliminate any remaining liquid.
  • MTP is placed into a multiplex colorimetric reader for the determination of colorimetric intensities of spots.
  • the results of the MIA are generated by software.
  • the Bovine MIA results in the formation of colorimetric spots, the intensity of which is directly proportional to the concentration of analytes, anti -MAP IgG and PAGs, present in the milk sample.
  • the colorimetric arrays are imaged by either using a handheld colorimetric reader device or commercially available colorimetric microarray readers.
  • Image analysis software will be used and is based on an image analysis algorithm.
  • the software will analyze the colorimetric images of microarray spots in each well, which are captured by commercial readers and generate the desired results.
  • the software will identify the array wells followed by the detection of positive control spots within each well.
  • the positive control spots act as alignment anchors and are used by the software to place a microarray grid for all spots in a well.
  • the image analysis software determines the pixel intensity for each colorimetric spot based on image analysis algorithm. The data generated from each spot is then collated.
  • the automated MIA will be performed inside an analyzer, where all the steps in the manual MIA will be automated.
  • the dispensing and aspiration of reagents is done by a needle attached to the robotic arm.
  • the analyzer would have a dedicated compartment for putting the patient sample vials, and dedicated spaces for putting the wash buffer, TMB substrate and other buffers.
  • the washing of the MTP wells will be done by the robotic needle using specific washing programs.
  • the needle will be washed after each dispensing step.
  • disposable tips could also be used, which would obviate the cleaning of the needle after each dispensing step. All the steps of the IA will be optimized for the automated MIA.
  • the readout of the colorimetric array spots in the processed 96-well MTP will be performed using an integrated colorimetric reader and an image analysis software.
  • the assay formats used for development of MIA could be further employed for the development of automated CLIAs, both multiplex as well as singleplex, for the diagnosis of Johne’s disease and pregnancy in cattle.
  • the MAP antigen could be bound covalently to paramagnetic beads (micron-sub-micron size) and used for the detection of bovine IgG against MAP via indirect immunoassay.
  • the detection of PAGs could also be done by sandwich immunoassay, where the capture Abs against PAG would be coated covalently onto the paramagnetic beads.
  • the detection signal in case of automated CLIAs could be generated by conjugating the detection Ab with acridinium or other chemiluminescent labels and providing the appropriate trigger solutions for the generation of chemiluminescent signal.
  • the assay reagents are stored in the form of assay cartridges that can used for up to 100 tests.
  • the buffers, wash solution and trigger solutions are stored at the respective places in the analyzer.
  • the Bovine MIA (Membrane-free, Two wells) format utilizes the 96-well microtiter plate as the assay surface. For each 96-well plate, anti -PAG capture Ab is immobilized as duplicate spots in wells of every odd 8-well strip while MAP antigen is immobilized in duplicate in wells of every even numbered 8-well strip. [0140]
  • the assay protocol follows the general procedure described above. The milk sample is diluted and added to a well from an odd and even numbered 8-well strip and incubated at 37°C then washed with wash buffer.
  • HRP labelled anti-PAG detection Ab is added to all odd numbered wells and HRP labelled anti-bovine IgG detection Ab is added to all even numbered wells of the 96-well plate.
  • the wells are incubated at 37°C and then washed followed by the addition of TMB substrate. After a short incubation, the wells are washed once and then analyzed.
  • Manual ELISA can be developed for the detection of anti -MAP IgG and PAGs separately using the developed MIA procedure with customization of some steps for ELISA.
  • the 96-well MTP would be coated with a mixture MAP antigen either passively or using a leach-proof biomolecular immobilization procedure based on silane chemistry. All the immunoassay steps for the detection of anti-MAP IgG by ELISA would then be performed exactly as specified in the bovine MIA except the last step.
  • the signal would be generated by enzyme-substrate reaction by providing TMB and H2O2 to the HRP-labeled detection Ab.
  • the enzyme-substrate reaction is stopped by providing a stop solution comprising of IN H2SO4.
  • the optical density of the colorimetric solution is then read at 450 nm with reference at 650 nm.
  • the detection of anti-MAP IgG is done by indirect assay while the detection of PAGs is could done by sandwich assay.
  • Ab immobilization and Casein blocking Mix anti-PAG capture Ab (optimal concentration, in pg mL' 1 ) with 0.5-3 % (v/v) APTES in the ratio of 1 : 1 (v/v). Incubate each of the desired wells of a 96-well MTP with 100 pL of the freshly prepared anti-PAG capture Ab solution for 30-60 min at room temperature. Wash five times with 300 pL of 0.1M PBS, pH 7.4. Washing can also be performed with an automatic plate washer. Passive Ab immobilization, by incubating with the Ab overnight at 4 deg C, could also done. Block the Ab-bound wells with 300 pL of 0.5- 5% (w/v) casein or other appropriate blocking buffer for 30 min-2 h at 37°C followed by extensive PBS washing.
  • Sandwich ELISA Add 100 pL of the milk/serum/plasma sample to different casein or other buffer blocked wells. Incubate for 30 min-1 h at 37°C and wash extensively with PBS. Add 100 pL of HRP-labeled anti-PAG detection Ab in each of the PAG-captured wells. Incubate for 30 min-1 h at 37 °C and wash extensively with PBS. Add 100 pL of TMB-H2O2 mixture to each of these wells and incubate at room temperature to develop color for 5-30 min. Stop the enzymesubstrate reaction by adding 50 pL of 1 N H2SO4 to each well. Determine the absorbance at a primary wavelength of 450 nm taking 540 nm as the reference wavelength a microplate reader.
  • MAP antigen solution with 0.5-3% (v/v) is mixed with APTES in the ratio of 1 : 1 (v/v). Desired wells of a 96-well MTP are incubated with 100 pL of the freshly prepared antigen solution for 30 min at room temperature. Wells are washed five times with 300 pL of 0.1M PBS, pH 7.4. Washing can also be performed with an automatic plate washer. Passive Ag immobilization, by incubating with the Ag overnight at 4 deg C, could also be done. MAP Ag-bound wells are blocked with 300 pL of 0.5-5% (w/v) casein or other blocking buffer for 30 min - 2 h at 37°C followed by extensive PBS washing.
  • ELISA 100 pL of the milk/serum/plasma sample is added to different casein-blocked wells. Wells or plate are incubated for 30 min-2 h at 37°C and washed extensively with PBS. 100 pL of HRP-labeled anti-bovine IgG detection Ab is added in each of the wells. Wells are incubated for 30 min-1 h at 37 °C and washed extensively with PBS. 100 pL of TMB-H2O2 mixture is added to each of these wells and incubated at room temperature to develop color for 5-30 min. The enzyme-substrate reaction is stopped by adding 50 pL of 1 N H2SO4 to each well. The absorbance at a primary wavelength of 450 nm taking 540 nm as the reference wavelength is determined in a microplate reader.
  • a customized rapid one step kinetics-based ELISA procedure will be used for the detection of PAGs and anti-MAP IgG, as described by Vashist et al. in Biosensors and Bioelectronics 67, 73-78, 2015.
  • Another wash- free MIA format such as that based on electrochemiluminescent ELISA, could also be developed. It will involve the detection of analytes in sample using biomolecule- coated (antibody- or antigen-coated) carbon electrode surface- based microwell plates and SULFO- TAG-labeled detection Ab that emits light upon electrochemical stimulation.
  • Peptide antigen synthesis Three peptides, designed from the antigenic sites that are common to all the PAGs, were synthesized via “solid-phase peptide synthesis” that involves stepwise amino-acid coupling, which leads to the desired peptide chain (Table 2). Peptides will be used individually as antigen (Peptide 1, Peptide 2, Peptide 3).
  • Peptide conjugation The synthesized PAGs peptides will be conjugated by Proteogenix, France to KLH, OVA and/or BSA for the phage display technology based custom synthesis of anti-PAGs Ab.
  • the peptides (containing the immunogenic epitopes) bound to carriers would be used as baits for the phage display.
  • Phage display screening A mixture of the 3 custom synthesized PAG peptides bound to carriers in equimolar ratio would be used as bait for the screening of specific responders in a high diverse rabbit Ab phage display library, which consists of 1.09 xlO 10 clones. .
  • Biopanning 4-6 rounds of library screening against peptide conjugated to carriers, with specific strategy to achieve efficient (1) enrichment in binders to peptide, and (2) depletion in binders to carriers: - KLH, BSA and OVA conjugates to be alternately used: e.g. round 1 with KLH conjugate, round 2 with BSA-conjugate, round 3 with OVA-conjugate.
  • the phage DNA of the screened unique binders is then extracted and sequenced.
  • the sequences of phage display binders will be owned and filed for patent protection in this application, as clearly specified by our contract manufacturer, Proteogenix, France.
  • Affinity mapping of anti-PAGs Abs The anti-PAGs Abs corresponding to the screened unique binders would be analyzed for their binding kinetics (KON, KOFF, KD) and then graded on the basis of their affinity. The affinity mapping would be done by surface plasmon resonance based BiaCore instruments or bilayer interferometry-based Octet systems. The anti-PAGs Abs with the highest affinity would then be used for the development of sandwich IA for PAGs in the bovine MIA.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Gynecology & Obstetrics (AREA)
  • Pregnancy & Childbirth (AREA)
  • Reproductive Health (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La maladie de Johne, une infection provoquée par Mycobacterium avium paratuberculosis (MAP), est une maladie majeure limitant la production de vaches laitières. Les veaux peuvent être infectés par la maladie de Johne pendant la naissance. La présente invention concerne des méthodes pour détecter à la fois la maladie de Johne et des glycoprotéines associées à la grossesse dans un seul dosage immunologique multiplex.
PCT/NZ2021/050139 2020-08-21 2021-08-20 Dosage multiplex pour la maladie de johne et la grossesse WO2022039604A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
NZ76733320 2020-08-21
NZ767333 2020-08-21

Publications (1)

Publication Number Publication Date
WO2022039604A1 true WO2022039604A1 (fr) 2022-02-24

Family

ID=80322882

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/NZ2021/050139 WO2022039604A1 (fr) 2020-08-21 2021-08-20 Dosage multiplex pour la maladie de johne et la grossesse

Country Status (1)

Country Link
WO (1) WO2022039604A1 (fr)

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998036278A1 (fr) * 1997-02-15 1998-08-20 Beth Israel Deaconess Medical Center, Inc. Essai d'anticorps par immunocapture en plusieurs sites et necessaire correspondant
US20020192838A1 (en) * 2001-06-19 2002-12-19 Ott Troy L. Determination of pregnancy status
WO2003028582A2 (fr) * 2001-09-28 2003-04-10 Aspenbio, Inc. Test de grossesse pour bovins
WO2008067091A2 (fr) * 2006-11-28 2008-06-05 Pictor Limited Membrane de dosage et procédé d'utilisation de celle-ci
WO2008134806A1 (fr) * 2007-05-04 2008-11-13 The University Of Sydney Antigènes de mycobactérium
US20090136991A1 (en) * 2007-11-09 2009-05-28 Jones Douglas E Identifying Naive, Infected, or Vaccinated Mammals
US20140116352A1 (en) * 2012-10-31 2014-05-01 Gilles R.G. Monif Fuidi herd management and risk stratification methods
WO2019143891A1 (fr) * 2018-01-18 2019-07-25 The Penn State Research Foundation Antigènes immunodiagnostiques de mycobacterium avium subsp. paratuberculosis, procédés et kits comprenant ceux-ci

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998036278A1 (fr) * 1997-02-15 1998-08-20 Beth Israel Deaconess Medical Center, Inc. Essai d'anticorps par immunocapture en plusieurs sites et necessaire correspondant
US20020192838A1 (en) * 2001-06-19 2002-12-19 Ott Troy L. Determination of pregnancy status
WO2003028582A2 (fr) * 2001-09-28 2003-04-10 Aspenbio, Inc. Test de grossesse pour bovins
WO2008067091A2 (fr) * 2006-11-28 2008-06-05 Pictor Limited Membrane de dosage et procédé d'utilisation de celle-ci
WO2008134806A1 (fr) * 2007-05-04 2008-11-13 The University Of Sydney Antigènes de mycobactérium
US20090136991A1 (en) * 2007-11-09 2009-05-28 Jones Douglas E Identifying Naive, Infected, or Vaccinated Mammals
US20140116352A1 (en) * 2012-10-31 2014-05-01 Gilles R.G. Monif Fuidi herd management and risk stratification methods
WO2019143891A1 (fr) * 2018-01-18 2019-07-25 The Penn State Research Foundation Antigènes immunodiagnostiques de mycobacterium avium subsp. paratuberculosis, procédés et kits comprenant ceux-ci

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CHO D ET AL.: "Comparison of the proteosomes and antigenicities of secreted and cellular proteins produced by Mycobacterium paratuberculosis.", CLINICAL AND VACCINE IMMUNOLOGY, vol. 13, no. 10, October 2006 (2006-10-01), pages 1155 - 61, XP002464447, DOI: 10.1128/CVI.00058-06 *
SHIN SJ ET AL.: "Diagnosis of bovine paratuberculosis by a novel enzyme-linked immunosorbent assay based on early secreted antigens of Mycobacterium avium subsp. paratuberculosis", CLINICAL AND VACCINE IMMUNOLOGY, vol. 15, no. 8, August 2008 (2008-08-01), pages 1277 - 81, XP055097327, DOI: 10.1128/CVI.00105-08 *

Similar Documents

Publication Publication Date Title
JP6742978B2 (ja) ラテラルフローおよび関連する免疫アッセイにおけるシグナル増幅
JP5591693B2 (ja) 回虫糞便抗原の検出
US20070166776A1 (en) Immunoassay and kit for an early and simultaneous detection of biochemical markers in a patient's sample
JP2007532905A (ja) ウイルス感染の検出のための方法と装置
JP5813111B2 (ja) イムノアッセイにおける試薬の反応性を制御するためのコカップリング
US20110312922A1 (en) EDTA Resistant S100A12 Complexes (ERAC)
US11703507B2 (en) Immunoassay for SARS-CoV-2 antibodies
WO2022039604A1 (fr) Dosage multiplex pour la maladie de johne et la grossesse
US20240183852A1 (en) Multiplex immunoassay for the detection of mycoplasma bovis infection
CA2939789C (fr) Detection de globules rouges
US20220034904A1 (en) IMMUNOASSAY FOR SARS-CoV-2 ANTIBODIES
Crowther et al. Systems in ELISA
CN115616219A (zh) 高灵敏度免疫缀合物、其制备方法、包含其的体外诊断试剂以及体外诊断试剂盒
AU2022252931A9 (en) Multiplex immunoassay for the detection of mycoplasma bovis infection
WO2022025773A1 (fr) Dosage immunologique pour anticorps anti-sras-cov-2
JP7105970B1 (ja) SARS-CoV-2の免疫測定方法及び免疫測定キット
KR102504175B1 (ko) 항체의 항원 결합 부위에 결합하는 펩타이드, 이를 이용한 약물 복합체, 바이오센서 및 분석 방법
WO2022202876A1 (fr) Procédé d'analyse immunologique pour un télopeptide c-terminal de collagène de type i
RU2396565C1 (ru) Способ выявления больных описторхозом и инвазированных opisthorchis felineus кошек
JPH1048210A (ja) 特異結合分析における感度調節方法
KR101539150B1 (ko) 자가면역 항체 검출방법 및 이를 이용한 자가면역 항체 검출키트
CN113710698A (zh) 用于生物样品中的游离aim的免疫分析方法
AU2002346529B2 (en) Immunoassay and kit for an early and simulataneous detection of biochemical markers in a patient's sample
JPH1114626A (ja) Ca125の測定方法
JP2003166995A (ja) 抗体又はタンパク質を固定化したチップ

Legal Events

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

Ref document number: 21858678

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21858678

Country of ref document: EP

Kind code of ref document: A1