WO2016159960A1 - Automated immunoanalyzer system for performing diagnostic assays for autoimmune and infectious diseases - Google Patents

Automated immunoanalyzer system for performing diagnostic assays for autoimmune and infectious diseases Download PDF

Info

Publication number
WO2016159960A1
WO2016159960A1 PCT/US2015/023408 US2015023408W WO2016159960A1 WO 2016159960 A1 WO2016159960 A1 WO 2016159960A1 US 2015023408 W US2015023408 W US 2015023408W WO 2016159960 A1 WO2016159960 A1 WO 2016159960A1
Authority
WO
WIPO (PCT)
Prior art keywords
complex
conjugate
incubating
capture reagent
immune
Prior art date
Application number
PCT/US2015/023408
Other languages
French (fr)
Inventor
Mark David Van Cleve
Elaine Grace TAINE
Douglas John Canfield
Stephanie Tuvi Ortega
Taylor Addison Reid
Original Assignee
Hycor Biomedical, 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 Hycor Biomedical, Inc. filed Critical Hycor Biomedical, Inc.
Priority to EP15887969.2A priority Critical patent/EP3278109A4/en
Priority to AU2015389978A priority patent/AU2015389978A1/en
Priority to CN201580078733.XA priority patent/CN107548461A/en
Priority to CA2981455A priority patent/CA2981455A1/en
Priority to JP2017551241A priority patent/JP2018510354A/en
Priority to PCT/US2015/023408 priority patent/WO2016159960A1/en
Publication of WO2016159960A1 publication Critical patent/WO2016159960A1/en
Priority to HK18108131.3A priority patent/HK1248804A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/564Immunoassay; Biospecific binding assay; Materials therefor for pre-existing immune complex or autoimmune disease, i.e. systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, rheumatoid factors or complement components C1-C9
    • 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

Definitions

  • the present teachings are related to a system and process for performing diagnostic assays, and more particularly to an automated immunoanalyzer system and process for performing diagnostic assays for infectious and autoimmune diseases.
  • a quantitative method for performing an automated diagnostic assay for autoimmune or infectious diseases comprises the steps of incubating a capture reagent with a streptavid in-coated medium to form a solid phase complex, wherein the capture reagent is a biotinylated autoantigen or infectious disease antigen; washing the solid phase complex to remove excess capture reagent; incubating the solid phase complex with a serum sample to form an immune complex; washing the immune complex to remove any unbound sample; incubating the immune complex with a conjugate to create an immune-conjugate complex; washing the immune-conjugate complex to remove any unbound conjugate; introducing a substrate capable of generating a quantifiable response; and calibrating the response generated from the substrate.
  • a controlled process for binding fluorescent labels to particles within a patient sample includes binding luminescent labels to the particles, and quantifying the particles remaining after a series of washing steps in order to normalize a luminescence signal from the patient sample.
  • the luminescent labels are bound to the particles in proportion to a number of bound analyte molecules.
  • biotinylated capture reagent is incubated with a streptavidin-coated solid phase to illicit adhesion of the capture reagent to the solid phase by exploitation of the biotin-streptavidin interaction.
  • the capture-reagent solid phase complex is then washed to remove excess biotinylation capture reagent.
  • a serum sample is then incubated with the capture-reagent solid phase complex to illicit binding of autoantigen-specific immunoglobulins (IgG, IgM, or IgA) present in the serum to the presented capture reagent and to create an immune complex.
  • the immune complex is then washed to remove unbound immunoglobulin and then incubated with labeled antiimmunoglobulin conjugates to illicit binding of the conjugate to the autoantigen-specific immunoglobulin component of the immune complex and to create an immune-conjugate complex.
  • the immune-conjugate complex is washed to remove the unbound labeled antiimmunoglobulin and then a substrate capable of generating a quantifiable response is introduced. The quantifiable response generated from adding the substrate is calibrated and the reported value adjusted for bead retention.
  • biotinylated capture reagent is incubated with a streptavidin-coated solid phase to illicit adhesion of the capture reagent to the solid phase by exploitation of the biotin-streptavidin interaction.
  • the capture-reagent solid phase complex is then washed to remove excess biotinylation capture reagent.
  • a serum sample is then incubated with the capture-reagent solid phase complex to illicit binding of infectious agent-specific immunoglobulins (IgG, IgM, or IgA) present in the serum to the presented capture reagent and to create an immune complex.
  • infectious agent-specific immunoglobulins IgG, IgM, or IgA
  • the immune complex is then washed to remove unbound immunoglobulin and then incubated with labeled anti-immunoglobulin conjugates to illicit binding of the conjugate to the autoantigen-specific immunoglobulin component of the immune complex and to create an immune-conjugate complex.
  • the immune-conjugate complex is washed to remove the unbound labeled anti-immunoglobulin and then a substrate capable of generating a quantifiable response is introduced.
  • the quantifiable response generated from adding the substrate is calibrated and the reported value adjusted for bead retention.
  • the biotinylated capture reagent is a biotinylated autoantigen or a biotinylated infectious agent.
  • the biotinylated capture reagent is derived from the biotinylation of a purified protein, enzyme, or antibody.
  • the biotinylated capture reagent is derived from the biotinylation of an infectious agent extract comprised of a multiplicity of antigens.
  • the biotinylated capture reagent exists as an amalgam of multiple biotinylated capture reagents of different origins including purified proteins, enzymes, antibodies and extracts.
  • the streptavidin-coated solid phase is a universal fluorescent-labeled magnetic microparticle.
  • one or more of the washing steps include washing the solid phase complexes by magnetically sequestering the complex within a confined area of a reaction cuvette.
  • the step of incubating the capture reagent-solid phase complex with a serum sample includes incubating a capture reagent-solid phase complex that is retained in a suspension by a reaction diluent including a high concentration of human serum albumin (HSA).
  • a reaction diluent including a high concentration of human serum albumin (HSA).
  • the step of incubating the immune complex with a labeled anti-immmunoglobulin conjugate comprises incubating an immune complex that is retained in a suspension by a conjugate diluent including a nominal concentration of polyethylene glycol.
  • HRP horseradish peroxidase conjugated to an anti-immunoglobulin antibody
  • HRP horseradish peroxidase
  • the addition of a substrate to the immune-conjugate complex comprises adding Lumigen PS-Atto as the substrate capable of generating a quantifiable response, the quantifiable response existing as a chemiluminescent signal generated by HRP-PS-Atto reporter system and detected by a luminometer within an optics box.
  • the step of adjusting a quantifiable response for bead retention includes the steps of: transferring the substrate and immune-conjugate complex to an optics box wherein both fluorescent and chemiluminescent signals are quantified; employing a ratio of initial to final fluorescence to adjust the quantified chemiluminescent signal for bead retention; and calibrating the adjusted chemiluminescent signal to calculate a reported value.
  • an automated pipette arm with a reusable pipette tip aspirating the sample can be utilized.
  • fluorescence is measured to determine bead retention, and luminescence is measured to detect the RLU signal generated by the chemistry.
  • the measurements are entered into an algorithm to generate a "bead retention adjusted RLU" that is compared the calibration curve RLU, thereafter an immunoglubulin concentration is assigned.
  • FIG. 1 is a schematic illustration of a method for performing an automated diagnostic assay in accordance with the present application.
  • FIG. 2 is a top schematic view of an automated immunochemistry analyzer and reagent system in accordance with the teachings of the present application.
  • Disclosed herein is an automated immunoanalyzer instrument, reagent system, and methods for performing diagnostic assays for infectious and autoimmune diseases.
  • immunoassays generally require that one or more separation phases be carried out in the reaction cuvette.
  • a variety of techniques can be used, including, but not limited to, well coating techniques, bead coating techniques, or the use of paramagnetic particles.
  • Each of these separation media are coated with a capture reagent that will bind analyte molecules of interest in the patient's blood sample.
  • the biotinylated capture reagent can exist as an amalgam or mixture (i.e., capture reagents from a similar category but from different genus species).
  • capture reagents from a similar category but from different genus species.
  • numerous capture reagents are available and can be used in accordance with the present teachings. It should be understood herein that the amount and volume of each of the individual capture reagents used in accordance with the present teachings depends on their potency (i.e. their ability to produce a detectable response).
  • paramagnetic particles When paramagnetic particles are used as the separation media, the paramagnetic particles are pulled to the wall of the cuvette by magnets during the washing process and then all of the liquid is aspirated. As those of skill in the art will understand and appreciate herein, during conventional washing processes, some of the paramagnetic particles may be aspirated along with the liquid and will therefore be lost for further chemistry processing. The loss of the magnetic particles becomes even more significant if the immunoassay procedure involves several wash steps.
  • One of the objectives of the present teachings is to take into account the loss of paramagnetic particles that occur on an immunochemistry analyzer during these washing processes.
  • the analytes of interest in a patient's blood sample bind to a capture reagent that has in turn been bound to the surface of a paramagnetic particle.
  • Luminescent labels are then bound to these analyte molecules.
  • a luminescing reagent or substrate is added to the cuvette it reacts with the luminescent label to produce light that is detectable by the analyzer's optical detector.
  • the paramagnetic particles have a fluorescent label attached, fluorescently reading the contents within the cuvette will provide a means for determining the fraction of the particles that were lost during the wash steps.
  • the automated analyzer utilizes common paramagnetic particles for the assays, including, but not limited to, magnetic beads or microparticles.
  • a capture reagent is incubated and bound to the universal particles in a reaction cuvette to produce an assay- specific, particle-based reagent, sometimes referring to herein as a capture-reagent solid phase complex.
  • a capture reagent that can be used for performing a diagnostic immunoassay is comprised of biotin- antigen, 10 mM sodium phosphate, pH 7.4, 0.9% NaCI, 0.05% Tween-20, 1 % (w/v) human serum albumin, 1 %(v/v) ProClin 950, up to 5% (v/v) glycerol.
  • another capture reagent that can be used for performing a diagnostic immunoassay is comprised of biotin-antigen, 10 mM sodium phosphate, pH 7.4, 0.9% (w/v) NaCI, 0.05% Tween-20, 1 % (w/v) bovine serum albumin, 1 %(v/v) ProClin 950, 1 % protease inhibitor cocktail, 0.1 mM DTT, 25% (up to 30%) (v/v) glycerol.
  • the patient sample After undergoing a washing process, the patient sample, and optionally a diluent if needed, is added to the particles in the cuvettes and incubated. This results in the capture of specific analyte molecules in the patient's blood sample.
  • the reaction diluent (sample diluent) is comprised of 10 mM sodium phosphate, pH 7.4, 500 mM NaCI, 0.02% Tween-20, 1 % (w/v) human serum albumin, 1 % (v/v) human IgG, 1 %(v/v) ProClin 950, 0.005% Antifoam-B v/v, 2% (w/v) PEG 6,000.
  • the reaction diluent (sample diluent) is comprised of 10 mM sodium phosphate, pH 7.4, 500 mM NaCI, 0.02% Tween-20, 25% (w/v) human serum albumin, 1 %(v/v) ProClin 950.
  • HSA relative light unit
  • Another washing process is then performed to remove any excess or unbound sample, and then a luminescent label and a conjugate is added to the cuvette.
  • a luminescent label and a conjugate is added to the cuvette.
  • the particles then undergo another wash process to remove any unbound conjugate, and then the substrate is added to the cuvette and incubated for a short period of time to allow the chemiluminescent glow reaction to reach equilibrium.
  • F represents the corrected fluorescent signal (i.e., the measured signal corrected by the counting efficiency of the optical detector). Because the optical detector has a certain time resolution, as the number of photons detected per unit time increases, the likelihood of two photons arriving at the detector within that time resolution also increases. Since these two photons cannot be resolved by the detector, they will count as a single photon. Thus, the detection efficiency of the optical detector decreases as the incident photon flux increases.
  • the biotinylated capture reagents can be derived from components including, but not necessarily limited to, purified proteins, enzymes, antibodies, DNA, nuclear extracts, cellular extracts and non-protein antigens (e.g., drugs or materials cross-linked to a protein).
  • biotinylation processes and techniques commonly used for diagnostic infectious and autoimmune disease immunoassays can be utilized in accordance with the present teachings; however the biotin/protein ratio for the reaction can be optimized as needed to ensure optimum performance of the multiple biotinylated reagents used in the chemistry.
  • a specific size linker arm of the biotin reagent is NHS-PEGi 2 -Biotin.
  • the material can be crosslinked to a biotinylated protein for coating onto the streptavidin bead solid phase, while for autoimmune antigens, such as DNA, biotinylated dideoxynucleotides can be incorporated into the DNA.
  • FIG. 1 A schematic illustration of an automated diagnostic assay process in accordance with certain aspects of the present disclosure is shown in FIG. 1 .
  • magnetic beads or microparticles manufactured with a streptavidin coating are mixed (incubated) with a known biotinylated antigen (step 10).
  • biotinylated antigen step 10
  • affinity binding between streptavidin and biotin facilitates antigen coating onto the surface of the beads and thereby allows for the use of a universal bead with on-board reagent preparation.
  • a particularly useful incubation time range is from about 1 minute to about 15 minutes, more particularly from about 5 minutes to about 10 minutes and at a temperature of from about 2°C to about 40°C, more particularly from about 36.8°C to about 37.2°C.
  • the products are mixed in a reaction cuvette so that the antigens coat the beads due to the biotin/streptavidin interaction.
  • 10 ⁇ _ of streptavidin (SA)-coated bead is dispensed into the reaction cuvette, followed by 40 ⁇ _ biotinylated antigen, which is mixed during dispensation. The mixture is incubated for 1-15 minutes.
  • the buffer can be comprised of 10 mM sodium phosphate, pH 7.4, 0.9% (w/v) NaCI, 0.05% (v/v) Tween-20, 10 mg/mL HSA and 1 % (v/v) ProClin 950.
  • an external magnet is used to immobilize the magnetic beads while the washing step is performed.
  • the streptavidin coated magnetic beads are then released from the magnetic field and allowed to move freely within the reaction cuvette.
  • a biological sample (serum or plasma) is then added to the reaction cuvette, followed by 40 ⁇ _ reaction buffer, thereby re- suspending the magnetic beads (step 30).
  • a high concentration of human serum albumin (HSA) is also used within the suspension to promote macromolecular binding, as well as to keep the magnetic beads within the solution.
  • Human IgG is added into the buffer to keep the reaction linear.
  • the sample contains any antibodies (e.g., IgE, IgG, IgM, IgA) that are reactive to any of the antigen bead coatings, such antibodies will bind during this sample incubation step.
  • the sample incubation is kept at 37°C for 40 minutes. After any antibodies within the patient sample bind with the beads, a second washing step is then performed to remove any non-bound patient sample (step 40).
  • wash buffer concentrate 50 mM sodium phosphate, pH 7.4, 4.5% (w/v) NaCI, 0.05% Tween-20, 0.05%(v/v) ProClin 950, 0.02% (v/v) Antifoam-C v/v) is added to resuspend the bead and then the beads are pulled down with a magnet for 1.5 min. After the solution is removed, the magnet is moved away, and 200 ⁇ _ of wash buffer is added to resuspend the bead. The wash is then repeated one more time.
  • the beads are re-suspended in an antibody that is specific for human immunoglobulin G, M, E, or A (IgG/M/E/A).
  • the antibody is conjugated to an enzyme (such as horseradish peroxidase) to bind to any specific patient antibodies that have been captured by the beads (step 50).
  • the beads are then washed once again to remove any excess antibody (step 60), and a highly sensitive light-forming reagent (e.g., chemiluminescent substrate) is added to maximize detection sensitivity (step 70).
  • a highly sensitive light-forming reagent e.g., chemiluminescent substrate
  • Illustrative reagents that can be used as the chemiluminescent substrate in accordance with the teachings of the present disclosure include, but are not limited to, Lumigen ® PS-atto, SuperSignal ® ELISA Pico Chemiluminescent Substrate or SuperSignal ® ELISA Femto Maximum Sensitivity Substrate.
  • Lumigen ® PS-atto SuperSignal ® ELISA Pico Chemiluminescent Substrate or SuperSignal ® ELISA Femto Maximum Sensitivity Substrate.
  • xanthene dyes aromatic amines and heterocyclic amines
  • chemiluminescent compounds include, but are not limited to, dioxetane type molecules, luciferin, Lumigen ® PS- 2, Lumigen ® PS-3, Lumigen ® TMA-6, Lumigen ® TMA-3.
  • step 80 the highly sensitive light-forming reagent is added to the reaction cuvette, light is produced (step 80).
  • this light can be measured, by transferring the solution in a pipette tip into a reading station to read both the luminescent signal and fluorescent signal.
  • light emitted in accordance with the present disclosure can be detected by any suitable known detection means available within the art, including, but not limited to, a luminometer, x-ray film, high speed photographic film, a CCD camera, a scintillation counter, a chemical actinometer or visually.
  • each detection means has a different spectral sensitivity, as such; the chosen detection device can be governed by several factors including, the application and use, cost and convenience.
  • Ig antigen-specific immunoglobulin
  • RLU relative light unit
  • RLU generated by a positive/negative sample for any antigen-specific Ig (slg) is compared to RLU generated by a total IgE (tig) calibration curve.
  • the calibration curve is generated by subjecting a range of pre-diluted total Ig (tig) calibrators (which are generated from WHO standards) evaluated using a biotinylated anti-lg capture reagent.
  • FIG. 2 illustrates an automated immunochemistry analyzer and reagent system 100 that can be used to quantify and normalize the luminescence signal of an analyte sample in accordance with the teachings of the present disclosure.
  • the automated immunochemistry analyzer 100 begins by first dispensing fluorescently labelled paramagnetic particles, or fluo-beads, into a cuvette located within the reaction rotor 106.
  • an exemplary Fluo-Bead includes Fluo-Bead (SA- Speed Bead, Atto 590 labeled), 1 mg/mL.
  • the fluo-beads may initially be located in the vortexer 102 and be transferred to the reaction rotor 106 by the R1 pipettor 104.
  • the R1 pipettor 104 can aspirate a desired quantity of the fluo-bead mixture and transfer the aspirated quantity to the reaction rotor 106 where it is injected into the cuvette of the reaction rotor 106.
  • the optics pipettor 108 may aspirate a test sample from the cuvette of the reaction rotor 106 and transfer the test sample to the optics box 1 10. Once the sample is disposed within the optics box 1 10, fluorescence and luminescence measurements can be recorded.
  • the initial recording of the fluorescence and luminescence signal can be used as a baseline measurement for the fluorescence signal that can correspond to the initial concentration of fluo-beads in a sample.
  • the multi rinse pipettor 1 12 can rinse the cuvettes using a wash buffer.
  • fluo-beads may be transferred from the vortexer 102 to a cuvette in the reaction rotor 106 via the R1 pipettor 104. Then, the R1 pipettor 104 may aspirate a capture reagent from the reagent rotor 1 14 and inject the capture reagent into the cuvette located in the reaction rotor 106. After an incubation period, the single rinse pipettor 1 16 may inject a rinse buffer to resuspend the fluo-bead. A substantial amount of the suspended fluo-bead may then be localized by magnets within the reaction rotor 106 over a period of time.
  • the multi rinse pipettor 1 12 may aspirate and dispose of a portion of the rinse buffer, leaving a portion of the fluo-beads localized within the cuvette.
  • the multi rinse pipettor 1 12 may proceed to inject a wash buffer into the cuvette of the reaction rotor 106, resuspending the fluo-beads.
  • the fluo- beads may again be localized by the magnets within the reaction rotor 106 to be followed by the multi rinse pipettor 1 12 aspirating and discarding a portion of the sample that was not localized from the cuvette in the reaction rotor 106.
  • a patient sample may be contained in a sample tube on in the sample rotor 1 18.
  • the patient sample may further be partially diluted with a sample diluent.
  • the sample pipettor 120 may aspirate a portion of the patient sample and inject the patient sample into the cuvette of the reaction rotor 106 to resuspend the fluo-beads.
  • the cuvette containing the patient sample within the reaction rotor 106 may then incubate at a specific temperature, for a specific amount of time.
  • the single rinse pipettor 1 16 may inject the rinse buffer to again resuspend the fluo-beads.
  • Another localization process is performed by the reaction rotor 106 by allowing the fluo-beads to substantially collect within the cuvette near the magnets in the reaction rotor 106.
  • the multi rinse pipettor 1 12 may aspirate and discard a portion of the fluid within the cuvette of the reaction rotor 106 that was not localized during the localization process.
  • a couple of rinse cycles may then be performed on the sample within the cuvette of the reaction rotor 106.
  • the rinse cycle may comprise using the multi rinse pipettor 1 12 to inject a wash buffer into the cuvette to resuspend the fluo-beads.
  • Another localization step may allow the fluo-beads to collect within the cuvette by the magnets within the reaction rotor 106.
  • the multi rinse pipettor 1 12 may aspirate and unintentionally discard a portion of the sample, leaving a portion of the fluo-beads within the cuvette of the reaction rotor 106.
  • Another rinse cycle may then occur by using the multi rinse pipettor 1 12 to again inject wash buffer into the cuvette and allow the fluo-beads to resuspend.
  • Another fluo-bead localization process may utilize the magnets within the reaction rotor 106 to localize the fluo-beads from the rest of the sample.
  • the multi rinse pipettor 1 12 may aspirate a portion of the sample that was not localized by the localization process.
  • the R2 pipettor 122 may aspirate a conjugate contained in a conjugate cuvette within the reagent rotor 1 14.
  • the R2 pipettor 122 may then inject the previously aspirated conjugate into the cuvette of the reaction rotor 106.
  • the single rinse pipettor 1 16 may inject a rinse buffer into the cuvette in the reaction rotor 106.
  • Another fluo-bead localization cycle may be performed by allowing magnets within the reaction rotor 106 to substantially localize the fluo-beads within the cuvette.
  • the multi rinse pipettor 1 12 may aspirate and discard a portion of the sample within the cuvette that has not been localized during the localization cycle.
  • Two more rinse cycles may be performed on the sample within the cuvette of the reaction rotor 106.
  • the multi rinse pipettor 1 12 may inject a wash buffer to resuspend the fluo-beads within the cuvette.
  • Another fluo-bead localization cycle may localize the fluo- beads by locating the cuvette within close proximity to the magnets in the reaction rotor 106 over an adequate period of time.
  • the multi rinse pipettor 1 12 may aspirate and discard a portion of the sample that was not localized during the localization cycle.
  • a second wash cycle may then occur by using the multi rinse pipettor 1 12 to inject the wash buffer to resuspend the fluo-beads.
  • Another localization cycle may utilize the magnets within the reaction rotor 106 to localize the fluo-beads within the cuvette.
  • the multi rinse pipettor 1 12 may again aspirate and discard a portion of the sample that was not localized during the localization cycle.
  • the R2 pipettor 122 may aspirate a portion of conjugate from the reagent rotor 1 14 and inject the conjugate into the mixed substrate container 124 creating a mixed substrate sample.
  • the R2 pipettor may then aspirate the mixed substrate sample from the mixed substrate container 124 and inject the mixed substrate sample into the cuvette of the reaction rotor 106, resuspending the fluo-bead with the mixed substrate sample.
  • the sample in the cuvette of the reaction rotor 106 may then be aspirated by the optics pipettor 108 and placed in the optics box 1 10. After the optics box makes fluorescence and luminescence optical observations, the sample is discarded and the multi rinse pipettor rinses the cuvettes of the reaction rotor 106 in preparation for the next test.
  • Antigens can be proteins, polypeptides, peptides, nucleic acids, lipids, enzymes, immunoglobulins, or nucleotides.
  • protein refers to amino acid sequences longer than 100 amino acids.
  • polypeptides refers to amino acid sequences between 10 and 100 amino acids in length.
  • peptides refers to amino acid sequences less than 10 amino acids in length.
  • a "nucleotide” refers to a single purine or pyrimidine molecule while the term “nucleic acid” refers to a nucleotide sequence.
  • DNA is a nucleic acid while guanine is a nucleotide.
  • Non-limiting examples of autoimmune disorders that can be diagnosed using the method disclosed herein include acute disseminated encephalomyelitis (ADEM), Addison's disease, an anti-phospholipid antibody syndrome (APS), arthritis (e.g., monoarthritis, oligoarthritis, or a polyarthritis like osteoarthritis, rheumatoid arthritis, juvenile idiopathic arthritis, septic arthritis) spondyloarthropathy, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune inner ear disease, bullous pemphigoid, celiac disease, Chagas disease, chronic obstructive pulmonary disease (COPD), diabetes mellitus type 1 (IDDM), endometriosis, a gastrointestinal disorder (e.g., irritable bowel disease or an inflammatory bowel disease like Crohn's disease or ulcerative colitis), Goodpasture's syndrome, Graves' disease, Guillain-Barre syndrome (GBS),
  • Non-limiting examples of autoantigens include, but are not limited to, aggrecan, alanyl-tRNA syntetase (PL-12), alpha beta crystallin, alpha fodrin (Sptan 1 ), alpha-actinin, a1 antichymotrypsin, a1 antitripsin, a1 microglobulin, alsolase, aminoacyl-tRNA synthetase, amyloid (e.g., amyloid beta, amyloid P), annexins (e.g., annexin II, annexin V), apolipoproteins (e.g., ApoB, ApoE, ApoE4, ApoJ), aquaporin (e.g., AQP1 , AQP2, AQP3, AQP4), bactericidal/permeability-increasing protein (BPI), ⁇ -globin precursor BP1 , ⁇ -actin, ⁇ - lactoglobulin
  • Infectious agents include, but are not limited to bacteria, virus, viroids, prions, nemotodes (e.g., roundworms, pinworms), parasites (malaria, tapeworm), and fungi (e.g., yeast, ringworm).
  • infectious agent includes, but are not limited to bacteria, virus, viroids, prions, nemotodes (e.g., roundworms, pinworms), parasites (malaria, tapeworm), and fungi (e.g., yeast, ringworm).
  • infectious agent “pathogen”, “pathogenic microorganism” and “microorganism” all refer to the infectious agents listed above.
  • Antigens from infectious agents can be any isolated protein, glycoprotein, nucleic acid, enzyme, lipid, liposaccharide, or combination thereof, from an infectious agent. Antigens can also include extracts or homogenized preparations from infectious agents which include a plurality of different antigenic moieties.
  • a sample contains autoimmune antibodies to self antigens (autoantigens), or antibodies to an infectious agent
  • fluorescently labeled paramagnetic microparticles e.g., fluo-beads
  • a sample of the cuvette is obtained prior to washing for a fluorescent reading to determination of the starting number of magnetic beads in the reaction cuvette.
  • the patient sample, and optionally a diluent if needed is added to the particles in the cuvettes and incubated. This results in the capture of specific analyte molecules (antibodies) in the patient's blood sample.
  • Another washing process is then performed to remove any excess or unbound sample, and then a luminescent label and a conjugate is added to the cuvette.
  • a luminescent label and a conjugate is added to the cuvette.
  • the particles then undergo another wash process to remove any unbound conjugate, and then the substrate is added to the cuvette and incubated for a short period of time to allow the chemiluminescent glow reaction to reach equilibrium.
  • the reagent solution is mixed and placed on ice for 2 hours. Free biotin reagent is separated from the biotinylated antibody by dialysis against two changes of PBS (volume ratio of antibody to buffer - 1 : 100) at 2-8 °C for 4 hours and overnight.
  • PBS volume ratio of antibody to buffer - 1 : 100
  • Fluo-Bead Fluorescence labeled para-magnetic microparticles
  • bead concentration 1 mg/mL
  • 40 ⁇ of biotin-antigen e.g., infectious disease antigen, autoantigen, etc
  • biotin-anti-lg antibody e.g., biotin-anti-lg antibody
  • a 10 ⁇ _ sample was added to 40 ⁇ _ of suspended antigen-coated beads in reaction cuvette.
  • 10 ⁇ _ of serum standards (secondary standards calibrated against a WHO Ig Standard) are each added to 40 ⁇ _ of anti-lg-coated beads in a reaction cuvette.
  • anti-lg conjugates are utilized: for assays: anti-lgE-HRP; anti-lgA-HRP, anti-lgG-HRP, anti-lgM-HRP, anti-ECP-HRP, and anti-tryptase-HRP.
  • each conjugate has an optimized HRP incorporation ratio for use in the chemistry.
  • the range of HRP incorporation ratio used for the listed conjugates is between about 1.2 and about 5.4.
  • the present teachings also contemplate the incorporation of other types of conjugate-reporter systems including, but not limited to: alkaline phosphatase conjugate and b-galactosidase conjugate.
  • a list of illustrative reagents and components that may be used in accordance with the present teachings include, but are not necessarily limited to: Bead: Fluo-Bead (SA- Speed Bead, Atto 590 labeled), 1 mg/mL; Capture Reagent Diluent: IgE: 10 mM sodium phosphate, pH 7.4, 0.9% (w/v) NaCI, 0.05% Tween-20, 1 % (w/v) human serum albumin, 1 %(v/v) ProClin 950, up to 5% (v/v) glycerol; ANA: 10 mM sodium phosphate, pH 7.4, 0.9% (w/v) NaCI, 0.05% Tween-20, 1 % (w/v) bovine serum albumin, 1 % Protease inhibitor cocktail, 0.1 mM DTT, 1 %(v/v) ProClin 950, 25% (up to 30%) (v/v) glycerol; Wash
  • Fluo-Bead Fluorescence labeled para-magnetic microparticles
  • bead concentration 1 mg/mL
  • 40 ⁇ _ of biotin-antigen e.g., myelin basic protein, collagen, insulin, etc
  • biotin-anti-lgG antibody or alternatively, anti-lgM or anti-lgA
  • a sample of the labeled Fluo-Bead is obtained to determine the starting number of beads.
  • antigen- or anti-lgG-coated beads are resuspended in 40 ⁇ _ of reaction buffer.
  • Serum samples obtained from individuals are assayed against antigens.
  • a 10 ⁇ _ sample is added to 40 ⁇ _ of suspended antigen-coated beads in reaction cuvette.
  • 10 ⁇ _ of serum standards (secondary standards calibrated against the WHO IgG Standard) are each added to 40 ⁇ _ of anti-lgG-coated beads in a reaction cuvette.
  • Fluo-Bead Fluorescence labeled para-magnetic microparticles
  • bead concentration 1 mg/mL
  • 40 ⁇ of biotin-antigen from e.g., human immunodeficiency virus, Ebola virus, Mycobacterium tuberculosis, etc
  • biotin-anti-lgG antibody or alternatively, anti-lgM or anti-lgA
  • a sample of the labeled Fluo-Bead is obtained to determine the starting number of beads.
  • antigen- or anti-lgG- coated beads are resuspended in 40 ⁇ of reaction buffer. Serum samples obtained from individuals are assayed against antigens. A 10 ⁇ sample is added to 40 ⁇ of suspended antigen-coated beads in reaction cuvette. For the six point standard curve, 10 ⁇ of serum standards (secondary standards calibrated against the WHO IgG Standard) are each added to 40 ⁇ L of anti-lgG-coated beads in a reaction cuvette. [0072] The solutions are mixed and incubated for 40 min at 37°C. After washing, beads are resuspended in 50 ⁇ _ of anti-human IgG-HRP conjugate, and incubated at 37°C for 30 min.
  • PS-atto 50 ⁇ _ of PS-atto (Lumigen) is added into each cuvette and the beads resuspended.
  • the bead suspension is transferred into a pipette tip and read in the optic box for both fluorescence and luminescence signal.
  • the standard curve is determined using a four parameter logistic function equation and levels of specific IgG to the infectious agent antigens interpolated from the standard curve.

Abstract

A quantitative method for diagnosing an autoimmune disease or an infectious disease comprising performing an automated diagnostic assay, comprising: incubating a capture reagent with a streptavidin-coated medium to form a solid phase complex, wherein the capture reagent is a biotinylated autoantigen or infectious disease antigen; washing the solid phase complex to remove excess capture reagent; incubating the solid phase complex with a serum sample to form an immune complex; washing the immune complex to remove any unbound sample; incubating the immune complex with a conjugate to create an immune-conjugate complex; washing the immune-conjugate complex to remove any unbound conjugate; introducing a substrate capable of generating a quantifiable response; and calibrating the response generated from introducing the substrate.

Description

AUTOMATED IMMUNOANALYZER SYSTEM FOR PERFORMING DIAGNOSTIC ASSAYS FOR AUTOIMMUNE AND INFECTIOUS DISEASES
TECHNICAL FIELD
[0001] The present teachings are related to a system and process for performing diagnostic assays, and more particularly to an automated immunoanalyzer system and process for performing diagnostic assays for infectious and autoimmune diseases.
SUMMARY OF THE DISCLOSURE
[0002] In accordance with certain aspects of the present application, a quantitative method for performing an automated diagnostic assay for autoimmune or infectious diseases is provided and comprises the steps of incubating a capture reagent with a streptavid in-coated medium to form a solid phase complex, wherein the capture reagent is a biotinylated autoantigen or infectious disease antigen; washing the solid phase complex to remove excess capture reagent; incubating the solid phase complex with a serum sample to form an immune complex; washing the immune complex to remove any unbound sample; incubating the immune complex with a conjugate to create an immune-conjugate complex; washing the immune-conjugate complex to remove any unbound conjugate; introducing a substrate capable of generating a quantifiable response; and calibrating the response generated from the substrate.
[0003] In accordance with yet other aspects of the present application, a controlled process for binding fluorescent labels to particles within a patient sample is provided. In accordance with this aspect of the present disclosure, the process includes binding luminescent labels to the particles, and quantifying the particles remaining after a series of washing steps in order to normalize a luminescence signal from the patient sample. According to this illustrative process, the luminescent labels are bound to the particles in proportion to a number of bound analyte molecules.
[0004] In accordance with still other aspects of the present disclosure, quantitative methods for evaluating autoimmune-specific immunoglobulins in a serum sample designed for use on an automated platform are provided. In accordance with this method, biotinylated capture reagent is incubated with a streptavidin-coated solid phase to illicit adhesion of the capture reagent to the solid phase by exploitation of the biotin-streptavidin interaction. The capture-reagent solid phase complex is then washed to remove excess biotinylation capture reagent. A serum sample is then incubated with the capture-reagent solid phase complex to illicit binding of autoantigen-specific immunoglobulins (IgG, IgM, or IgA) present in the serum to the presented capture reagent and to create an immune complex. The immune complex is then washed to remove unbound immunoglobulin and then incubated with labeled antiimmunoglobulin conjugates to illicit binding of the conjugate to the autoantigen-specific immunoglobulin component of the immune complex and to create an immune-conjugate complex. The immune-conjugate complex is washed to remove the unbound labeled antiimmunoglobulin and then a substrate capable of generating a quantifiable response is introduced. The quantifiable response generated from adding the substrate is calibrated and the reported value adjusted for bead retention.
[0005] In accordance with still other aspects of the present disclosure, quantitative methods for evaluating infectious agent-specific immunoglobulins in a serum sample designed for use on an automated platform is provided. In accordance with this method, biotinylated capture reagent is incubated with a streptavidin-coated solid phase to illicit adhesion of the capture reagent to the solid phase by exploitation of the biotin-streptavidin interaction. The capture-reagent solid phase complex is then washed to remove excess biotinylation capture reagent. A serum sample is then incubated with the capture-reagent solid phase complex to illicit binding of infectious agent-specific immunoglobulins (IgG, IgM, or IgA) present in the serum to the presented capture reagent and to create an immune complex. The immune complex is then washed to remove unbound immunoglobulin and then incubated with labeled anti-immunoglobulin conjugates to illicit binding of the conjugate to the autoantigen-specific immunoglobulin component of the immune complex and to create an immune-conjugate complex. The immune-conjugate complex is washed to remove the unbound labeled anti-immunoglobulin and then a substrate capable of generating a quantifiable response is introduced. The quantifiable response generated from adding the substrate is calibrated and the reported value adjusted for bead retention.
[0006] In accordance with certain aspects herein, the biotinylated capture reagent is a biotinylated autoantigen or a biotinylated infectious agent.
[0007] In accordance with certain aspects herein, the biotinylated capture reagent is derived from the biotinylation of a purified protein, enzyme, or antibody.
[0008] In accordance with other aspects herein, the biotinylated capture reagent is derived from the biotinylation of an infectious agent extract comprised of a multiplicity of antigens.
[0009] According to specific illustrative aspects of the present disclosure, the biotinylated capture reagent exists as an amalgam of multiple biotinylated capture reagents of different origins including purified proteins, enzymes, antibodies and extracts.
[0010] According to yet another specific illustrative aspect of the present disclosure, the streptavidin-coated solid phase is a universal fluorescent-labeled magnetic microparticle. [0011] In accordance with certain aspects of the present disclosure, one or more of the washing steps include washing the solid phase complexes by magnetically sequestering the complex within a confined area of a reaction cuvette.
[0012] In accordance with yet other illustrative aspects of the present disclosure, the step of incubating the capture reagent-solid phase complex with a serum sample includes incubating a capture reagent-solid phase complex that is retained in a suspension by a reaction diluent including a high concentration of human serum albumin (HSA).
[0013] In accordance with still another illustrative aspect of the present disclosure, the step of incubating the immune complex with a labeled anti-immmunoglobulin conjugate comprises incubating an immune complex that is retained in a suspension by a conjugate diluent including a nominal concentration of polyethylene glycol.
[0014] According to certain specific aspects of the present disclosure, horseradish peroxidase (HRP) conjugated to an anti-immunoglobulin antibody can be used as an indirect label when washing the immune complex to remove unbound immunoglobulin, particularly as the reaction of PS-Atto with an HRP labeled conjugate generates sustained high-intensity luminescence for maximum detection sensitivity in solution assays.
[0015] According to still other specific aspects of the present disclosure, the addition of a substrate to the immune-conjugate complex comprises adding Lumigen PS-Atto as the substrate capable of generating a quantifiable response, the quantifiable response existing as a chemiluminescent signal generated by HRP-PS-Atto reporter system and detected by a luminometer within an optics box.
[0016] In accordance with certain aspects of the present teachings, the step of adjusting a quantifiable response for bead retention includes the steps of: transferring the substrate and immune-conjugate complex to an optics box wherein both fluorescent and chemiluminescent signals are quantified; employing a ratio of initial to final fluorescence to adjust the quantified chemiluminescent signal for bead retention; and calibrating the adjusted chemiluminescent signal to calculate a reported value. To transfer the substrate and immune-conjugate complex to the optics box, an automated pipette arm with a reusable pipette tip aspirating the sample can be utilized. Within the optics box, fluorescence is measured to determine bead retention, and luminescence is measured to detect the RLU signal generated by the chemistry. The measurements are entered into an algorithm to generate a "bead retention adjusted RLU" that is compared the calibration curve RLU, thereafter an immunoglubulin concentration is assigned.
[0017] Still other objects and benefits of the invention will become apparent from the following written description along with the accompanying figures. BRIEF DESCRIPTION OF THE DRAWINGS
[0018] The above-mentioned aspects of the present disclosure and the manner of obtaining them will become more apparent and the disclosure itself will be better understood by reference to the following description of the embodiments of the disclosure taken in conjunction with the accompanying drawings, wherein:
[0019] FIG. 1 is a schematic illustration of a method for performing an automated diagnostic assay in accordance with the present application; and
[0020] FIG. 2 is a top schematic view of an automated immunochemistry analyzer and reagent system in accordance with the teachings of the present application.
[0021] Corresponding reference characters indicate corresponding parts throughout the several views. Although the exemplification set out herein illustrates embodiments of the disclosure, in several forms, the embodiments disclosed below are not intended to be exhaustive or to be construed as limiting the scope of the disclosure to the precise forms disclosed.
DETAILED DESCRIPTION
[0022] Disclosed herein is an automated immunoanalyzer instrument, reagent system, and methods for performing diagnostic assays for infectious and autoimmune diseases.
[0023] Before describing in detail the illustrative automated immunoanalyzer system and method of the present disclosure, it should be understood and appreciated herein that as a way of minimizing background signals from excess or unbound materials, immunoassays generally require that one or more separation phases be carried out in the reaction cuvette. To facilitate the separation or washing process, a variety of techniques can be used, including, but not limited to, well coating techniques, bead coating techniques, or the use of paramagnetic particles. Each of these separation media are coated with a capture reagent that will bind analyte molecules of interest in the patient's blood sample. In accordance with certain aspects of the present teachings, the biotinylated capture reagent can exist as an amalgam or mixture (i.e., capture reagents from a similar category but from different genus species). As those of skill in the art would understand and appreciated herein, numerous capture reagents are available and can be used in accordance with the present teachings. It should be understood herein that the amount and volume of each of the individual capture reagents used in accordance with the present teachings depends on their potency (i.e. their ability to produce a detectable response).
[0024] When paramagnetic particles are used as the separation media, the paramagnetic particles are pulled to the wall of the cuvette by magnets during the washing process and then all of the liquid is aspirated. As those of skill in the art will understand and appreciate herein, during conventional washing processes, some of the paramagnetic particles may be aspirated along with the liquid and will therefore be lost for further chemistry processing. The loss of the magnetic particles becomes even more significant if the immunoassay procedure involves several wash steps.
[0025] One of the objectives of the present teachings is to take into account the loss of paramagnetic particles that occur on an immunochemistry analyzer during these washing processes. To accomplish this, in accordance with certain aspects of the present teachings, the analytes of interest in a patient's blood sample bind to a capture reagent that has in turn been bound to the surface of a paramagnetic particle. Luminescent labels are then bound to these analyte molecules. When a luminescing reagent or substrate is added to the cuvette it reacts with the luminescent label to produce light that is detectable by the analyzer's optical detector. In addition, if the paramagnetic particles have a fluorescent label attached, fluorescently reading the contents within the cuvette will provide a means for determining the fraction of the particles that were lost during the wash steps.
[0026] According to certain aspects of the present disclosure, the automated analyzer utilizes common paramagnetic particles for the assays, including, but not limited to, magnetic beads or microparticles. For each assay onboard the analyzer, a capture reagent is incubated and bound to the universal particles in a reaction cuvette to produce an assay- specific, particle-based reagent, sometimes referring to herein as a capture-reagent solid phase complex. In accordance with certain aspects of the present disclosure, a capture reagent that can be used for performing a diagnostic immunoassay is comprised of biotin- antigen, 10 mM sodium phosphate, pH 7.4, 0.9% NaCI, 0.05% Tween-20, 1 % (w/v) human serum albumin, 1 %(v/v) ProClin 950, up to 5% (v/v) glycerol. In accordance with still other aspects of the present disclosure, another capture reagent that can be used for performing a diagnostic immunoassay is comprised of biotin-antigen, 10 mM sodium phosphate, pH 7.4, 0.9% (w/v) NaCI, 0.05% Tween-20, 1 % (w/v) bovine serum albumin, 1 %(v/v) ProClin 950, 1 % protease inhibitor cocktail, 0.1 mM DTT, 25% (up to 30%) (v/v) glycerol.
[0027] After undergoing a washing process, the patient sample, and optionally a diluent if needed, is added to the particles in the cuvettes and incubated. This results in the capture of specific analyte molecules in the patient's blood sample. In accordance with one specific illustrative aspect of the present disclosure, the reaction diluent (sample diluent) is comprised of 10 mM sodium phosphate, pH 7.4, 500 mM NaCI, 0.02% Tween-20, 1 % (w/v) human serum albumin, 1 % (v/v) human IgG, 1 %(v/v) ProClin 950, 0.005% Antifoam-B v/v, 2% (w/v) PEG 6,000. In accordance with yet another specific illustrative aspect of the present disclosure, the reaction diluent (sample diluent) is comprised of 10 mM sodium phosphate, pH 7.4, 500 mM NaCI, 0.02% Tween-20, 25% (w/v) human serum albumin, 1 %(v/v) ProClin 950.
[0028] In accordance with these illustrative embodiments, it should be understood herein that the high percentage of HSA (25%) functions in part to increase the viscosity of the reaction medium in order to retain beads in suspension during the incubation step. In addition, high HSA also reduces non-specific binding during this incubation, and improves relative light unit (RLU) linearity upon dilution of the patient sample.
[0029] Another washing process is then performed to remove any excess or unbound sample, and then a luminescent label and a conjugate is added to the cuvette. When added to the cuvette, it can be expected that some portion of the conjugate will bind to the capture reagent/sample complex on the paramagnetic particles after an incubation period. The particles then undergo another wash process to remove any unbound conjugate, and then the substrate is added to the cuvette and incubated for a short period of time to allow the chemiluminescent glow reaction to reach equilibrium.
[0030] After equilibrium is reached, luminescence and fluorescence readings of the sample are taken. Since the paramagnetic particles are contained on the analyzer in a common reagent vial and are maintained in a state of uniform suspension before being pipetted into the reaction cuvettes, initial fluorescence measurements of the particles after they are pipetted into the cuvettes, when combined with the final fluorescence measurement for each test, can be used to determine the fraction of the initial particles that remain in the cuvette after the immunoassay process. The fraction remaining is given by the following formula:
_ , . , . , . . . Ffinal"Fback2round
Particle fraction remaining = ,
F initial - F background
where F represents the corrected fluorescent signal (i.e., the measured signal corrected by the counting efficiency of the optical detector). Because the optical detector has a certain time resolution, as the number of photons detected per unit time increases, the likelihood of two photons arriving at the detector within that time resolution also increases. Since these two photons cannot be resolved by the detector, they will count as a single photon. Thus, the detection efficiency of the optical detector decreases as the incident photon flux increases.
[0031] Because of the very high flux of fluorescence excitation photons which interact with and scatter from the container walls for the paramagnetic particles, there will be a certain number of photons that will be counted by the optical detector, even when no fluorescent material is present. This corrected background signal is represented by Fbackground. [0032] The use of a fluorescent measurement to determine the percentage of the initial paramagnetic particles that remain in a reaction cuvette throughout the immunoassay process is beneficial because the process does not limit system throughput, particularly because the process does not restrict the timing or parallel processing that can be achieved. Most conventional immunoassay analyzers, on the other hand, rely on very reproducible processing of paramagnetic particles and samples, which do indeed restrict the timing or parallel processing that can be achieved, and as a result, also limit system throughput. While changes in processing efficiency over time in these conventional immunoassay analyzers may go undetected, these changes can be detected with fluorescence detection. The teachings of the present disclosure permit the use of parallel processing (e.g. multiple wash arms), which vary in wash efficiency due to minor mechanical alignment or fluidic differences. Fluorescence readings taken after each step of an immunoassay process are useful for verifying the equivalent functionality of the parallel processes.
[0033] An automated immunoanalyzer instrument and reagent system for performing diagnostic assays for infectious and autoimmune diseases in accordance with the above- described methods and techniques is now described in greater detail. As this process is described, it should be understood and appreciated herein that the disclosed instrumentation used to perform the assay can be configured to accept standard or universal collection tubes so that a variety of different tests can be conducted by the system. Those of skill in the art will also understand and appreciate herein that there are many known methods for isolating antigens, including infectious disease and autoimmune antigens from source materials. Because these isolating methods are widely known and accepted within the art, they are not discussed in detail herein, particularly as those of skill in the art will recognize that any acceptable antigen isolation methods may be incorporated into the inventive system without departing from its spirit or scope. After the infectious disease or autoimmune antigens have been isolated, they can then be conjugated with biotin to create biotinylated antigens or capture reagents. The biotinylated antigens are then contacted with a streptavidin-linked solid support or membrane. In accordance with certain aspects of the present disclosure, the biotinylated capture reagents can be derived from components including, but not necessarily limited to, purified proteins, enzymes, antibodies, DNA, nuclear extracts, cellular extracts and non-protein antigens (e.g., drugs or materials cross-linked to a protein).
[0034] As those of skill in the art will understand and appreciate herein, standard biotinylation processes and techniques commonly used for diagnostic infectious and autoimmune disease immunoassays can be utilized in accordance with the present teachings; however the biotin/protein ratio for the reaction can be optimized as needed to ensure optimum performance of the multiple biotinylated reagents used in the chemistry. In accordance with certain aspects of the present teachings, a specific size linker arm of the biotin reagent is NHS-PEGi2-Biotin. Moreover, for non-protein antigens, the material can be crosslinked to a biotinylated protein for coating onto the streptavidin bead solid phase, while for autoimmune antigens, such as DNA, biotinylated dideoxynucleotides can be incorporated into the DNA.
[0035] A schematic illustration of an automated diagnostic assay process in accordance with certain aspects of the present disclosure is shown in FIG. 1 . In accordance with this illustrative embodiment, magnetic beads or microparticles manufactured with a streptavidin coating are mixed (incubated) with a known biotinylated antigen (step 10). As those of skill in the art will understand and appreciate herein, the well-known affinity binding between streptavidin and biotin facilitates antigen coating onto the surface of the beads and thereby allows for the use of a universal bead with on-board reagent preparation. It should also be understood and appreciated herein that the amount of time and associated laboratory conditions required in order to incubate the biotinylated capture reagents with the streptavidin-coated solid phase in accordance with the present teachings may vary in light of the specific experiment being conducted, however, in accordance with certain aspects of the present disclosure, a particularly useful incubation time range is from about 1 minute to about 15 minutes, more particularly from about 5 minutes to about 10 minutes and at a temperature of from about 2°C to about 40°C, more particularly from about 36.8°C to about 37.2°C.
[0036] As shown in Table 1 below, in accordance with this aspect of the present disclosure, the following beads may be used for the magnetic supports disclosed herein:
Table 1
Figure imgf000009_0001
[0037] While numerous processes can be utilized for mixing or incubating the biotinylated antigens with the streptavidin coated beads, in accordance with certain specific embodiments, the products are mixed in a reaction cuvette so that the antigens coat the beads due to the biotin/streptavidin interaction. According to one illustrative embodiment, 10 μΙ_ of streptavidin (SA)-coated bead is dispensed into the reaction cuvette, followed by 40 μΙ_ biotinylated antigen, which is mixed during dispensation. The mixture is incubated for 1-15 minutes. Excess biotinylated antigen can then be washed off by pulling the magnetic beads to one side of the reaction cuvette and immobilizing them while buffer is washed through the reaction cuvette (step 20). In accordance with one illustrative embodiment, the buffer can be comprised of 10 mM sodium phosphate, pH 7.4, 0.9% (w/v) NaCI, 0.05% (v/v) Tween-20, 10 mg/mL HSA and 1 % (v/v) ProClin 950. While those of skill in the art can utilize any readily available immobilization techniques known within the art to cause the magnetic beads to stay on one side of the reaction cuvette, in accordance with certain specific illustrative embodiments, an external magnet is used to immobilize the magnetic beads while the washing step is performed.
[0038] The streptavidin coated magnetic beads are then released from the magnetic field and allowed to move freely within the reaction cuvette. A biological sample (serum or plasma) is then added to the reaction cuvette, followed by 40 μΙ_ reaction buffer, thereby re- suspending the magnetic beads (step 30). In addition to the biological sample, in accordance with certain aspects of the present disclosure, a high concentration of human serum albumin (HSA) is also used within the suspension to promote macromolecular binding, as well as to keep the magnetic beads within the solution. Human IgG is added into the buffer to keep the reaction linear.
[0039] If the sample contains any antibodies (e.g., IgE, IgG, IgM, IgA) that are reactive to any of the antigen bead coatings, such antibodies will bind during this sample incubation step. The sample incubation is kept at 37°C for 40 minutes. After any antibodies within the patient sample bind with the beads, a second washing step is then performed to remove any non-bound patient sample (step 40). 150 μΙ_ of wash buffer concentrate (50 mM sodium phosphate, pH 7.4, 4.5% (w/v) NaCI, 0.05% Tween-20, 0.05%(v/v) ProClin 950, 0.02% (v/v) Antifoam-C v/v) is added to resuspend the bead and then the beads are pulled down with a magnet for 1.5 min. After the solution is removed, the magnet is moved away, and 200 μΙ_ of wash buffer is added to resuspend the bead. The wash is then repeated one more time.
[0040] After the second washing step is performed, the beads are re-suspended in an antibody that is specific for human immunoglobulin G, M, E, or A (IgG/M/E/A). In accordance with certain aspects of the present disclosure, the antibody is conjugated to an enzyme (such as horseradish peroxidase) to bind to any specific patient antibodies that have been captured by the beads (step 50). The beads are then washed once again to remove any excess antibody (step 60), and a highly sensitive light-forming reagent (e.g., chemiluminescent substrate) is added to maximize detection sensitivity (step 70). Illustrative reagents that can be used as the chemiluminescent substrate in accordance with the teachings of the present disclosure include, but are not limited to, Lumigen® PS-atto, SuperSignal® ELISA Pico Chemiluminescent Substrate or SuperSignal® ELISA Femto Maximum Sensitivity Substrate. Those of skill in the art will understand and appreciate that numerous compounds of various structural classes, including xanthene dyes, aromatic amines and heterocyclic amines can be used to produce chemiluminescence under these conditions. These compounds are well-known within the patent literatures and are readily available through many commercial venders. Some non-limiting chemiluminescent compounds include, but are not limited to, dioxetane type molecules, luciferin, Lumigen® PS- 2, Lumigen® PS-3, Lumigen® TMA-6, Lumigen® TMA-3.
[0041] Once the highly sensitive light-forming reagent is added to the reaction cuvette, light is produced (step 80). In accordance with certain embodiments, this light can be measured, by transferring the solution in a pipette tip into a reading station to read both the luminescent signal and fluorescent signal. It should be understood, however, that light emitted in accordance with the present disclosure can be detected by any suitable known detection means available within the art, including, but not limited to, a luminometer, x-ray film, high speed photographic film, a CCD camera, a scintillation counter, a chemical actinometer or visually. As those of skill in the art readily understand and appreciate, each detection means has a different spectral sensitivity, as such; the chosen detection device can be governed by several factors including, the application and use, cost and convenience. Moreover, as used herein, a quantifiable or detectable response that can be measured in accordance with the present disclosure implies that a positive sample with antigen-specific immunoglobulin (Ig, e.g., IgE, IgG, IgM, IgA) caused binding of Anti-lg-HRP which would generate luminosity (i.e., RLU = relative light unit) upon addition of the substrate. In addition, it should also be understood herein that a quantifiable or detectable response can also apply to a quantifiable response generated by a negative sample.
[0042] In accordance with certain aspects of the present teachings, RLU generated by a positive/negative sample for any antigen-specific Ig (slg) is compared to RLU generated by a total IgE (tig) calibration curve. The calibration curve is generated by subjecting a range of pre-diluted total Ig (tig) calibrators (which are generated from WHO standards) evaluated using a biotinylated anti-lg capture reagent.
[0043] To better understand the mechanical aspects of this disclosure, FIG. 2 illustrates an automated immunochemistry analyzer and reagent system 100 that can be used to quantify and normalize the luminescence signal of an analyte sample in accordance with the teachings of the present disclosure. According to this illustrative aspect, the automated immunochemistry analyzer 100 begins by first dispensing fluorescently labelled paramagnetic particles, or fluo-beads, into a cuvette located within the reaction rotor 106. In accordance with one embodiment herein, an exemplary Fluo-Bead includes Fluo-Bead (SA- Speed Bead, Atto 590 labeled), 1 mg/mL.
[0044] The fluo-beads may initially be located in the vortexer 102 and be transferred to the reaction rotor 106 by the R1 pipettor 104. The R1 pipettor 104 can aspirate a desired quantity of the fluo-bead mixture and transfer the aspirated quantity to the reaction rotor 106 where it is injected into the cuvette of the reaction rotor 106. Following the injection into the cuvette, the optics pipettor 108 may aspirate a test sample from the cuvette of the reaction rotor 106 and transfer the test sample to the optics box 1 10. Once the sample is disposed within the optics box 1 10, fluorescence and luminescence measurements can be recorded. The initial recording of the fluorescence and luminescence signal can be used as a baseline measurement for the fluorescence signal that can correspond to the initial concentration of fluo-beads in a sample. After recording the measurements, the multi rinse pipettor 1 12 can rinse the cuvettes using a wash buffer.
[0045] Next, fluo-beads may be transferred from the vortexer 102 to a cuvette in the reaction rotor 106 via the R1 pipettor 104. Then, the R1 pipettor 104 may aspirate a capture reagent from the reagent rotor 1 14 and inject the capture reagent into the cuvette located in the reaction rotor 106. After an incubation period, the single rinse pipettor 1 16 may inject a rinse buffer to resuspend the fluo-bead. A substantial amount of the suspended fluo-bead may then be localized by magnets within the reaction rotor 106 over a period of time. After the magnets have substantially localized the fluo-beads within the cuvette, the multi rinse pipettor 1 12 may aspirate and dispose of a portion of the rinse buffer, leaving a portion of the fluo-beads localized within the cuvette. The multi rinse pipettor 1 12 may proceed to inject a wash buffer into the cuvette of the reaction rotor 106, resuspending the fluo-beads. The fluo- beads may again be localized by the magnets within the reaction rotor 106 to be followed by the multi rinse pipettor 1 12 aspirating and discarding a portion of the sample that was not localized from the cuvette in the reaction rotor 106.
[0046] A patient sample may be contained in a sample tube on in the sample rotor 1 18. The patient sample may further be partially diluted with a sample diluent. At this point, the sample pipettor 120 may aspirate a portion of the patient sample and inject the patient sample into the cuvette of the reaction rotor 106 to resuspend the fluo-beads. The cuvette containing the patient sample within the reaction rotor 106 may then incubate at a specific temperature, for a specific amount of time. After incubation, the single rinse pipettor 1 16 may inject the rinse buffer to again resuspend the fluo-beads. Another localization process is performed by the reaction rotor 106 by allowing the fluo-beads to substantially collect within the cuvette near the magnets in the reaction rotor 106. After the localization of the fluo- beads, the multi rinse pipettor 1 12 may aspirate and discard a portion of the fluid within the cuvette of the reaction rotor 106 that was not localized during the localization process.
[0047] A couple of rinse cycles may then be performed on the sample within the cuvette of the reaction rotor 106. The rinse cycle may comprise using the multi rinse pipettor 1 12 to inject a wash buffer into the cuvette to resuspend the fluo-beads. Another localization step may allow the fluo-beads to collect within the cuvette by the magnets within the reaction rotor 106. After a period allowing for adequate localization of the fluo-beads, the multi rinse pipettor 1 12 may aspirate and unintentionally discard a portion of the sample, leaving a portion of the fluo-beads within the cuvette of the reaction rotor 106. Another rinse cycle may then occur by using the multi rinse pipettor 1 12 to again inject wash buffer into the cuvette and allow the fluo-beads to resuspend. Another fluo-bead localization process may utilize the magnets within the reaction rotor 106 to localize the fluo-beads from the rest of the sample. Finally, the multi rinse pipettor 1 12 may aspirate a portion of the sample that was not localized by the localization process.
[0048] At this point, the R2 pipettor 122 may aspirate a conjugate contained in a conjugate cuvette within the reagent rotor 1 14. The R2 pipettor 122 may then inject the previously aspirated conjugate into the cuvette of the reaction rotor 106. After incubating the cuvette under controlled time and temperature in the reaction rotor 106, the single rinse pipettor 1 16 may inject a rinse buffer into the cuvette in the reaction rotor 106. Another fluo-bead localization cycle may be performed by allowing magnets within the reaction rotor 106 to substantially localize the fluo-beads within the cuvette. The multi rinse pipettor 1 12 may aspirate and discard a portion of the sample within the cuvette that has not been localized during the localization cycle.
[0049] Two more rinse cycles may be performed on the sample within the cuvette of the reaction rotor 106. The multi rinse pipettor 1 12 may inject a wash buffer to resuspend the fluo-beads within the cuvette. Another fluo-bead localization cycle may localize the fluo- beads by locating the cuvette within close proximity to the magnets in the reaction rotor 106 over an adequate period of time. After the localization cycle, the multi rinse pipettor 1 12 may aspirate and discard a portion of the sample that was not localized during the localization cycle. A second wash cycle may then occur by using the multi rinse pipettor 1 12 to inject the wash buffer to resuspend the fluo-beads. Another localization cycle may utilize the magnets within the reaction rotor 106 to localize the fluo-beads within the cuvette. After the localization process, the multi rinse pipettor 1 12 may again aspirate and discard a portion of the sample that was not localized during the localization cycle. [0050] At this point, the R2 pipettor 122 may aspirate a portion of conjugate from the reagent rotor 1 14 and inject the conjugate into the mixed substrate container 124 creating a mixed substrate sample. The R2 pipettor may then aspirate the mixed substrate sample from the mixed substrate container 124 and inject the mixed substrate sample into the cuvette of the reaction rotor 106, resuspending the fluo-bead with the mixed substrate sample. The sample in the cuvette of the reaction rotor 106 may then be aspirated by the optics pipettor 108 and placed in the optics box 1 10. After the optics box makes fluorescence and luminescence optical observations, the sample is discarded and the multi rinse pipettor rinses the cuvettes of the reaction rotor 106 in preparation for the next test.
[0051] Antigens can be proteins, polypeptides, peptides, nucleic acids, lipids, enzymes, immunoglobulins, or nucleotides. As used herein, the term "protein" refers to amino acid sequences longer than 100 amino acids. As used herein, the term "polypeptides" refers to amino acid sequences between 10 and 100 amino acids in length. As used herein, the term "peptides" refers to amino acid sequences less than 10 amino acids in length. As used herein, a "nucleotide" refers to a single purine or pyrimidine molecule while the term "nucleic acid" refers to a nucleotide sequence. For example, DNA is a nucleic acid while guanine is a nucleotide.
[0052] Also disclosed herein are quantitative methods for performing an automated diagnostic assay for detection of autoantigen-specific antibodies in a serum sample from a subject for the diagnosis of an autoimmune disease.
[0053] Non-limiting examples of autoimmune disorders that can be diagnosed using the method disclosed herein include acute disseminated encephalomyelitis (ADEM), Addison's disease, an anti-phospholipid antibody syndrome (APS), arthritis (e.g., monoarthritis, oligoarthritis, or a polyarthritis like osteoarthritis, rheumatoid arthritis, juvenile idiopathic arthritis, septic arthritis) spondyloarthropathy, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune inner ear disease, bullous pemphigoid, celiac disease, Chagas disease, chronic obstructive pulmonary disease (COPD), diabetes mellitus type 1 (IDDM), endometriosis, a gastrointestinal disorder (e.g., irritable bowel disease or an inflammatory bowel disease like Crohn's disease or ulcerative colitis), Goodpasture's syndrome, Graves' disease, Guillain-Barre syndrome (GBS), Hashimoto's thyroiditis, hidradenitis suppurativa, idiopathic thrombocytopenic purpura, interstitial cystitis, lupus (e.g., discoid lupus erythematosus, drug-induced lupus erythematosus, lupus nephritis, neonatal lupus, subacute cutaneous lupus erythematosus, systemic lupus erythematosus), morphea, multiple sclerosis (MS), myasthenia gravis, myopathy (e.g., dermatomyositis, inclusion body myositis, polymyositis, myositis), neuromyotonia, Parkinson's disease, pemphigus vulgaris, pernicious anaemia, primary biliary cirrhosis, psoriasis, recurrent disseminated encephalomyelitis, rheumatic fever, schizophrenia, scleroderma, Sjogren's syndrome, a skin disorder (e.g., dermatitis, eczema, statis dermatitis, hidradenitis suppurativa, psoriasis, rosacea or a scleroderma), tenosynovitis, uveitis, vasculitis (e.g., Buerger's disease, cerebral vasculitis, Churg-Strauss arteritis, cryoglobulinemia, essential cryoglobulinemic vasculitis, giant cell arteritis, Golfer's vasculitis, Henoch-Schonlein purpura, hypersensitivity vasculitis, Kawasaki disease, microscopic polyarteritis/polyangiitis, polyarteritis nodosa, polymyalgia rheumatica (PMR), rheumatoid vasculitis, Takayasu arteritis, Wegener's granulomatosis), or vitiligo.
[0054] Non-limiting examples of autoantigens include, but are not limited to, aggrecan, alanyl-tRNA syntetase (PL-12), alpha beta crystallin, alpha fodrin (Sptan 1 ), alpha-actinin, a1 antichymotrypsin, a1 antitripsin, a1 microglobulin, alsolase, aminoacyl-tRNA synthetase, amyloid (e.g., amyloid beta, amyloid P), annexins (e.g., annexin II, annexin V), apolipoproteins (e.g., ApoB, ApoE, ApoE4, ApoJ), aquaporin (e.g., AQP1 , AQP2, AQP3, AQP4), bactericidal/permeability-increasing protein (BPI), β-globin precursor BP1 , β-actin, β- lactoglobulin A, 32-glycoprotein I, 32-microglobulin, blood group antigens (e.g., Rh blood group antigens, I blood group antigens, ABO blood group antigens), C reactive protein (CRP), calmodulin, calreticulin, cardiolipin, catalase, cathepsin B, centromere proteins (e.g., CENP-A, CENP-B), chondroitin sulfate, chromatin, collagen (e.g., types I, II, III, IV, V, VI collagen), complement components (e.g., C1q, C3, C3a, C3b, C4, C5, C6, C7, C8, C9), cytochrome C, cytochrome P450 2D6, cytokeratins, decorin, dermatan sulfate, DNA (e.g., double stranded DNA, single stranded DNA), DNA topoisomerase I, elastin, Epstein-Barr nuclear antigen 1 (EBNA1 ), elastin, entaktin, extractable nuclear antigens (Ro, La, Sm, RNP, Scl-70, Jo1 ), Factor I, Factor P, Factor B, Factor D, Factor H, Factor X, fibrinogen (e.g., fibrinogen IV, fibrinogen S), fibronectin, formiminotransferase cyclodeaminase (LC-1 ), gliadin and amidated gliadin peptides (DGPs), gp210 nuclear envelope protein, GP2 (major zymogen granule membrane glycoprotein), glycoprotein gpllb/llla, glial fibrillary acidic protein (GFAP), glycated albumin, glyceraldehyde 3-phosphate dehydrogenase (GAPDH), haptoglobin A2, heat shock proteins (e.g., Hsp60, HSP70), hemocyanin, heparin, histones (e.g., histones H1 , H2A, H2B, H3, H4), histidyl-tRNA synthetase (Jo-1 ), hyaluronidase, immunoglobulins, insulin, insulin receptor, integrins (e.g., integrins α1 βι , α2βι, α3βι, α4βι, α5βι, α6βι, α7βι, αι_β2, αΜβ2, α^β3, ανβι , ανβ3, ανβ5, ανβ6, ανβ8, α6β3, αιβ , interstitial retinol- binding protein 3, intrinsic factor, Ku (p70/p80), lactate dehydrogenase, laminin, liver cytosol antigen type 1 (LC1 ), liver/kidney microsomal antigen 1 (LKM1 ), lysozyme, melanoma differentiation-associated protein 5 (MDA5), Mi-2 (chromodomain helicase DNA binding protein 4), mitochondrial proteins (e.g., M1 , M2, M3, M4, M5, M6, M7, M8, M9, BCOADC-E2, OGDC-E2, PDC-E2), muscarinic receptors, myelin-associated glycoprotein, myosin, myelin basic protein, myelin oligodendrocyte glycoprotein, myeloperoxidase (MPO), rheumatoid factor (IgM anti-lgG), neuron-specific enolase, nicotinic acetylcholine receptor a chain, nucleolin, nucleoporin (e.g., Nup62), nucleosome antigen, PM/ScM OO, PM/Scl 75, pancreatic β-cell antigen, pepsinogen, peroxiredoxin 1 , phosphoglucose isomerase, phospholipids, phosphotidyl inositol, platelet derived growth factors, polymerase beta (POLB), potassium channel KIR4.1 , proliferating cell nuclear antigen (PCNA), proteinase-3, proteolipid protein, proteoglycan, prothrombin, recoverin, rhodopsin, ribonuclease, ribonucleoproteins (e.g., Ro, La, snRNP, scRNP), ribosomes, ribosomal phosphoproteins (e.g., P0, P1 , P2), RNA (double stranded RNA, single stranded RNA), Sm proteins (e.g., SmB, SmB', SmD1 , SmD2, SmD3, SmF, SmG, SmN), Sp100 nuclear protein, SRP54 (signal recognition particle 54 kDa), selectin, smooth muscle proteins, sphinomyelin, streptococcal antigens, superoxide dismutase, synovial joint proteins, T1 F1 gamma collagen, threonyl-tRNA synthetase (PL-7), tissue transglutaminase, thyroid peroxidase, thyroglobulin, thyroid stimulating hormone receptor, transferrin, triosephosphate isomerase, tubulin, tumor necrosis alpha, topoisomerase, U1 -dnRNP 68/70 kDa, U1 -snRNP A, U 1 -snRNP C, U-snRNP B/B', ubiquitin, vascular endothelial growth factor, vimentin, and vitronectin.
[0055] Also disclosed herein are quantitative methods for performing an automated diagnostic assay for detection of an infectious disease by detecting antibodies to the infectious agent in a serum sample from a subject for the diagnosis of an autoimmune disease.
[0056] Infectious agents include, but are not limited to bacteria, virus, viroids, prions, nemotodes (e.g., roundworms, pinworms), parasites (malaria, tapeworm), and fungi (e.g., yeast, ringworm). As used herein the terms "infectious agent," "pathogen", "pathogenic microorganism" and "microorganism" all refer to the infectious agents listed above.
[0057] Antigens from infectious agents can be any isolated protein, glycoprotein, nucleic acid, enzyme, lipid, liposaccharide, or combination thereof, from an infectious agent. Antigens can also include extracts or homogenized preparations from infectious agents which include a plurality of different antigenic moieties.
[0058] In order to determine if a sample contains autoimmune antibodies to self antigens (autoantigens), or antibodies to an infectious agent, fluorescently labeled paramagnetic microparticles (e.g., fluo-beads) are bound to the autoantigen or infectious disease antigen according to methods disclosed herein to form a capture reagent. A sample of the cuvette is obtained prior to washing for a fluorescent reading to determination of the starting number of magnetic beads in the reaction cuvette. [0059] After undergoing a washing process, the patient sample, and optionally a diluent if needed, is added to the particles in the cuvettes and incubated. This results in the capture of specific analyte molecules (antibodies) in the patient's blood sample. Another washing process is then performed to remove any excess or unbound sample, and then a luminescent label and a conjugate is added to the cuvette. When added to the cuvette, it can be expected that some portion of the conjugate will bind to the capture reagent/sample complex on the paramagnetic particles after an incubation period. The particles then undergo another wash process to remove any unbound conjugate, and then the substrate is added to the cuvette and incubated for a short period of time to allow the chemiluminescent glow reaction to reach equilibrium.
[0060] After equilibrium is reached, luminescence and fluorescence readings of the sample are taken and the levels of antibodies to the antigen are determined as disclosed herein.
[0061] Advantages and improvements of the processes, methods of the present disclosure are demonstrated in the following examples. These examples are illustrative only and are not intended to limit or preclude other embodiments of the present disclosure.
EXAMPLE 1 : Biotinylation of anti-human Ig or antigen extracts:
[0062] Two microliters of NHS-PEG12-Biotin (Pierce) 250 mM in DMSO is added to 1 ml_ of affinity purified anti-human Ig (e.g., IgE, IgG, IgA, IgM) 5.0 mg/mL in phosphate buffered saline (PBS). Or, 1.6 μΙ_ of NHS-PEG12-Biotin (Pierce) 250 mM in DMSO is added to 1 ml_ of antigen extracts 1.0 mg/mL in PBS.
[0063] The reagent solution is mixed and placed on ice for 2 hours. Free biotin reagent is separated from the biotinylated antibody by dialysis against two changes of PBS (volume ratio of antibody to buffer - 1 : 100) at 2-8 °C for 4 hours and overnight.
EXAMPLE 2: Preparation of Fluo-Bead:
[0064] Five microliters of Biotin-Fluo (Alexa Fluor 594 Biocytin, Sodium Salt, Life Technologies) 1 mM in ddH20 is added into 45 mL of PBSTHP Buffer (10 mM sodium phosphate, pH 7.4, 0.9% (w/v) NaCI, 0.05% (v/v) Tween-20, 10 mg/mL HSA, 1 % (v/v) ProClin 950). Mix well.
[0065] Five milliliters of SA-Speed Bead (Sera-mag Speedbeads Streptavidin-Coated Magnetic Particles, Thermo) 10 mg/mL is added into the Biotin-Fluo solution and mixed well.
EXAMPLE 3: Assay for specific immunoglobulin levels to antigens
[0066] Ten microliters Fluo-Bead (Fluorescence labeled para-magnetic microparticles) at bead concentration 1 mg/mL is dispensed into the reaction cuvette; 40 μί of biotin-antigen (e.g., infectious disease antigen, autoantigen, etc) or biotin-anti-lg antibody, is dispensed and mixed into the Fluo-Bead, and incubated for 1-10 min at 37°C. After washing, antigen- or anti-lg-coated beads are resuspended in 40 μΙ_ of reaction buffer. Serum samples obtained from appropriate individuals are assayed against the antigens. A 10 μΙ_ sample was added to 40 μΙ_ of suspended antigen-coated beads in reaction cuvette. For the six point standard curve, 10 μΙ_ of serum standards (secondary standards calibrated against a WHO Ig Standard) are each added to 40 μΙ_ of anti-lg-coated beads in a reaction cuvette. While various different labeled anti-lg conjugates can be utilized in accordance with the present teachings, in accordance with certain teachings, the following anti-lg conjugates are utilized: for assays: anti-lgE-HRP; anti-lgA-HRP, anti-lgG-HRP, anti-lgM-HRP, anti-ECP-HRP, and anti-tryptase-HRP. Moreover, as used herein, each conjugate has an optimized HRP incorporation ratio for use in the chemistry. In accordance with certain aspects of the present teachings, the range of HRP incorporation ratio used for the listed conjugates is between about 1.2 and about 5.4. In addition, the present teachings also contemplate the incorporation of other types of conjugate-reporter systems including, but not limited to: alkaline phosphatase conjugate and b-galactosidase conjugate.
[0067] The solutions are mixed and incubated for 40 min at 37°C. After washing, beads are resuspended in 50 μΙ_ of anti-human IgE-HRP conjugate, and incubated at 37°C for 30 min. 50 μΙ_ of PS-atto (Lumigen) is added into each cuvette and the beads resuspended. The bead suspension is transferred into a pipette tip and read in the optic box for both fluorescence and luminescence signal. The standard curve was determined using a four parameter logistic function equation and levels of specific Ig to antigens interpolated from the standard curve.
[0068] A list of illustrative reagents and components that may be used in accordance with the present teachings include, but are not necessarily limited to: Bead: Fluo-Bead (SA- Speed Bead, Atto 590 labeled), 1 mg/mL; Capture Reagent Diluent: IgE: 10 mM sodium phosphate, pH 7.4, 0.9% (w/v) NaCI, 0.05% Tween-20, 1 % (w/v) human serum albumin, 1 %(v/v) ProClin 950, up to 5% (v/v) glycerol; ANA: 10 mM sodium phosphate, pH 7.4, 0.9% (w/v) NaCI, 0.05% Tween-20, 1 % (w/v) bovine serum albumin, 1 % Protease inhibitor cocktail, 0.1 mM DTT, 1 %(v/v) ProClin 950, 25% (up to 30%) (v/v) glycerol; Wash Buffer Concentrate (5*): 50 mM sodium phosphate, pH 7.4, 4.5% (w/v) NaCI, 0.05% Tween-20, 0.05%(v/v) ProClin 950, 0.02% (v/v) Antifoam-C v/v; Reagent Diluent (Reaction Diluent & Sample Diluent) IgE -10 mM sodium phosphate, pH 7.4, 500 mM NaCI, 0.02% Tween-20, 1 % (w/v) human serum albumin, 1 % (v/v) human IgG, 1 %(v/v) ProClin 950, 0.005% Antifoam-B v/v, 2% (w/v) PEG 6,000; ANA -10 mM sodium phosphate, pH 7.4, 500 mM NaCI, 0.02% Tween-20, 25% (w/v) human serum albumin, 1 %(v/v) ProClin 950; Calibrator s Control: Calibrator: Patient sample diluted into Sample Diluent; Control: Patient Sample pool; Conjugate: Conjugate Diluent: 50 mM sodium phosphate, pH 6.7, 150 mM NaCI, 0.05% Tween-20, 1 % BSA, 5% (w/v) PEG 6,000, 1 %(v/v) ProClin 950; IgE :100 ng/mL anti-lgE- HRP, 100 μg mL apo-HRP, in diluent, 0.015% Antifoam-B v/v; and Substrate: PS-atto A & B, 0.01 % Antifoam-B v/v.
EXAMPLE 4: Assay for specific immunoglobulin levels to autoantiqens
[0069] Ten microliters Fluo-Bead (Fluorescence labeled para-magnetic microparticles) at bead concentration 1 mg/mL is dispensed into the reaction cuvette; 40 μΙ_ of biotin-antigen (e.g., myelin basic protein, collagen, insulin, etc) or biotin-anti-lgG antibody (or alternatively, anti-lgM or anti-lgA), is dispensed and mixed into the Fluo-Bead, and incubated for 1-10 min at 37°C. A sample of the labeled Fluo-Bead is obtained to determine the starting number of beads. After washing, antigen- or anti-lgG-coated beads are resuspended in 40 μΙ_ of reaction buffer. Serum samples obtained from individuals are assayed against antigens. A 10 μΙ_ sample is added to 40 μΙ_ of suspended antigen-coated beads in reaction cuvette. For the six point standard curve, 10 μΙ_ of serum standards (secondary standards calibrated against the WHO IgG Standard) are each added to 40 μΙ_ of anti-lgG-coated beads in a reaction cuvette.
[0070] The solutions are mixed and incubated for 40 min at 37°C. After washing, beads are resuspended in 50 μΙ_ of anti-human IgG-HRP conjugate, and incubated at 37°C for 30 min. 50 μΙ_ of PS-atto (Lumigen) is added into each cuvette and the beads resuspended. The bead suspension is transferred into a pipette tip and read in the optic box for both fluorescence and luminescence signal. The standard curve is determined using a four parameter logistic function equation and levels of specific IgG to the autoantigens interpolated from the standard curve.
EXAMPLE 5: Assay for specific IgG levels to infectious agent antigens
[0071] Ten microliters Fluo-Bead (Fluorescence labeled para-magnetic microparticles) at bead concentration 1 mg/mL is dispensed into the reaction cuvette; 40 μί of biotin-antigen (from e.g., human immunodeficiency virus, Ebola virus, Mycobacterium tuberculosis, etc) or biotin-anti-lgG antibody (or alternatively, anti-lgM or anti-lgA), is dispensed and mixed into the Fluo-Bead, and incubated for 1 -10 min at 37°C. A sample of the labeled Fluo-Bead is obtained to determine the starting number of beads. After washing, antigen- or anti-lgG- coated beads are resuspended in 40 μί of reaction buffer. Serum samples obtained from individuals are assayed against antigens. A 10 μί sample is added to 40 μί of suspended antigen-coated beads in reaction cuvette. For the six point standard curve, 10 μί of serum standards (secondary standards calibrated against the WHO IgG Standard) are each added to 40 \}L of anti-lgG-coated beads in a reaction cuvette. [0072] The solutions are mixed and incubated for 40 min at 37°C. After washing, beads are resuspended in 50 μΙ_ of anti-human IgG-HRP conjugate, and incubated at 37°C for 30 min. 50 μΙ_ of PS-atto (Lumigen) is added into each cuvette and the beads resuspended. The bead suspension is transferred into a pipette tip and read in the optic box for both fluorescence and luminescence signal. The standard curve is determined using a four parameter logistic function equation and levels of specific IgG to the infectious agent antigens interpolated from the standard curve.
[0073] Unless otherwise indicated, all numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, and so forth used in the specification and claims are to be understood as being modified in all instances by the term "about." As used herein the terms "about" and "approximately" means within 10 to 15%, preferably within 5 to 10%. Accordingly, unless indicated to the contrary, the numerical parameters set forth in the specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained by the present invention. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. Notwithstanding that the numerical ranges and parameters setting forth the broad scope of the invention are approximations, the numerical values set forth in the specific examples are reported as precisely as possible. Any numerical value, however, inherently contains certain errors necessarily resulting from the standard deviation found in their respective testing measurements.
[0074] The terms "a," "an," "the" and similar referents used in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each individual value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., "such as") provided herein is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention otherwise claimed. No language in the specification should be construed as indicating any non-claimed element essential to the practice of the invention.
[0075] Groupings of alternative elements or embodiments of the invention disclosed herein are not to be construed as limitations. Each group member may be referred to and claimed individually or in any combination with other members of the group or other elements found herein. It is anticipated that one or more members of a group may be included in, or deleted from, a group for reasons of convenience and/or patentability. When any such inclusion or deletion occurs, the specification is deemed to contain the group as modified thus fulfilling the written description of all Markush groups used in the appended claims.
[0076] Certain embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Of course, variations on these described embodiments will become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventor expects skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.
[0077] Specific embodiments disclosed herein may be further limited in the claims using consisting of or consisting essentially of language. When used in the claims, whether as filed or added per amendment, the transition term "consisting of" excludes any element, step, or ingredient not specified in the claims. The transition term "consisting essentially of" limits the scope of a claim to the specified materials or steps and those that do not materially affect the basic and novel characteristic(s). Embodiments of the invention so claimed are inherently or expressly described and enabled herein.
[0078] Furthermore, numerous references have been made to patents and printed publications throughout this specification. Each of the above-cited references and printed publications are individually incorporated herein by reference in their entirety.
[0079] In closing, it is to be understood that the embodiments of the invention disclosed herein are illustrative of the principles of the present invention. Other modifications that may be employed are within the scope of the invention. Thus, by way of example, but not of limitation, alternative configurations of the present invention may be utilized in accordance with the teachings herein. Accordingly, the present invention is not limited to that precisely as shown and described.

Claims

What is claimed is:
1. A quantitative method for performing an automated diagnostic assay for an autoimmune disease comprising:
incubating a capture reagent with a streptavidin-coated medium to form a solid phase complex, wherein the capture reagent is a biotinylated autoantigen;
washing the solid phase complex to remove excess capture reagent;
incubating the solid phase complex with a serum sample to form an immune complex;
washing the immune complex to remove any unbound sample; incubating the immune complex with a conjugate to create an immune- conjugate complex;
washing the immune-conjugate complex to remove any unbound conjugate; introducing a substrate capable of generating a quantifiable response; and calibrating the response generated from the substrate.
2. The method of claim 1 , wherein the step of incubating the solid phase complex with the serum sample comprises binding an autoimmune-specific human immunoglobulin G (IgG), an autoimmune-specific human immunoglobulin M (IgM) or an autoimmune-specific human immunoglobulin A (IgA) present in the serum sample to the capture reagent.
3. The method of claim 1 , wherein the autoantigen is aggrecan, alanyl-tRNA syntetase (PL-12), alpha beta crystallin, alpha fodrin (Sptan 1 ), alpha-actinin, a1
antichymotrypsin, a1 antitripsin, a1 microglobulin, alsolase, aminoacyl-tRNA synthetase, amyloid (e.g., amyloid beta, amyloid P), annexins (e.g., annexin II, annexin V),
apolipoproteins (e.g., ApoB, ApoE, ApoE4, ApoJ), aquaporin (e.g., AQP1 , AQP2, AQP3, AQP4), bactericidal/permeability-increasing protein (BPI), β-globin precursor BP1 , β-actin, β- lactoglobulin A, 32-glycoprotein I, β2-microglobulin, blood group antigens (e.g., Rh blood group antigens, I blood group antigens, ABO blood group antigens), C reactive protein (CRP), calmodulin, calreticulin, cardiolipin, catalase, cathepsin B, centromere proteins (e.g., CENP-A, CENP-B), chondroitin sulfate, chromatin, collagen (e.g., types I, II, III, IV, V, VI collagen), complement components (e.g., C1q, C3, C3a, C3b, C4, C5, C6, C7, C8, C9), cytochrome C, cytochrome P450 2D6, cytokeratins, decorin, dermatan sulfate, DNA (e.g., double stranded DNA, single stranded DNA), DNA topoisomerase I, elastin, Epstein-Barr nuclear antigen 1 (EBNA1 ), elastin, entaktin, extractable nuclear antigens (Ro, La, Sm, RNP, Scl-70, Jo1 ), Factor I, Factor P, Factor B, Factor D, Factor H, Factor X, fibrinogen (e.g., fibrinogen IV, fibrinogen S), fibronectin, formiminotransferase cyclodeaminase (LC-1 ), gliadin and amidated gliadin peptides (DGPs), gp210 nuclear envelope protein, GP2 (major zymogen granule membrane glycoprotein), glycoprotein gpllb/llla, glial fibrillary acidic protein (GFAP), glycated albumin, glyceraldehyde 3-phosphate dehydrogenase (GAPDH), haptoglobin A2, heat shock proteins (e.g., Hsp60, HSP70), hemocyanin, heparin, histones (e.g., histones H1 , H2A, H2B, H3, H4), histidyl-tRNA synthetase (Jo-1 ), hyaluronidase, immunoglobulins, insulin, insulin receptor, integrins (e.g., integrins α1 β1 , α2βι, α3βι, α4βι, α5βι, α6βι, α7βι , αι_β2, αΜβ2, α,^β3, ανβι, ανβ3, ανβ5, ανβ6, ανβ8, α6β3, αιβ , interstitial retinol- binding protein 3, intrinsic factor, Ku (p70/p80), lactate dehydrogenase, laminin, liver cytosol antigen type 1 (LC1 ), liver/kidney microsomal antigen 1 (LKM1 ), lysozyme, melanoma differentiation-associated protein 5 (MDA5), Mi-2 (chromodomain helicase DNA binding protein 4), mitochondrial proteins (e.g., M1 , M2, M3, M4, M5, M6, M7, M8, M9, BCOADC-E2, OGDC-E2, PDC-E2), muscarinic receptors, myelin-associated glycoprotein, myosin, myelin basic protein, myelin oligodendrocyte glycoprotein, myeloperoxidase (MPO), rheumatoid factor (IgM anti-lgG), neuron-specific enolase, nicotinic acetylcholine receptor a chain, nucleolin, nucleoporin (e.g., Nup62), nucleosome antigen, PM/Scl100, PM/Scl 75, pancreatic β-cell antigen, pepsinogen, peroxiredoxin 1 , phosphoglucose isomerase, phospholipids, phosphotidyl inositol, platelet derived growth factors, polymerase beta (POLB), potassium channel KIR4.1 , proliferating cell nuclear antigen (PCNA), proteinase-3, proteolipid protein, proteoglycan, prothrombin, recoverin, rhodopsin, ribonuclease, ribonucleoproteins (e.g., Ro, La, snRNP, scRNP), ribosomes, ribosomal phosphoproteins (e.g., P0, P1 , P2), RNA (double stranded RNA, single stranded RNA), Sm proteins (e.g., SmB, SmB', SmD1 , SmD2, SmD3, SmF, SmG, SmN), Sp100 nuclear protein, SRP54 (signal recognition particle 54 kDa), selectin, smooth muscle proteins, sphinomyelin, streptococcal antigens, superoxide dismutase, synovial joint proteins, T1 F1 gamma collagen, threonyl-tRNA synthetase (PL-7), tissue transglutaminase, thyroid peroxidase, thyroglobulin, thyroid stimulating hormone receptor, transferrin, triosephosphate isomerase, tubulin, tumor necrosis alpha,
topoisomerase, U1-dnRNP 68/70 kDa, U1-snRNP A, U1-snRNP C, U-snRNP B/B', ubiquitin, vascular endothelial growth factor, vimentin, or vitronectin
4. The method of claim 1 , wherein the streptavidin-coated medium comprises a universal fluorescent-labeled magnetic microparticle.
5. The method of claim 1 , wherein one or more of the washing steps include washing the complexes by magnetically sequestering the complex being washed within a confined area of a reaction cuvette.
6. The method of claim 1 , wherein the step of incubating a capture reagent with a streptavidin-coated medium includes incubating a capture reagent that is retained in a suspension by a reaction diluent including a high concentration of human serum albumin (HSA).
7. The method of claim 1 , wherein the step incubating the immune complex with a conjugate comprises incubating an immune complex that is retained in a suspension by a conjugate diluent including a nominal concentration of polyethylene glycol.
8. The method of claim 1 , further comprising the step of using horseradish peroxidase (HRP) as an indirect label when washing the immune complex to remove unbound sample.
9. The method of claim 1 , further comprising the step of adjusting the quantifiable response for bead retention by:
transferring the substrate and immune-conjugate complex to an optics box, wherein both fluorescent and chemiluminescent signals are quantified; and
employing a ratio of initial to final fluorescence to adjust the quantified
chemiluminescent signal to calculate a reported value.
10. The method of claim 9, further comprising:
measuring fluorescence within the optics box to determine bead retention; and
measuring luminescence within the optics box to detect a generated relative light unit signal.
1 1. The method of claim 10, further comprising entering the fluorescence and luminescence measurements into an algorithm to generate a bead retention adjusted relative light unit signal.
12. The method of claim 10, further comprising comparing the generated bead retention adjusted relative light unit signal to a calibration curve relative light unit signal.
13. A quantitative method for performing an automated diagnostic assay for an infectious disease comprising:
incubating a capture reagent with a streptavidin-coated medium to form a solid phase complex, wherein the capture reagent is a biotinylated infectious disease antigen;
washing the solid phase complex to remove excess capture reagent;
incubating the solid phase complex with a serum sample to form an immune complex;
washing the immune complex to remove any unbound sample; incubating the immune complex with a conjugate to create an immune- conjugate complex;
washing the immune-conjugate complex to remove any unbound conjugate; introducing a substrate capable of generating a quantifiable response; and calibrating the response generated from the substrate.
14. The method of claim 13, wherein the step of incubating the solid phase complex with the serum sample comprising binding an infectious agent-specific human IgG, IgM, or IgA present in the serum sample to the biotinylated capture reagent.
15. The method of claim 13, wherein the infectious disease antigen is from an infectious agent selected from bacteria, virus, viroids, prions, nemotodes, parasites, and fungi.
16. The method of claim 13, wherein the capture reagent is derived from the biotinylation of an infectious agent extract comprised of a multiplicity of antigens.
17. The method of claim 13, wherein the infectious disease antigen is a protein, glycoprotein, nucleic acid, enzyme, lipid, liposaccharide, antibodies, or combination thereof.
18. The method of claim 13, wherein the capture reagent is an amalgam of multiple biotinylated capture reagents selected from purified proteins, enzymes, antibodies, and infectious agent extracts.
19. The method of claim 13, wherein the streptavidin-coated medium comprises a universal fluorescent-labeled magnetic microparticle.
20. The method of claim 13, wherein one or more of the washing steps include washing the complexes by magnetically sequestering the complex being washed within a confined area of a reaction cuvette.
21. The method of claim 13, wherein the step of incubating a capture reagent with a streptavidin-coated medium includes incubating a capture reagent that is retained in a suspension by a reaction diluent including a high concentration of human serum albumin (HSA).
22. The method of claim 13, wherein the step incubating the immune complex with a conjugate comprises incubating an immune complex that is retained in a suspension by a conjugate diluent including a nominal concentration of polyethylene glycol.
23 The method of claim 13, further comprising the step of using horseradish peroxidase (HRP) as an indirect label when washing the immune complex to remove unbound sample.
24. The method of claim 13, further comprising the step of adjusting the quantifiable response for bead retention by:
transferring the substrate and immune-conjugate complex to an optics box, wherein both fluorescent and chemiluminescent signals are quantified; and employing a ratio of initial to final fluorescence to adjust the quantified
chemiluminescent signal to calculate a reported value.
25. The method of claim 24, further comprising:
measuring fluorescence within the optics box to determine bead retention; and
measuring luminescence within the optics box to detect a generated relative light unit signal.
26. The method of claim 25, further comprising entering the fluorescence and luminescence measurements into an algorithm to generate a bead retention adjusted relative light unit signal.
27. The method of claim 25, further comprising comparing the generated bead retention adjusted relative light unit signal to a calibration curve relative light unit signal.
PCT/US2015/023408 2015-03-30 2015-03-30 Automated immunoanalyzer system for performing diagnostic assays for autoimmune and infectious diseases WO2016159960A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
EP15887969.2A EP3278109A4 (en) 2015-03-30 2015-03-30 Automated immunoanalyzer system for performing diagnostic assays for autoimmune and infectious diseases
AU2015389978A AU2015389978A1 (en) 2015-03-30 2015-03-30 Automated immunoanalyzer system for performing diagnostic assays for autoimmune and infectious diseases
CN201580078733.XA CN107548461A (en) 2015-03-30 2015-03-30 Immunoassay instrument system for the automation of the diagnostic measure that performs autoimmune disease and infectious diseases
CA2981455A CA2981455A1 (en) 2015-03-30 2015-03-30 Automated immunoanalyzer system for performing diagnostic assays for autoimmune and infectious diseases
JP2017551241A JP2018510354A (en) 2015-03-30 2015-03-30 Automated immunoassay system for performing diagnostic assays for autoimmunity and infectious diseases
PCT/US2015/023408 WO2016159960A1 (en) 2015-03-30 2015-03-30 Automated immunoanalyzer system for performing diagnostic assays for autoimmune and infectious diseases
HK18108131.3A HK1248804A1 (en) 2015-03-30 2018-06-25 Automated immunoanalyzer system for performing diagnostic assays for autoimmune and infectious diseases

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2015/023408 WO2016159960A1 (en) 2015-03-30 2015-03-30 Automated immunoanalyzer system for performing diagnostic assays for autoimmune and infectious diseases

Publications (1)

Publication Number Publication Date
WO2016159960A1 true WO2016159960A1 (en) 2016-10-06

Family

ID=57004502

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/023408 WO2016159960A1 (en) 2015-03-30 2015-03-30 Automated immunoanalyzer system for performing diagnostic assays for autoimmune and infectious diseases

Country Status (7)

Country Link
EP (1) EP3278109A4 (en)
JP (1) JP2018510354A (en)
CN (1) CN107548461A (en)
AU (1) AU2015389978A1 (en)
CA (1) CA2981455A1 (en)
HK (1) HK1248804A1 (en)
WO (1) WO2016159960A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017015662A1 (en) 2015-07-23 2017-01-26 Hycor Biomedical, Inc On-board kitting
CN107022548A (en) * 2017-03-22 2017-08-08 陕西脉元生物科技有限公司 A kind of anti-AQP4 autoantibodies detection material of human body and preparation method thereof
CN107870239A (en) * 2017-09-14 2018-04-03 上海长海医院 Application of the ECD albumen of nAChR α 1 in medical treatment detects
EP3358355A1 (en) 2017-02-04 2018-08-08 Warszawski Uniwersytet Medyczny Use of serum 2-cysteine peroxiredoxins (2-cys-prdx) as biomarkers of chronic kidney diseases (ckd) such as lupus nephritis (ln), iga nephropathy (igan) and autosomal-dominant polycystic kidney disease (adpkd) useful for diagnosing, monitoring, and prognosing in these diseases and method of differentiation of these diseases
CN109142743A (en) * 2018-07-25 2019-01-04 滴准生物科技(常州)有限公司 A kind of multinomial detection kit for exempting from liver antibody certainly
JP2019537717A (en) * 2016-11-01 2019-12-26 ハイコア バイオメディカル エルエルシー An immunoassay system that can propose assays based on input data
US10928399B2 (en) 2015-07-15 2021-02-23 Hycor Biomedical, Llc Customizable instrument
CN112505319A (en) * 2020-10-26 2021-03-16 绍兴梅奥心磁医疗科技有限公司 Immune quantitative detection device, detection method and application of to-be-detected marker
CN112710833A (en) * 2021-01-13 2021-04-27 上海交通大学 Cell capture method based on microtubule fluidic chip
CN113125422A (en) * 2021-04-16 2021-07-16 合肥工业大学 Preparation method of chemiluminescent hydrogel microspheres, prepared hydrogel microspheres and application thereof
CN113702647A (en) * 2021-08-31 2021-11-26 普十生物科技(北京)有限公司 Human growth differentiation factor 15 instant detection kit, preparation method and application thereof
WO2023087942A1 (en) * 2021-11-18 2023-05-25 上海北昂医药科技股份有限公司 Delisa sample capable of increasing proportion of effective microbeads, and preparation and detection method therefor

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108440666B (en) * 2018-03-07 2021-03-23 深圳市伯劳特生物制品有限公司 Biotinylated insulin antigen and biotinylation process thereof
AU2019274450A1 (en) * 2018-05-22 2020-10-29 AllerGenis LLC Peptides and methods for detecting peanut allergies
CN109142750B (en) * 2018-08-14 2021-10-22 江苏浩欧博生物医药股份有限公司 Kit for determining anti-histone antibody IgG and detection method
CN109212191A (en) * 2018-09-27 2019-01-15 郑州安图生物工程股份有限公司 It is a kind of for detecting the kit of cytokeratin 19 fragment
CN110320087B (en) * 2019-07-30 2021-03-02 河南赛诺特生物技术有限公司 Pigment-containing tissue immunohistochemical staining kit and staining method
CN110596371B (en) * 2019-09-20 2023-04-07 郑州安图生物工程股份有限公司 Method for evaluating magnetic bead recovery rate and reagent needle precision of full-automatic chemiluminescence determinator and kit
CN111007256A (en) * 2019-12-10 2020-04-14 迪瑞医疗科技股份有限公司 Calmodulin chemiluminescence immunoassay kit and preparation method thereof
CN111272999B (en) * 2020-02-17 2022-11-22 珠海丽珠试剂股份有限公司 Antigen-coupled magnetic particle, preparation method thereof, detection method of anti-U1-snRNP antibody and kit
CN112485432A (en) * 2020-10-26 2021-03-12 绍兴梅奥心磁医疗科技有限公司 Immune quantitative detection device, detection method and application of cardiovascular disease marker
CN113447658B (en) * 2021-07-01 2022-04-19 浙江大学 Kit for detecting anti-peroxiredoxin-1-IgG antibody
CN114895024A (en) * 2021-07-01 2022-08-12 浙江大学 Kit for detecting anti-serine/arginine-rich splicing factor 9-IgG antibody
CN114594245A (en) * 2022-01-26 2022-06-07 浙江莱阅病理诊断科技有限公司 Cleaning buffer solution adapted to full-automatic immunohistochemical dyeing machine and preparation method thereof
CN116148463A (en) * 2022-09-08 2023-05-23 浙江省血液中心 Magnetic bead-antibody-membrane glycoprotein complex, application and kit
CN115656154A (en) * 2022-10-26 2023-01-31 广东优尼德生物科技有限公司 anti-RNP antibody chemiluminescence detection kit based on recombinant RNP multiple antigens

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110151582A1 (en) * 2008-09-02 2011-06-23 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Method for detection of antigen-specific antibodies in biological samples
US20140274784A1 (en) * 2013-03-15 2014-09-18 Hycor Biomedical, Inc. Automated immunoanalyzer system for performing diagnostic assays for allergies and autoimmune diseases

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE4418223A1 (en) * 1994-05-25 1995-11-30 Boehringer Mannheim Gmbh Procedure for the detection of the presence of insulin-dependent diabetes mellitus
WO2001003721A1 (en) * 1999-07-12 2001-01-18 Merlin Biomedical & Pharmaceutical, Ltd. Improved assays for detecting human immunodeficiency virus infection
AU2003273219A1 (en) 2002-07-12 2004-02-02 Dana-Farber Cancer Institute, Inc. Novel compositions and methods for the generation of mhc class ii compounds by peptide exchange
EP3325965B1 (en) 2015-07-23 2022-02-09 Hycor Biomedical, Inc. On-board kitting
TW201832726A (en) 2016-11-01 2018-09-16 美商海科生醫有限責任公司 Immunoassay system capable of suggesting assays based on input data

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110151582A1 (en) * 2008-09-02 2011-06-23 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Method for detection of antigen-specific antibodies in biological samples
US20140274784A1 (en) * 2013-03-15 2014-09-18 Hycor Biomedical, Inc. Automated immunoanalyzer system for performing diagnostic assays for allergies and autoimmune diseases

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3278109A1 *

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10928399B2 (en) 2015-07-15 2021-02-23 Hycor Biomedical, Llc Customizable instrument
WO2017015662A1 (en) 2015-07-23 2017-01-26 Hycor Biomedical, Inc On-board kitting
EP3325965A4 (en) * 2015-07-23 2019-02-20 Hycor Biomedical, Inc. On-board kitting
JP7069151B2 (en) 2016-11-01 2022-05-17 ハイコア バイオメディカル エルエルシー An immunoassay system that can propose an assay based on input data
JP2019537717A (en) * 2016-11-01 2019-12-26 ハイコア バイオメディカル エルエルシー An immunoassay system that can propose assays based on input data
EP3358355A1 (en) 2017-02-04 2018-08-08 Warszawski Uniwersytet Medyczny Use of serum 2-cysteine peroxiredoxins (2-cys-prdx) as biomarkers of chronic kidney diseases (ckd) such as lupus nephritis (ln), iga nephropathy (igan) and autosomal-dominant polycystic kidney disease (adpkd) useful for diagnosing, monitoring, and prognosing in these diseases and method of differentiation of these diseases
WO2018141975A1 (en) 2017-02-04 2018-08-09 Warszawski Uniwersytet Medyczny Use of serum 2-cysteine peroxiredoxins (2-cys-prdx) as biomarkers of chronic kidney diseases
CN107022548A (en) * 2017-03-22 2017-08-08 陕西脉元生物科技有限公司 A kind of anti-AQP4 autoantibodies detection material of human body and preparation method thereof
CN107870239B (en) * 2017-09-14 2020-10-23 上海长海医院 Application of nAChR-alpha 1-ECD protein in medical detection
CN107870239A (en) * 2017-09-14 2018-04-03 上海长海医院 Application of the ECD albumen of nAChR α 1 in medical treatment detects
CN109142743A (en) * 2018-07-25 2019-01-04 滴准生物科技(常州)有限公司 A kind of multinomial detection kit for exempting from liver antibody certainly
CN112505319A (en) * 2020-10-26 2021-03-16 绍兴梅奥心磁医疗科技有限公司 Immune quantitative detection device, detection method and application of to-be-detected marker
CN112710833A (en) * 2021-01-13 2021-04-27 上海交通大学 Cell capture method based on microtubule fluidic chip
CN113125422A (en) * 2021-04-16 2021-07-16 合肥工业大学 Preparation method of chemiluminescent hydrogel microspheres, prepared hydrogel microspheres and application thereof
CN113125422B (en) * 2021-04-16 2023-07-25 合肥工业大学 Preparation method of chemiluminescent hydrogel microbead, prepared hydrogel microbead and application thereof
CN113702647A (en) * 2021-08-31 2021-11-26 普十生物科技(北京)有限公司 Human growth differentiation factor 15 instant detection kit, preparation method and application thereof
CN113702647B (en) * 2021-08-31 2024-03-15 普十生物科技(北京)有限公司 Human growth differentiation factor 15 instant detection kit, preparation method and application thereof
WO2023087942A1 (en) * 2021-11-18 2023-05-25 上海北昂医药科技股份有限公司 Delisa sample capable of increasing proportion of effective microbeads, and preparation and detection method therefor

Also Published As

Publication number Publication date
AU2015389978A1 (en) 2017-09-28
EP3278109A1 (en) 2018-02-07
CN107548461A (en) 2018-01-05
JP2018510354A (en) 2018-04-12
EP3278109A4 (en) 2018-08-15
CA2981455A1 (en) 2016-10-06
HK1248804A1 (en) 2018-10-19

Similar Documents

Publication Publication Date Title
US20200371029A1 (en) Automated immunoanalyzer system for performing diagnostic assays for autoimmune and infectious diseases
WO2016159960A1 (en) Automated immunoanalyzer system for performing diagnostic assays for autoimmune and infectious diseases
JP5005511B2 (en) Immunodiagnostics with reduced non-specific reactions
EP3829759A1 (en) Method for detecting biomarkers
EP3325965B1 (en) On-board kitting
US20220260557A1 (en) Methods for depletion and enrichment
RU2132070C1 (en) Method of chemiluminescent compound-assisted detection of antibody in sample and method of measuring concentration and/or relative content of specific antibody in sample
JP2024019568A (en) Enrichment of antigen-specific antibodies for analytical and therapeutic use

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: 15887969

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2015389978

Country of ref document: AU

Date of ref document: 20150330

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2981455

Country of ref document: CA

Ref document number: 2017551241

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2015887969

Country of ref document: EP