WO1993021346A1 - Dosage pour la detection de l'antigene du vih et de l'anticorps du vih - Google Patents

Dosage pour la detection de l'antigene du vih et de l'anticorps du vih Download PDF

Info

Publication number
WO1993021346A1
WO1993021346A1 PCT/US1993/003224 US9303224W WO9321346A1 WO 1993021346 A1 WO1993021346 A1 WO 1993021346A1 US 9303224 W US9303224 W US 9303224W WO 9321346 A1 WO9321346 A1 WO 9321346A1
Authority
WO
WIPO (PCT)
Prior art keywords
hiv
antibody
antigen
analyte
assay
Prior art date
Application number
PCT/US1993/003224
Other languages
English (en)
Inventor
James L. Gallarda
Barbara L. Stone
Douglas J. Liu
Susan K. Harrington
George J. Dawson
Ping Wu
Original Assignee
Abbott Laboratories
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 Abbott Laboratories filed Critical Abbott Laboratories
Priority to EP93908748A priority Critical patent/EP0644950A4/fr
Priority to JP51843193A priority patent/JP2001504572A/ja
Priority to CA002133826A priority patent/CA2133826A1/fr
Publication of WO1993021346A1 publication Critical patent/WO1993021346A1/fr

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • G01N33/56988HIV or HTLV

Definitions

  • This invention relates generally to immunoassays, and more particularly, relates to an immunoassay and products for the simultaneous detection of HIV-1 antigen analyte and HIV-1 antibody analyte and/or HIV-2 antibody analyte in a test sample.
  • HIV Acquired immunodeficiency syndrome
  • HIV type 1 HIV-1
  • ARC AIDS-related complex
  • HIV-1 is transmitted by sexual contact, exposure to blood and certain blood products, or from an infected mother to her fetus or child.
  • AIDS and ARC patients and persons at risk is high, and the virus can be isolated from nearly 90% of ail seropositive individuals. See, for example, M. G.
  • HIV-2 a second human immunodeficiency virus
  • HIV-2 appears to be endemic only in west Africa, it appears likely that, based on the experience with HIV-1 , HIV-2 will spread to other parts of the world.
  • HIV-2 virus is similar to HIV-1 virus in its morphology, cell tropism, interaction with the CD4 cellular receptor, in vitro cytopathic effect on CD4 cells, overall genomic structure and its ability to cause AIDS.
  • F. Clavel AIDS 1 :135-140 (19&7).
  • HIV-2 differs from HIV-1 in several respects. See F. Clavel, Ibid and R.A. Weiss et al., AIDS 2:95-100 (1988).
  • HIV-1 serological assays for HIV-1 to react with certain sera from individuals with antibody to HIV-2.
  • a commercially available assay for HIV-1/HIV-2 antibody recently available from Abbott Laboratories (Abbott Park, IL 60064), designated as the Abbott HIVAB ® HIV-1/HIV-2 (rDNA) EIA, uses recombinant antigens corresponding to the two viral proteins, HIV-1 envelope and HIV-2 envelope.
  • the use of these recombinant antigens allows for the improved detection of anti-HIV-1 and/or anti-HIV-2 containing test samples, while minimizing non-specific reactions largely due to cross reactions with whole virus or viral lysate.
  • the use of at least one recombinant HIV protein to detect HIV antibody in a test sample with the use of labelled recombinant HIV antigens is described in the parent patent applications previously incorporated herein by reference.
  • HIV genomic RNA Based on the nucleotide analysis of the viral genome the HIV genomic RNA encodes (beginning at the 5' end):
  • ( i ) a gag gene extending between nucleotides 310 to 1869 and encoding for the internal structure core or nucleocapsid proteins including p24, the most antigenic core protein;
  • the detection of more than one analyte in a test sample usually involves the separate detection of each analyte in a separate assay. Such detection methods have been preferred since they allow for stringent quality assurance
  • Great Britain Patent No. 2188418 teaches an assay tray assembly having reaction wells each with openings in the top surface from which a projection is extended and wherein the inner surface of each well sidewall and the outer surface of each projection may be incubated simultaneously for detecting two or more specific substances present in a specimen which has been introduced into the reaction wells.
  • simultaneous assays The critical factors which have been identified to the development of simultaneous assays are that the two assays to be performed simultaneously must have the same sample volumes, identical incubation times and identical cut-off calculations. Such a simultaneous assay also should be capable of being separately quality controlled for each analyte, both at the manufacturer and at the laboratory using the assay, to ensure the sensitivity, specificity and reproducibility of the immunoassay.
  • an assay wherein the presence of more than one HIV analyte could be simultaneously detected, yet each separate analyte to be detected could be individually quality controlled.
  • Such an assay would be an improvement over other known assays since the simultaneous determinations of the presence of either HIV antigen analyte and/or HIV antibody analyte would be performed in one well, separation of solid phase components would not be required if more than one solid phase was utilized, and the assay could be quality controlled for individual analytes which were to be detected in the simultaneous assay.
  • This invention provides an assay to simultaneously detect the presence, if any, of an HIV antigen analyte and/or an HIV antibody analyte which may be present in a test sample.
  • the assay comprises simultaneously contacting the test sample with (i) at least one HIV antigen analyte capture reagent specific for said HIV antigen analyte attached to a solid phase, and (ii) at least one HIV antibody analyte capture reagent specific for said HIV antibody analyte attached to a solid phase, to form a first mixture; incubating said first mixture for a time and under conditions sufficient to form HIV antigen capture reagent/HIV antigen analyte complexes and/or HIV antibody capture reagent/HIV antibody analyte complexes; contacting said complexes with (i) an HIV antigen analyte indicator reagent which comprises a member of a binding pair specific for said HIV antigen analyte labelled with a signal generating compound, and (i
  • analyte/indicator reagent complexes determining the presence, if any, of said HIV antigen analyte and/or said HIV antibody analyte in the test sample by detecting a signal generated an indication of the presence of either or both analytes in the test sample.
  • the present invention also provides an assay to simultaneously detect the presence, if any, of an HIV antigen analyte and/or an HIV antibody analyte in a test sample, comprising simultaneously contacting the test sample with: (i) at least one HIV antigen capture reagent specific for said HIV antigen analyte attached to a solid phase, (ii) at least one HIV antibody capture reagent specific for said HIV antibody analyte attached to a solid phase, and (iii) an HIV antigen analyte indicator reagent which comprises a member of a binding pair specific for said HIV antigen analyte labelled with a signal generating compound, to form a first mixture; incubating said first mixture for a time and under conditions sufficient to form HIV antigen capture reagent/HIV antigen analyte/HIV antigen indicator reagent complexes and HIV antibody capture reagent/HIV antibody analyte complexes; contacting said complexes with an HIV antibody analyte indicator
  • analyte/HIV antibody analyte indicator reagent complexes and determining the presence, if any, of said HIV antigen analyte and/or said HIV antibody analyte in the test sample by detecting a signal generated.
  • the HIV antibody capture reagent and said antigen capture reagent can be attached (bound) to the same or different solid phase.
  • Preferred solid phases include magnetic beads, non-magnetic beads, wells of a reaction tray,
  • the HIV antigen analyte is HIV p24 gag antigen and HIV antigen analyte capture reagent is anti-HIV p24 antibody, either monoclonal or polyclonal or recombinantly derived, or fragments thereof.
  • the HIV antigen capture reagent is a mixture of monoclonal antibodies secreted by hybridoma cell lines ATCC Deposit Nos. HB 9725 and HB 9726, while the HIV antibody capture reagent is HIV-1 p41 protein , or a mixture of HIV-1 p41 protein and HIV-2 p41 env protein. Most preferably, these antigens are recombinantly produced.
  • the signal generating compound of the HIV antigen analyte indicator reagent and the signal generating compound of the HIV antibody analyte indicator reagent is selected from the group consisting of enzymes, luminescent compounds, chemiluminescent compounds and radioactive elements. Additionally, either or both indicator reagents may further comprise a hapten.
  • the steps comprise (a) contacting the test sample with a solid phase to which at least one anti-HIV p24 antibody and at least one recombinant HIV-1 p41 antigen and at least one HlV-2 p41 antigen have been attached, to form a mixture; (b) incubating said mixture for a time and under conditions sufficient to form anti- HIV p24 antibody/HIV-1 p24 antigen complexes and/or HIV-1 p41 antigen/HIV- 1 antibody and/or HIV-2 p41 antigen/anti-HIV-2 p41 antigen complexes; (c) detecting the presence of HIV p24 antigen in the test sample by: (i) contacting said anti-HIV p24 antibody/HIV p24 antigen complexes with an anti-HIV p24 antibody capable of specifically binding HIV p24 antigen
  • HIV-2 p41 antibody in the test sample can be performed using two (or more) solid phases.
  • Test kits for performing the assays of the present invention also are provided.
  • An assay for the detection of HIV antigen analyte and/or HIV antibody analyte in a test sample is provided.
  • the assay preferably is performed as an immunoassay, although the present invention is not limited to immunoreactive assays. Any assay utilizing specific binding members can be performed.
  • a "specific binding member,” as used herein, is a member of a specific binding pair. That is, two different molecules where one of the molecules through chemical or physical means specifically binds to the second molecule. Therefore, in addition to antigen and antibody specific binding pairs of common
  • immunoassays other specific binding pairs can include biotin and avidin, carbohydrates and lectins, complementary nucleotide sequences, effector and receptor molecules, cofactors and enzymes, enzyme inhibitors and enzymes, and the like.
  • specific binding pairs can include members that are analogs of the original specific binding member, for example, an analyte-analog.
  • Immunoreactive specific binding members include antigens, antigen fragments; antibodies and antibody fragments, both monoclonal and polyclonal; and
  • complexes thereof including those formed by recombinant DNA methods.
  • analyte is the substance to be detected which may be present in the test sample.
  • the analyte can be any substance for which there exists a naturally occurring specific binding member (such as, an antibody), or for which a specific binding member can be prepared.
  • an analyte is a substance that can bind to one or more specific binding members in an assay.
  • “Analyte” also includes any antigenic substances, haptens, antibodies, and combinations thereof.
  • the analyte can be detected by means of naturally occurring specific binding partners (pairs) such as the use of intrinsic factor protein in the capture and/or indicator reagents for the determination of vitamin B 12 , or the use of a lectin in the capture and/or indicator reagents for the determination of a carbohydrate.
  • the analyte can include a protein, a peptide, an amino acid, a hormone, a steroid, a vitamin, a drug including those administered for therapeutic purposes as well as those administered for illicit purposes, a bacterium, a virus, and metabolites of or antibodies to any of the above substances.
  • the test sample can be a mammalian biological fluid such as whole blood or whole blood components including red blood ceils, white blood cells including lymphocyte or lymphocyte subset preparations, platelets, serum and plasma; ascites; saliva; stools; cerebrospinal fluid; urine; sputum; tracheal aspirates and other constituents of the body which may contain or be suspected of containing the analyte(s) of interest.
  • the test sample also can be a culture fluid supernatant, or a suspension of cultured cells. Mammals whose body fluids can be assayed for HIV antigen analyte or HIV antibody analyte according to the present invention include humans and primates, as well as other mammals who are suspected of containing these analytes of interest. It also is contemplated that non-biological fluid samples can be utilized.
  • the indicator reagent comprises a label conjugated to a specific binding member of each analyte.
  • Each indicator reagent produces a detectable signal at a level relative to the amount of the analyte in the test sample.
  • each indicator reagent while comprising a specific binding member of a different analyte, is conjugated to the same signal generating compound (label), which is capable of generating a detectable signal.
  • the indicator reagent is detected or measured after it is captured on the solid phase material.
  • the total signal generated by the indicator reagent(s) indicates the presence of one or more of the analytes in the test sample. It is contemplated that different signal generating compounds can be utilized in the practice of the present invention.
  • different fluorescent compounds could be utilized as the signal generating compounds, one for each indicator reagent, and detection could be determined by reading at different wavelengths.
  • a short-lived chemiluminescent compound such as an acridinium or phenanthridinium compound and a long-lived chemiluminescent compound such as a dioxetane can be utilized to generate signals at different times for different analytes.
  • the specific binding member of the indicator reagent can be a member of any specific binding pair, including either biotin or avidin, a carbohydrate or a lectin, a complementary nucleotide sequence, an effector or a receptor molecule, an enzyme cofactor or an enzyme, an enzyme inhibitor or an enzyme, and the like.
  • An immunoreactive specific binding member can be an antibody, an antigen, or an antibody/antigen complex that is capable of binding either to the analyte as in a sandwich assay, to the capture reagent as in a competitive assay, or to the ancillary specific binding member as in an indirect assay.
  • an antibody can be a monoclonal antibody, a polyclonal antibody, an antibody fragment, a recombinant antibody, a mixture thereof, or a mixture of an antibody and other specific binding members.
  • the details of the preparation of such antibodies and their suitability for use as specific binding members are well known to those in the art.
  • the signal generating compound (label) of the indicator reagent is capable of generating a measurable signal detectable by external means.
  • the various signal generating compounds (labels) contemplated include chromagens; catalysts such as enzymes for example, horseradish peroxidase, alkaline phospatase, and B-galactosidase; luminescent compounds such as fiuorescein and rhodamine; chemiluminescent compounds such as acridinium compounds, phenanthridinium compounds and dioxetane compounds; radioactive elements; and direct visual labels.
  • the selection of a particular label is not critical, but it will be capable of producing a signal either by itself or in conjunction with one or more additional substances.
  • a variety of different indicator reagents can be formed by varying either the label or the specific binding member.
  • the capture reagents of the present invention comprise a specific binding member for each of the analytes of interest which are attached to at least one solid phase and which are unlabeled.
  • the capture reagent is specific for the analyte as in a sandwich assay, it can be specific for indicator reagent or analyte in a competitive assay, or for an ancillary specific binding member, which itself is specific for the analyte, as in an indirect assay.
  • the capture reagent can be directly or indirectly bound to a solid phase material before the performance of the assay or during the performance of the assay, thereby enabling the separation of immobilized complexes from the test sample. This attachment can be achieved, for example, by coating the specific binding member onto the solid phases by absorption or covalent coupling. Coating methods, and other known means of attachment, are known to those in the art.
  • the specific binding member of the capture reagent can be any molecule capable of specifically binding with another molecule.
  • the specific binding member of the capture reagent can be an immunoreactive compound such as an antibody, antigen, or antibody/antigen complex. If an antibody is used, it can be a monoclonal antibody, a polyclonal antibody, an antibody fragment, a recombinant antibody, a mixture thereof, or a mixture of an antibody and other specific binding members.
  • solid phase is not critical and can be selected by one skilled in the art.
  • latex particles, microparticles, magnetic or non-magnetic beads, membranes, plastic tubes, walls of wells of reaction trays, glass or silicon chips and tanned sheep red blood cells are all suitable examples.
  • Suitable methods for immobilizing capture reagents on solid phases include ionic, hydrophobic, covalent interactions and the like.
  • 60- well polystyrene reaction trays and 1/4 inch polystyrene beads are utilized, while in another example, a 96-well reaction tray is the only solid phase utilized. It is contemplated that all solid phases be present during the
  • a “solid phase”, as used herein, refers to any material which is insoluble, or can be made insoluble by a subsequent reaction.
  • the solid phase can be chosen for its intrinsic ability to attract and immobilize the capture reagent.
  • the solid phase can retain an additional receptor which has the ability to attract and Immobilize the capture reagent.
  • the additional receptor can include a charged substance that is oppositely charged with respect to the capture reagent itself or to a charged substance conjugated to the capture reagent.
  • the receptor molecule can be any specific binding member which is immobilized upon (attached to) the solid phase and which has the ability to immobilize the capture reagent through a specific binding reaction. The receptor molecule enables the indirect binding of the capture reagent to a solid phase material before the performance of the assay or during the performance of the assay.
  • the solid phase thus can be a plastic, derivatized plastic, magnetic or non-magnetic metal, glass or silicon surface of a test tube, microtiter well, sheet, bead, microparticle, chip, and other configurations known to those of ordinary skill in the art.
  • the solid phase also can comprise any suitable porous material with sufficient porosity to allow access by detection antibodies and a suitable surface affinity to bind antigens.
  • Microporous structures are generally preferred, but materials with gel structure, in the hydrated state may be used as well.
  • Such useful solid supports include:
  • Natural polymeric carbohydrates and their synthetically modified, cross-linked or substituted derivatives such as agar, agarose, cross-linked alginic acid, substituted and cross-linked guar gums, cellulose esters, especially with nitric acid and carboxylic acids, mixed cellulose esters, and cellulose ethers; natural polymers containing nitrogen, such as proteins and derivatives, including cross- linked or modified gelatins; natural hydrocarbon polymers, such as latex and rubber; synthetic polymers which may be prepared with suitably porous structures, such as vinyl polymers, including polyethylene, polypropylene, polystyrene, polyvinylchloride, polyvinylacetate and its partially hydrolyzed derivatives, polyacrylamides, polymethacrylates, copolymers and terpolymers of the above polycondensates, such as polyesters, polyamides, and other polymers, such as polyurethanes or polyepoxides; porous inorganic materials such as sulfates or carbonates of alka
  • inert carriers such as paper, glass, plastic films, or fabrics.
  • the porous structure of nitrocellulose has excellent absorption and adsorption qualities for a wide variety of reagents including monoclonal antibodies.
  • Nylon also possesses similar characteristics and also is suitable.
  • porous solid supports described hereinabove are preferably in the form of sheets of thickness from about 0.01 to 0.5 mm, preferably about 0.1 mm.
  • the pore size may vary within wide limits, and is preferably from about 0.025 to 15 microns, especially from about 0.15 to 15 microns.
  • the surfaces of such supports may be activated by chemical processes which cause covalent linkage of the antigen or antibody to the support. The irreversible binding of the antigen or antibody is obtained, however, in general, by adsorption on the porous material by poorly understood hydrophobic forces.
  • Preferred solid phase materials for flow-through assay devices include filter paper such as a porous fiberglass material or other fiber matrix materials.
  • the thickness of such material is not critical and will be a matter of choice, largely based upon the properties of the sample or analyte being assayed, such as the fluidity of the test sample.
  • a charged substance can be coated directly to the material or onto microparticles which then are retained by a solid phase support material.
  • microparticles can serve as the solid phase, by being retained in a column or being suspended in the mixture of soluble reagents and test sample, or the particles themselves can be retained and immobilized by a solid phase support material.
  • retained and immobilized is meant that the particles on or in the support material are not capable of substantial movement to positions elsewhere within the support material.
  • the particles can be selected by one skilled in the art from any suitable type of particulate material and include those composed of polystyrene, polymethylacrylate, polypropylene, latex, polytetrafluoroethylene, polyacrylonitriie, polycarbonate, or similar materials.
  • the size of the particles is not critical, although it is preferred that the average diameter of the particles be smaller than the average pore size of the support material being used.
  • An immobilizable immune complex is separated from the rest of the reaction mixture by ionic interactions between the negatively charged poly-anion/immune complex and the previously treated, positively charged porous matrix and detected by using various signal generating systems previously described, including those described in chemiluminescent signal measurements as described in co-pending U.S. Patent Application Serial No. 921 ,979
  • the methods of the present invention can be adapted for use in systems which utilize microparticle technology including automated and semi-automated systems wherein the solid phase comprises a microparticle.
  • Such systems include those described in pending U. S. Patent Application 425,651 and
  • a test sample suspected of containing any of the HIV antigen analyte or HIV antibody analytes of interest is simultaneously contacted with a solid phase to which a first specific binding member of a first analyte is attached, and a solid phase to which a first specific binding member of a second analyte has been attached, thereby forming a mixture.
  • the specific binding members serve as capture reagents to bind the analyte(s) to the solid phases. If the specific binding member is an
  • immunoreactant it can be an antibody, antigen, or complex thereof, specific for each analyte of interest.
  • specific binding member is an antibody, it can be a monoclonal or polyclonal antibody, an antibody fragment, a recombinant antibody, as well as a mixture thereof, or a mixture of an antibody and other specific binding members.
  • This mixture is incubated for a time and under conditions sufficient for a binding reaction to occur and which incubation results in the formation of capture reagent/first analyte complexes of the first analyte if it is present in the test sample, and/or the formation of capture reagent/second analyte complexes of the second analyte if it is present in the test sample.
  • an indicator reagent for each analyte is contacted with the complexes.
  • the indicator reagent for the first analyte comprises a specific binding member of the first analyte of interest which has been labelled with a signal generating compound.
  • the indicator reagent for the second analyte comprises a specific binding member of the second analyte of interest which has been labelled with the same signal generating compound as the indicator reagent for the first analyte, thereby forming a second mixture.
  • This second mixture is incubated for a time and under conditions sufficient to form capture reagent/first analyte/indicator reagent complexes and/or capture reagent/second
  • analyte/indicator reagent complexes The presence of either analyte is determined by detecting the signal generated in connection with the complexes formed on the solid phase as an indication of the presence of one or more analytes in the test sample. If the indicator employs an enzyme as the signal generating compound (label), then the signal can be detected visually or measured spectrophotometrically. Or, the label can be detected by the measurement of fluorescence, chemiluminescence, radioactive energy emission, etc., depending on the label used to generate the signal.
  • the capture reagents can be attached to the same solid phase, or can be attached to different solid phases. It is contemplated that all capture reagents could be attached to the same solid phase, or that each capture reagent could be attached to a separate solid phase, or that combination of capture reagents could be attached to separate solid phases. For example, if microparticles were the solid phase of choice, then separate microparticles could have at least one capture reagent(s) attached to it. A mixture of microparticles (solid phases) could be used to capture the various analytes which may be present in the test sample by using the mixture of microparticles. It is contemplated that different ratios of capture reagents attached to solid phases could be utilized in such an assay, to optimize analyte(s) detection.
  • the specific binding member used as a capture reagent for the HIV-1 antibody analyte be HIV- 1 p41 antigen
  • the specific binding member used as the capture reagent for the HIV-1 antigen analyte be anti-HIV-1 p24 antibody.
  • the HIV-1 p41 used be a recombinantly prepared antigen (protein).
  • the specific binding member for the antibody analyte indicator reagent is HIV-1 p41 antigen, labelled with an enzyme, and that the specific binding member for the antigen analyte indicator reagent is anti-HIV p24 antibody, labelled with an enzyme.
  • HIV-1 p41 antigen be recombinantly produced, and that the enzyme be horseradish peroxidase (HRPO).
  • HRPO horseradish peroxidase
  • the specific binding members serve as capture reagents to bind the analyte(s) to the solid phases.
  • the specific binding member is an immunoreactant, it can be an antibody, antigen, or complex thereof, specific for each analyte of interest.
  • the specific binding member is an antibody, it can be a monoclonal or polyclonal antibody, an antibody fragment, a recombinant antibody, as well as a mixture thereof, or a mixture of an antibody and other specific binding members.
  • the indicator reagents comprise specific binding members of the first and second analytes of interest which have been labelled with a signal generating compound.
  • This mixture is incubated for a time and under conditions sufficient for a binding reaction to occur and which incubation results in the formation of capture reagent/first analyte/indicator reagent complexes of the first analyte and/or capture reagent/second analyte/indicator reagent complexes of the second analyte, if either or both the first or second analyte are present in the test sample.
  • the presence of either analyte is determined by detecting the signal generated in connection with the complexes formed on either or both solid phases as an indication of the presence of the first analyte and/or the second analyte in the test sample.
  • the indicator employs an enzyme as the signal generating compound (label)
  • the signal can be detected visually or measured spectrophotometrically.
  • the label can be detected by the measurement of fluorescence, chemiluminescence, radioactive energy emission, etc., depending on the label used.
  • the assay can include the use of a hapten-anti-hapten system, in which case the indicator reagent can further comprise a hapten such as biotin.
  • a biotin/anti-biotin system for assays is the subject matter of co-pending U.S. Patent Application Serial No. 687,785 which corresponds to published European Patent Application No.
  • the specific binding member used as a capture reagent for the HIV-1 antibody analyte be HIV- 1 p41 antigen, and that the specific binding member used as the capture reagent for the HIV-1 antigen analyte be anti-HIV-1 p24 antibody. It is most preferred that the HIV-1 p41 used be a recombinantly prepared antigen (protein). Also, it is preferred that the specific binding member for the antibody analyte indicator reagent is HIV-1 p41 antigen, labelled with an enzyme. It is most preferred this HIV-1 p41 antigen be recombinantly produced, and that the enzyme be
  • HRPO horseradish peroxidase
  • Solid phases preferred include a magnetic or non-magnetic bead, a well of a reaction tray, and microparticles, either alone or in any combination.
  • Positive and negative controls can be included in the assay of the present invention to ensure reliable results.
  • a blank solid phase(s), to which no capture reagent has been attached, can be utilized as the negative reagent control.
  • Positive controls can include a positive control for each analyte which control is tested separately, and a combined positive control wherein the presence of all analytes to be detected in the assay are determined.
  • recombinantly-prepared antigens be used in the assay.
  • inactivated whole virus which has been cultured in a cell line capable of virus replication as a reagent in an assay for HIV antibody
  • uninfected persons may occasionally give false positive results.
  • the detection of false positive (reactive) results in screening assays of blood products is one of the most pressing problems associated with many known methods of HIV detection. These false problems often are associated with nonspecific binding of immunoglobulins to cellular protein in the viral isolates.
  • These reagents also have the drawback of potentially dangerous manufacturing methods, since culturing of live virus in vitro and isolation and deactivation processes can expose workers to the HIV virss.
  • HIV protein antigen
  • assay methods may solve these problems.
  • Recombinant proteins are non-infectious and therefore the production and isolation of such proteins would be safer than culturing the whole virus.
  • the use of pure viral protein obtained by recombinant methods should eliminate some of the false positive reactions due to nonspecific reactions with contaminating proteins. Further, standardization of reagents should improve the specificity and predictive value of the assay. Expression of HIV gp41 or parts of HIV gp41 have demonstrated the utility of recombinant DNA (rDNA) derived HIV envelope sequences in diagnostic assays.
  • rDNA recombinant DNA
  • the present invention utilizes recombinantly-produced HIV envelope proteins as assay reagents.
  • the cloning of the HIV genome and expression of HIV envelope and core protein in E. coli, the purification and characterization of gp41 and p24, and various assay formats which utilize these recombinant proteins are described in EP-A-386 713, published September 12, 1990 (U.S. Patent Application Serial No. 07/020,282 filed February 27, 1987 and previously incorporated herein by reference, from which this present invention claims priority). Briefly, HIV-infected HT-9 cells were harvested and total cellular DNA was isolated an subjected to digestion.
  • the DNA segments encoding for the core protein and for the envelope glycoprotein were further subcloned into bacterial expression vectors using well-known recombinant technology.
  • the priority application also teaches that, in the detection of HIV-1 antibody, the use of recombinant antigens as the capture reagent and the indicator reagent allows for the detection of anti-HIV-1 antibodies of different immunoglobulin classes.
  • immunoglobulin classes include IgG, IgA, IgE and IgM.
  • the detection of anti-HIV-1 IgG, IgM and IgA using the Abbott HIVAB® HIV-1/HIV-2 (rDNA) EIA assay has been described in an abstract by J. L. Gallarda et al., 5th Annual Forum on AIDS, Hepatitis and Other Blood-Borne Diseases, Atlanta, Georgia, March 29- April 1 , 1992.
  • recombinant antigens produced in heterologous sources can be utilized in the assay and will contribute an even greater lessening of false positive results.
  • an E. coli prepared recombinant antigen such as p41 is used as the capture reagent, then a recombinant antigen p41 produced in any suitable source different than E. coli, such as in a suitable yeast host or other suitable host such as B. megaterium, can be used.
  • the present invention also utilizes antibodies which specifically bind to
  • HIV antigen analytes In a preferred embodiment, anti-HIV p24 antibody is used, in a most preferred embodiment, a mixture of monoclonal antibodies, both specific for HIV p24 antigen, is used. In this mixture, one monoclonal antibody which specifically binds to an epitope on HIV-1 p24 to which epitope human anti-HIV-1 p24 IgG does not competitively bind is used with another monoclonal antibody which specifically binds to a different epitope of HIV-1 p24 to which different epitope human anti-HIV-1 p24 IgG does competitively bind.
  • the monoclonal antibody which does not competitively bind human anti-HIV-1 p24 IgG also specifically binds to HIV-2 p24 antigen.
  • These monoclonal antibodies and their use in HIV antigen assays are the subject matter of co- pending U. S. Patent No. 5,173,399, issued December 22, 1992.
  • These monoclonal antibodies are designated as 31-42-19 and 31-90-25.
  • Hybridoma cell line 31-42-19 producing monoclonal antibody 31-42-19 was deposited at the American Type Culture Collection, 12301 Parklawn Drive, Rockville,. Maryland, 20852 on May 26, 1988 and has been accorded ATCC Deposit No. HB 9726.
  • Hybridoma cell line 31-90-25 producing monoclonal antibody 31-90- 25 was deposited at the American Type Culture Collection, 12301 Parklawn Drive, Rockville, Maryland, 20852 on May 26, 1988 and has been accorded ATCC Deposit No. HB 9725.
  • the use of these monoclonal antibodies as antibody fragments in HIV antigen assays also has been described in U.S. Patent No.
  • HIV-2 p41 would be attached to a solid support as the HIV-2 antigen capture reagent, in addition to the previously-described HIV-1 p41 antigen capture reagent and HIV-1 antibody capture reagents.
  • the solid support can be the same solid support to which all other capture reagents are attached, it can be the same solid support to which HIV-1 recombinant antigens have been attached, or it can be attached to a solid phase to which no other capture reagent (except for HIV- 2 p41) has been attached.
  • the assay procedure would be the same as described hereinabove for the various embodiments of the invention.
  • the HIV-2 antibody analyte indicator reagent would comprise HIV-2 p41 antigen attached to a detectable label.
  • the sequence for the HIV-2 virus (including p41 antigen) is described in EP-A 347,365, published December 20, 1989 to Diagen Corporation, which is incorporated herein by reference.
  • a most preferred HIV-2 recombinant antigen encodes the first 104 amino acids of the HIV-2 p41 antigen.
  • the resulting plasmid designated as pJC104 expresses the HIV-2 env protein as a fusion with CKS protein.
  • This plasmid encodes a recombinant protein containing the first 239 amino acids of the CKS protein, 13 amino acids from the pTB210N multiple restriction site linker, 104 amino acids from the HIV-2 env protein (amino acids 506-609), and an additional 15 amino acids from the pTB210N multiple restriction site linker, following the methods disclosed by Boiling and Mandecki, "CKS Method of Protein Synthesis", EP-A-475 182 published March 18, 1992 (U.S. Patent Application Serial No.
  • This procedure utilized 1/4 inch polystyrene beads (available from Abbott Laboratories, Abbott Park, IL 60064) and a 60-well polystyrene reaction tray (available from Abbott Laboratories, Abbott Park, IL 60064).
  • Two different anti-HIV-1 p24 monoclonal antibodies were coated on the beads, as follows.
  • the beads were coated at a concentration of 8 ⁇ g/ml (approximately 1.6 ⁇ g/ml/bead) in a 0.25 M sodium citrate buffer (pH 7.2) for two hours at 45°C.
  • the beads then were washed in the 0.25 M sodium citrate buffer (pH 7.2), arid then they were reacted with a detergent solution containing 0.1% Triton X- 100 ® (polyoxyethylene ether, available from Sigma Chemical Co., St. Louis, MO) for one hour at 45°C.
  • Triton X- 100 ® polyoxyethylene ether, available from Sigma Chemical Co., St. Louis, MO
  • the beads next were blocked with 1% bovine serum albumin (BSA) in 0.25 M sodium citrate buffer (pH 7.2) for 30 minutes at 45°C, and then overcoated with 2% sucrose, 1% phosphate glass for 15 minutes at 15-30°C in 0.25 M sodium citrate buffer and allowed to dry.
  • BSA bovine serum albumin
  • the two monoclonal antibodies used are designated as 31-42-19 and 31-90-25. They are the subject matter of a patent application U.S. Patent Application Serial No. 07/204,798 that describes their development and uses, previously incorporated herein by reference. Their use also has been described in U.S. Patent No.
  • Hybridoma cell line 31-42-19 producing monoclonal antibody 31-42-19 was deposited at the American Type Culture Collection, 12301 Parklawn Drive, Rockville, Maryland, 20852 on May 26, 1988 and has been accorded ATCC Deposit No. HB 9726.
  • Hybridoma cell line 31-90-25 producing monoclonal antibody 31-90-25 was deposited at the American Type Culture Collection, 12301 Parklawn Drive, Rockville, Maryland, 20852 on May 26, 1988 and has been accorded ATCC Deposit No. HB 9725.
  • the wells of the 60-weil reaction tray were coated with HIV antigen, as follows.
  • the recombinant protein HIV-1 p41 env protein designated as pTB319 was added to each well at a concentration of 1 ⁇ g/ml in 0.1 M 3- [cyclohexylamino]-1-propanesulfonic acid (CAPS buffer, pH 11), and incubated for two hours at 40°C.
  • the wells then were washed twice with 400 ⁇ l of phosphate buffered saline (PBS, pH 7.5), reacted with 0.1% Tween-20 ® for one hour at 40°C, and then blocked with 3% BSA in PBS for one hour at 40°C.
  • the wells next were overcoated with 5% sucrose in PBS for 20 minutes at room temperature and allowed to dry.
  • the pTB319 plasmid producing recombinant protein pTB319 is the subject matter of a patent application to Boiling and Mandecki, "CKS Method of Protein Synthesis," U.S. Patent Application Serial No. 167,067, filed March 11 , 1988, previously incorporated herein by reference.
  • pTB319 was produced by inserting a synthetically-produced DNA fragment which encoded the carboxy terminal 42 amino acids of HIV-1 p120 into the plasmid pTB315, as described in Boiling and Mandecki, Ibid.
  • the two solid phases prepared as described in Example 1 were used in an assay for detection of HIV antigen and/or HIV antibody in a test sample, as follows.
  • An HIV-1 seroconversion panel which contained 65 serum samples derived from nine HIV-1 infected individuals undergoing seroconversion, was used in the assay.
  • Each serum sample was diluted in a separate well of the 60-well tray previously prepared in Example 1 by adding 150 ⁇ l of the serum sample to 50 ⁇ l of specimen diluent, which contained 2% Tween 20®
  • Example 1 polyoxyethylenesorbitan, available from Sigma Chemical Co., St. Louis, MO. Then, a bead previously coated with anti-HIV p24 antibodies as described in Example 1 was placed in each well containing a serum sample. The wells of each tray were incubated for 60 minutes at 40°C under continuous rotation.
  • each well of the 60-well reaction tray was washed with 15 ml of deionized water (dH 2 O) in the Abbott Parallel Processing CenterTM (PPC, available from Abbott Laboratories, Abbott Park, IL). 200 ⁇ l of an HIV p24 antibody probe reagent (rabbit polyclonal F[ab']2 anti-HIV-1 [active
  • HIV-1 p41 antigen labelled with horseradish peroxidase pTB319 coupled to HRPO
  • HRPO-labelled goat anti-rabbit IgG antibody were added to each bead/well of the reaction tray and allowed to incubate for 60 minutes at 40°C without rotation.
  • Each bead/well of the 60-well reaction tray was washed with 15 ml of dH 2 O as previously described.
  • 300 ⁇ l of o-phenylenediamine- 2HCI (OPD) was added to each well/bead and then was incubated for 30 minutes at room temperature in the dark. The reaction then was stopped by adding 300 ⁇ l of a stopping reagent (1 N H 2 SO 4 ) to each well/bead.
  • OPD o-phenylenediamine- 2HCI
  • the reaction was read using the Abbott PPC which measured the optical density of the reaction at 492 nm using a 630 nm reference.
  • the cutoff value was established as 0.1 OD + mean OD of the negative control. Thus, serum samples were considered reactive (positive) if the sample to cutoff value was greater than 1.
  • HIV antigen assay HIV antigen assay
  • HIV antibody assay Human immunodeficiency Virus Types 1 and 2: E. coli and B. megaterium, recombinant antigen, Abbott HIVAB ® HIV-1/HlV-2 (rDNA) EIA; available from Abbott Laboratories, Abbott Park, IL
  • HIV-1/2 Ab HIV-1 Ag Comb designates the assay of the invention
  • HIV-1/2 Ab designates the HIVAB® HIV-1/HIV-2 (rDNA) EIA assay
  • HIV-1 Ag designates the Abbott HIVAG® assay.
  • the data from Table 1 indicates that the method of the present invention had greater sensitivity than either the HIV-1/2 Ab or the HIV-1 Ag test when the results from the three individual tests were compared separately to each other. It is expected that the assay can be optimized even further to detect both HIV p24 antigen which is present early in the course of infection and also in the final stages of HIV infections, as well as HIV antibodies which appear later in infection at the time of seroconversion.
  • blocking reagent comprising 5% non-fat dry milk, 10 mM Tris [pH 8.0] 150 mM NaCl and 0.05% Tween-20 ® ) for one hour at room temperature.
  • the solid phase prepared as described in Example 3 was used in an assay to detect the presence of HIV antigen and/or HIV antibody in a test sample, as follows.
  • Each serum sample of a 12-member seroconversion panel (Panel G available from Boston Biomedica Inc., Boston MA) as well as positive and negative controls were tested.
  • 150 ⁇ l of each serum sample or positive or negative control was diluted in a separate well of the microtiter plate with 50 ⁇ l of specimen diluent (containing 15 ⁇ l of Triton X-100® and 35 ⁇ l of blocking reagent, as described in Example 3) and incubated for 60 minutes at room temperature without rotation.
  • washing buffer 0.05% non-fat dry milk, 10 mM Tris [pH 8.0], 150 mM NaCl, 0.05% Tween 20®
  • HIV p24 antibody probe reagent goat polyclonal F[ab']2 anti-HIV-1 [active ingredient: anti-p24 antibody at a concentration of 2 to 6 ⁇ g/ml] in an antibody diluent (2.25% BSA, 7.5% calf serum, 7.5% goat serum, 25% human recaicified plasma, 0.1% sodium azide) were added to each well and incubated for 60 minutes at room temperature without rotation. After incubation the wells were washed with eight cycles of wash buffer as previously described.
  • conjugate diluent (as described in Example 2) which contained a mixture of recombinant HIV-1 p41 protein (pTB 319) labelled with HRPO and HRPO-labelled goat anti-rabbit IgG (previously described in Example 2) were added to each well and incubated for 60 minutes at room temperature without rotation. The wells were washed with eight cycles of wash buffer (described previously herein) and then rinsed with dH 2 O. Then, 125 ⁇ l of OPD substrate was added to each well and the wells were incubated at room temperature for 10 minutes in the dark. The reaction was stopped by adding 125 ⁇ l of stopping reagent (previously described in Example 2). The absorbance of each well was read at 490 nm with a 630 nm reference. The cutoff value of 0.025 OD + mean
  • the assay of the invention was capable of detecting the presence of HIV antibody and/or HIV antigen in the seroconversion panel.
  • the method of the present invention was more sensitive at detection than either test alone, based on detection of either antigen or antibody.
  • the results from Table 2 of the method of the present invention are compared to the combined results of the HIV-1/2 antibody test and the HIV antigen test, the method of the present invention was able to detect all specimens that were reactive by either test.
  • HIV Antibody and HIV Antigen Capture Reagents Both monoclonal anti-HIV p24 antibodies previously described (31-42- 19 and 31-90-25) and the recombinant HIV-1 p41 antigens previously described (HIV-1 p41 recombinant protein pTB319, and HIV-2 p41
  • recombinant protein pJC104 are together and simultaneously coated onto a uniform 0.5% suspension (wt/volume) of polystyrene microparticles (available from Seradyne Inc., Indianapolis, Indiana) at concentrations of 150 ⁇ g/ml each in 0.01 M carbonate buffer (pH 9.5) for two hours at room temperature (15- 30°C).
  • the suspension of microparticles is briefly centrifuged and the microparticle pellet is resuspended in 0.05M Tris buffer (pH 8.0) to wash away excess uncoupled protein. This washing is repeated until no uncoupled protein remains.
  • microparticles After blocking the microparticles with 10 mg/ml casein in 0.01 M Tris (pH 8.0), 0.15 M NaCl at 56°C for 18-24 hours, the microparticles again are washed as described herein and diluted to 0.015% suspension (wt./volume) in 0.05 M Tris (pH 8.0), 0.15 M NaCl, 1% BSA, 15% sucrose and 0.1% sodium azide.
  • the Abbott IM X ® Microparticle Enzyme Immunoassay (MEIA) system is used, although any system which employs microparticles can be used.
  • the Abbott IM X ® MEIA system is thoroughly described in the Abbott IM X ® Operation and Customer Training Manuals (available from Abbott Diagnostic Division, Abbott Laboratories, Abbott Park, IL).
  • 100 ⁇ l of the 0.15% suspension prepared in Example 5 is mixed together with 100 ⁇ l of test sample suspected of containing HIV-1 and/or
  • HIV-2 antibody and/or HIV-1 antigen incubated at 40°C for ten minutes in an Abbott IM X ® reaction cell to form a reaction mixture.
  • HIV antigens bind to the microparticles in an antibody/antigen/microparticle complex.
  • 150 ⁇ l of the reaction mixture is transferred onto a glass fiber matrix to which the microparticles are retained in an irreversible binding.
  • the antibody/antigen/microparticle complexes then are reacted with 50 ⁇ l of a probe consisting of biotinylated recombinant HIV-1 and HIV-2 recombinant p41 antigens (pTB319 and pJC104, previously described) and biotinylated F(Ab')2 anti-HIV-1 p24 in 0.05M Tris 9 pH 8.0), 2% BSA, 0.25% saponin and 0.1% sodium azide at 40°C for ten minutes.
  • an antibody conjugate consisting of goat anti-biotin alkaline phosphatase in 0.1 M Tris (pH 8.0), 0.5M NaCl, 0.9% Brij-35®, 1.0% BSA and 0.1% sodium azide then is allowed to react with the biotin probe/antibody/antigen/microparticle complexes for ten minutes at 40°C. Then, these microparticle complexes are washed six times with 0.05 M Tris (pH 8.0), 0.3 M NaCl and 0.1 % sodium azide, the biotin
  • probe/a ⁇ tibody/antigen microparticle complex is reacted with 50 ⁇ l of the substrate methylumbelliferyl phosphase (MUP, Abbott Laboratories, Abbott
  • methylumbelliferon (MU) is measured.
  • the rate of MU production is proportional to the concentration of analyte(s) in the test sample.
  • the various analyte capture reagents are coated separately onto polystyrene microparticles (available from Seradyne Inc.,
  • each of the reagents may be coated separately from each other or in various combinations with each other. After each of the analyte capture reagents is coated on their respective microparticles, the various coated microparticles are pooled together and used in the assay.
  • microparticles (31-42-19 and 31-90-25) are coated together onto microparticles separate from the microparticles coated simultaneously with recombinant HIV-1 and HIV-2 p41 antigens (pTB319 and pJC104). Although the exact amount may vary, in general, the coating procedure will approximate that described in Example 5. After blocking the physically separated microparticles with 10 mg/ml casein in 0.01 M Tris (pH 8.0), 0.15 M NaCl at 56°C for 18-24 hours, the
  • microparticles again are washed as described in Example 5, pooled together, and diluted to a 0.015% suspension (wt/volume) in 0.05 M Tris 9 pH 8.0), 0.15 M NaCl, 1% BSA and 15% sucrose.
  • the microparticles may be pooled at various ratios to affect sensitivity and specificity of the assay in order to optimize their use.
  • the Abbott IM X ® Microparticle Enzyme Immunoassay (MEIA) system is used, although any system which employs microparticles can be used.
  • the Abbott IM X ® MEIA system is thoroughly described in the Abbott IM X ® Operation and Customer Training Manuals (available from Abbott Diagnostic Division, Abbott Laboratories, Abbott Park, IL).
  • 100 ⁇ l of the 0.15% suspension prepared in Example 5 is mixed together with 100 ⁇ l of test sample suspected of containing HIV-1 and/or HIV-2 antibody and/or HIV-1 antigen, and incubated at 40°C for ten minutes in an Abbott IM X ® reaction cell to form a reaction mixture.
  • HIV antibodies and/or HIV antigens bind to the microparticles in an
  • antibody/antigen/microparticle complex 150 ⁇ l of the reaction mixture is transferred onto a glass fiber matrix to which the microparticles are retained in an irreversible binding.
  • the antibody/antigen/microparticle complexes then are reacted with 50 ⁇ l of a probe consisting of biotinylated recombinant HIV-1 and HIV-2 recombinant p41 antigens (pTB319 and pJC104, previously described) and biotinylated F(Ab')2 anti-HIV-1 p24 in 0.05M Tris 9 pH 8.0), 2% BSA, 0.25% saponin and 0.1% sodium azide at 40°C for ten minutes.
  • an antibody conjugate consisting of goat anti-biotin alkaline phosphatase in 0.1 M Tris (pH 8.0), 0.5M NaCl, 0.9% Brij-35 ® , 1.0% BSA and 0.1% sodium azide then is allowed to react with the biotin probe/antibody/antigen/microparticle complexes for ten minutes at 40°C. Then, these microparticle complexes are washed six times with 0.05 M Tris (pH 8.0), 0.3 M NaCl and 0.1 % sodium azide, the biotin probe/antibody/antigen microparticle complex is reacted with 50 ⁇ l of the substrate methylumbelliferyl phosphase (MUP, Abbott
  • methylumbelliferon, (MU) is measured.
  • the rate of MU production is proportional to the concentration of analyte(s) in the test sample.
  • the assay of the invention can be optimized even further by varying assay conditions and/or incubation times, using various combinations of antigen or antibody capture or probe reagents, and other methods, reagents and conditions known to those skilled in the art.
  • various other antibody capture reagents can be used, including HIV p24, gp120, gp160, p17, and others.
  • the variance of the antibody capture reagent may then require the use of a different antigen capture reagent. All these variations are contemplated to be within the scope of this invention.
  • manual methods or other automated analyzers can be used or adapted to the assay of the present invention.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Virology (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • AIDS & HIV (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Peptides Or Proteins (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

L'invention concerne un dosage qui permet de détecter simultanément la présence d'analyte antigénique du VIH et/ou l'anticorps du VIH-1 et/ou l'anticorps du VIH-2 dans un échantillon d'analyse. Les analytes sont capturés sur la même phase solide ou sur des phases solides différentes, et la présence d'analytes est déterminée par détection d'un signal émis par des réactifs indicateurs spécifiques de l'analyte.
PCT/US1993/003224 1992-04-09 1993-04-05 Dosage pour la detection de l'antigene du vih et de l'anticorps du vih WO1993021346A1 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP93908748A EP0644950A4 (fr) 1992-04-09 1993-04-05 Dosage pour la detection de l'antigene du vih et de l'anticorps du vih.
JP51843193A JP2001504572A (ja) 1992-04-09 1993-04-05 Hiv抗原及びhiv抗体を検出するためのアッセイ
CA002133826A CA2133826A1 (fr) 1992-04-09 1993-04-05 Test de detection de l'antigene du vih et de l'anticorps anti-vih

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US86638092A 1992-04-09 1992-04-09
US07/866,380 1992-04-09

Publications (1)

Publication Number Publication Date
WO1993021346A1 true WO1993021346A1 (fr) 1993-10-28

Family

ID=25347486

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1993/003224 WO1993021346A1 (fr) 1992-04-09 1993-04-05 Dosage pour la detection de l'antigene du vih et de l'anticorps du vih

Country Status (5)

Country Link
EP (1) EP0644950A4 (fr)
JP (1) JP2001504572A (fr)
AU (1) AU3945893A (fr)
CA (1) CA2133826A1 (fr)
WO (1) WO1993021346A1 (fr)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995023973A2 (fr) * 1994-03-02 1995-09-08 Abbott Laboratories Dosage immunologique vih a l'aide de proteines recombinantes et de reactifs peptidiques de synthese
WO1995033206A1 (fr) * 1994-05-31 1995-12-07 Abbott Laboratories Detection de differents genotypes de vih utilisant un immunodosage modifie par un peptide synthetique
WO1996041179A1 (fr) * 1995-06-07 1996-12-19 Abbott Laboratories Systeme d'expression pour genes non secreteurs
GB2313666A (en) * 1996-05-28 1997-12-03 Hoffmann La Roche Assay for detecting HIV antigens and HIV antibodies
WO1998040744A1 (fr) * 1997-03-10 1998-09-17 Roche Diagnostics Gmbh Procede de detection simultanee d'antigenes de vih et d'anticorps de vih
EP1020727A1 (fr) * 1998-07-30 2000-07-19 Advanced Life Science Institute, Inc. Procede pour la determination de l'hepatite a virus de type c
EP1072888A2 (fr) * 1999-07-28 2001-01-31 Fujirebio Kabushiki Kaisha Méthode et réactif d'immunoessai pour l'antigène HIV-1p24
US6528632B1 (en) 1997-05-02 2003-03-04 Biomerieux Vitek Nucleic acid assays
US6818392B2 (en) 2000-12-06 2004-11-16 Abbott Laboratories Monoclonal antibodies to human immunodeficiency virus and uses thereof
US6962982B2 (en) 2001-06-22 2005-11-08 Roche Diagnostics Corporation Soluble complexes of target proteins and peptidyl prolyl isomerase chaperones and methods of making and using them
US7094757B2 (en) 2001-06-22 2006-08-22 Roche Diagnostics Corporation Complexes comprising a prion protein and a peptidyl prolyl isomerase chaperone, and method for producing and using them
US7094884B2 (en) 2001-06-22 2006-08-22 Roche Diagnostics Corporation Soluble complexes of amylod β peptide and peptidyl prolyl isomerase chaperone and methods of making and using them
EP1798556A1 (fr) * 2005-12-14 2007-06-20 The Jordanian Pharmaceutical Manufacturing Co. Dispositif pour la détection immunochromatographique précoce et rapide de l HIV, et usages correspondants
US7262008B2 (en) 1997-05-02 2007-08-28 Biomerieux, Inc. Nucleic acid assays
US7364860B2 (en) * 2002-05-10 2008-04-29 Bio-Rad Pasteur Method for simultaneously detecting an antigen of, and an antibody against, an infectious microorganism
CN101512339A (zh) * 2006-09-08 2009-08-19 日东纺绩株式会社 检测抗原和针对该抗原的抗体的方法,以及在该方法中所使用的检测试剂
EP2267452A1 (fr) 2001-06-22 2010-12-29 Roche Diagnostics GmbH Complexe soluble comportant une glycoprotéine de surface rétrovirale
US7943375B2 (en) 1998-12-31 2011-05-17 Novartis Vaccines & Diagnostics, Inc Polynucleotides encoding antigenic HIV type C polypeptides, polypeptides and uses thereof
US8133494B2 (en) 2001-07-05 2012-03-13 Novartis Vaccine & Diagnostics Inc Expression cassettes endcoding HIV-1 south african subtype C modified ENV proteins with deletions in V1 and V2
CN106030306A (zh) * 2014-10-08 2016-10-12 艾维可股份有限公司 同时确定血清学特征和评估hiv感染后持续时间的方法
CN109725153A (zh) * 2017-10-26 2019-05-07 北京科美生物技术有限公司 一种均相免疫检测方法及其应用
US11437141B2 (en) 2009-04-30 2022-09-06 Dexcom, Inc. Performance reports associated with continuous sensor data from multiple analysis time periods

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4870003A (en) * 1987-06-15 1989-09-26 Coulter Corporation Simultaneous enzyme immunoassay for detecting antigen and/or antibody in humans
US5108891A (en) * 1988-06-09 1992-04-28 Beth Israel Medical Center Aids assay
CA1335880C (fr) * 1988-07-14 1995-06-13 Thomas P. O'connor Detection d'anticorps et d'antigenes dans le cadre d'un dosage immunologique

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Journal of Acquired Immune Deficiency Syndrome, Vol. 1, No. 3, issued 1988, HASELTINE, "Replication and Pathogenesis of the AIDS Virus", pages 217-240, see entire document. *
Science, Vol. 233, issued 18 July 1986, CLAVEL et al., "Isolation of a New Human Retrovirus from West African Patients with AIDS", pages 343-346, see entire document. *
See also references of EP0644950A4 *

Cited By (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995023973A2 (fr) * 1994-03-02 1995-09-08 Abbott Laboratories Dosage immunologique vih a l'aide de proteines recombinantes et de reactifs peptidiques de synthese
WO1995023973A3 (fr) * 1994-03-02 1995-09-28 Abbott Lab Dosage immunologique vih a l'aide de proteines recombinantes et de reactifs peptidiques de synthese
WO1995033206A1 (fr) * 1994-05-31 1995-12-07 Abbott Laboratories Detection de differents genotypes de vih utilisant un immunodosage modifie par un peptide synthetique
WO1996041179A1 (fr) * 1995-06-07 1996-12-19 Abbott Laboratories Systeme d'expression pour genes non secreteurs
GB2313666A (en) * 1996-05-28 1997-12-03 Hoffmann La Roche Assay for detecting HIV antigens and HIV antibodies
US6593079B1 (en) 1997-03-10 2003-07-15 Roche Diagnostics Gmbh Method for simultaneous detection of HIV antigens and HIV antibodies
WO1998040744A1 (fr) * 1997-03-10 1998-09-17 Roche Diagnostics Gmbh Procede de detection simultanee d'antigenes de vih et d'anticorps de vih
US6528632B1 (en) 1997-05-02 2003-03-04 Biomerieux Vitek Nucleic acid assays
US6558901B1 (en) 1997-05-02 2003-05-06 Biomerieux Vitek Nucleic acid assays
US6586234B1 (en) 1997-05-02 2003-07-01 Biomerieux Vitek, Inc. Device for automated detection of nucleic acids
US7309588B2 (en) 1997-05-02 2007-12-18 Biomerieux, Inc. Nucleic acid assays
US7262008B2 (en) 1997-05-02 2007-08-28 Biomerieux, Inc. Nucleic acid assays
EP1020727A1 (fr) * 1998-07-30 2000-07-19 Advanced Life Science Institute, Inc. Procede pour la determination de l'hepatite a virus de type c
EP1020727A4 (fr) * 1998-07-30 2004-12-29 Advanced Life Science Inst Inc Procede pour la determination de l'hepatite a virus de type c
US7316905B1 (en) 1998-07-30 2008-01-08 Advanced Life Science Institute, Inc. Method for measurement of hepatitis C virus
US7943375B2 (en) 1998-12-31 2011-05-17 Novartis Vaccines & Diagnostics, Inc Polynucleotides encoding antigenic HIV type C polypeptides, polypeptides and uses thereof
EP1072888A2 (fr) * 1999-07-28 2001-01-31 Fujirebio Kabushiki Kaisha Méthode et réactif d'immunoessai pour l'antigène HIV-1p24
EP1072888A3 (fr) * 1999-07-28 2003-06-18 Fujirebio Kabushiki Kaisha Méthode et réactif d'immunoessai pour l'antigène HIV-1p24
EP2336175A1 (fr) 2000-12-06 2011-06-22 Abbott Laboratories Anticorps monoclonaux de virus de l'immunodéficience humaine et leurs utilisations
US7531639B2 (en) 2000-12-06 2009-05-12 Abbott Laboratories, Inc. Monoclonal antibodies that recognize a shared epitope between the human immunodeficiency virus type 1 (HIV-1) capsid (CA/p24) and the human immunodeficiency virus type 2 (HIV-2) capsid (CA/p26)
US6818392B2 (en) 2000-12-06 2004-11-16 Abbott Laboratories Monoclonal antibodies to human immunodeficiency virus and uses thereof
US7531642B2 (en) 2000-12-06 2009-05-12 Abbott Laboratories, Inc. Monoclonal antibodies that recognize a shared epitope between the human immunodeficiency virus type 1 (HIV-1) capsid (CA/p24) and the human immunodeficiency virus type 2 (HIV-2) capsid (CA/p26).
US7531641B2 (en) 2000-12-06 2009-05-12 Abbott Laboratories, Inc. Monoclonal antibodies that recognize a shared epitope between the human immunodeficiency virus type 1 (HIV-1) capsid (CA/p24) and the human immunodeficiency virus type 2 (HIV-2) capsid (CA/p26)
US9115189B2 (en) 2000-12-06 2015-08-25 Abbott Laboratories Monoclonal antibodies to human immunodeficiency virus and uses thereof
US7897332B2 (en) 2000-12-06 2011-03-01 Abbott Laboratories Methods for the detection of HIV-1 or -2 antigens employing monoclonal antibodies that recognize a shared HIV-1/-2 epitope in the capsid p24/p26 proteins
US7528238B2 (en) 2000-12-06 2009-05-05 Abbott Laboratories, Inc. Monoclonal antibodies that recognize a shared epitope between the human immunodeficiency virus type 1 (HIV-1) capsid (CA/p24) and the human immunodeficiency virus type 2 (HIV-2) capsid (CA/p26)
US7531640B2 (en) 2000-12-06 2009-05-12 Abbott Laboratories, Inc. Monoclonal antibodies that recognize a shared epitope between the human immunodeficiency virus type 1 (HIV-1) capsid (CA/p24) and the human immunodeficiency virus type 2 (HIV-2) capsid (CA/p26)
US7531638B2 (en) 2000-12-06 2009-05-12 Abbott Laboratories, Inc. Monoclonal antibodies that recognize a shared epitope between the human immunodeficiency virus type 1 (HIV-1) capsid (CA/p24) and the human immunodeficiency virus type 2 (HIV-2) capsid (CA/p26)
US6962982B2 (en) 2001-06-22 2005-11-08 Roche Diagnostics Corporation Soluble complexes of target proteins and peptidyl prolyl isomerase chaperones and methods of making and using them
US7094757B2 (en) 2001-06-22 2006-08-22 Roche Diagnostics Corporation Complexes comprising a prion protein and a peptidyl prolyl isomerase chaperone, and method for producing and using them
US7094884B2 (en) 2001-06-22 2006-08-22 Roche Diagnostics Corporation Soluble complexes of amylod β peptide and peptidyl prolyl isomerase chaperone and methods of making and using them
US7244575B2 (en) 2001-06-22 2007-07-17 Roche Diagnostics Corporation Soluble complex comprising a retroviral surface glycoprotein
EP2267452A1 (fr) 2001-06-22 2010-12-29 Roche Diagnostics GmbH Complexe soluble comportant une glycoprotéine de surface rétrovirale
US8133494B2 (en) 2001-07-05 2012-03-13 Novartis Vaccine & Diagnostics Inc Expression cassettes endcoding HIV-1 south african subtype C modified ENV proteins with deletions in V1 and V2
US9598469B2 (en) 2001-07-05 2017-03-21 Novartis Vaccines And Diagnostics, Inc. HIV-1 south african subtype C env proteins
US8728723B2 (en) 2002-05-10 2014-05-20 Bio-Rad Innovations Method for simultaneously detecting an antigen of, and an antibody against, an infectious microorganism
US7364860B2 (en) * 2002-05-10 2008-04-29 Bio-Rad Pasteur Method for simultaneously detecting an antigen of, and an antibody against, an infectious microorganism
US8168394B2 (en) 2002-05-10 2012-05-01 Bio-Rad Pasteur Method for simultaneously detecting an antigen of, and an antibody against, an infectious microorganism
US8729234B2 (en) 2002-05-10 2014-05-20 Bio-Rad Innovations Method for simultaneously detecting an antigen of, and an antibody, against, an infectious microorganism
EP1798556A1 (fr) * 2005-12-14 2007-06-20 The Jordanian Pharmaceutical Manufacturing Co. Dispositif pour la détection immunochromatographique précoce et rapide de l HIV, et usages correspondants
WO2007068310A1 (fr) * 2005-12-14 2007-06-21 The Jordanian Pharmaceutical Manufacturing Co. Dispositif destine a la detection immunochromatographique rapide et precoce de vih et son utilisation
CN101512339A (zh) * 2006-09-08 2009-08-19 日东纺绩株式会社 检测抗原和针对该抗原的抗体的方法,以及在该方法中所使用的检测试剂
US11437141B2 (en) 2009-04-30 2022-09-06 Dexcom, Inc. Performance reports associated with continuous sensor data from multiple analysis time periods
CN106030306A (zh) * 2014-10-08 2016-10-12 艾维可股份有限公司 同时确定血清学特征和评估hiv感染后持续时间的方法
EP3204774A4 (fr) * 2014-10-08 2018-07-25 Avioq Inc. Procédés de détermination simultanée de profil sérologique et d'estimation de durée après infection par le vih
US11709163B2 (en) 2014-10-08 2023-07-25 Avioq, Inc. Methods for simultaneous determination of serological profile and estimation of duration post HIV infection
CN109725153A (zh) * 2017-10-26 2019-05-07 北京科美生物技术有限公司 一种均相免疫检测方法及其应用
CN109725153B (zh) * 2017-10-26 2022-05-13 科美诊断技术股份有限公司 一种均相免疫检测方法及其应用

Also Published As

Publication number Publication date
AU3945893A (en) 1993-11-18
JP2001504572A (ja) 2001-04-03
EP0644950A4 (fr) 1997-05-14
EP0644950A1 (fr) 1995-03-29
CA2133826A1 (fr) 1993-10-28

Similar Documents

Publication Publication Date Title
WO1993021346A1 (fr) Dosage pour la detection de l'antigene du vih et de l'anticorps du vih
EP0386136B1 (fr) Immuno-analyse enzymatique pour la detection d'antigenes hiv dans des serums humains
US4870003A (en) Simultaneous enzyme immunoassay for detecting antigen and/or antibody in humans
US5120662A (en) Multilayer solid phase immunoassay support and method of use
US5447837A (en) Multi-immunoassay diagnostic system for antigens or antibodies or both
US4748110A (en) Immunoassay for HTLV-III antigens
Ly et al. Seven human immunodeficiency virus (HIV) antigen-antibody combination assays: evaluation of HIV seroconversion sensitivity and subtype detection
EP0345462B1 (fr) Immunoessai pour des antigènes d'HIV-I utilisant des fragments de F(AB')2 comme sonde
EP0202890A2 (fr) ELISA-compétitif pour détecter les anticorps
JPH04233461A (ja) 同時アッセイ法
EP0698214A1 (fr) Dosage immunologique vih a l'aide de proteines recombinantes et de reactifs peptidiques de synthese
WO1995033206A1 (fr) Detection de differents genotypes de vih utilisant un immunodosage modifie par un peptide synthetique
CN101523216B (zh) 人类免疫缺陷病毒系列蛋白抗体测定方法
EP0386713B1 (fr) Essai immunologique des anticorps contre HIV
GB2313666A (en) Assay for detecting HIV antigens and HIV antibodies
JP3421336B2 (ja) 合成ペプチドを用いてのhtlv−▲i▼とhtlv−▲ii▼との区別
JPH11507635A (ja) Hivを検出するためのペプチド
EP0627625A1 (fr) Essai immunologique pour diagnostic différentiel
AU656221B2 (en) Method for immunological testing and composition and kit for use therein
Bonfanti et al. Detection of human immunodeficiency virus by antigen capture ELISA in culture fluids and blood specimens
Dewar et al. Principles and Procedures of Human Immunodeficiency Virus Serodiagnosis
AU2814889A (en) Enzyme immunoassay for detecting hiv antigens in human sera
Park et al. Evaluation of a simultaneous HIV antigen and antibody detection test in Korean population
JP2002519639A (ja) マルチエピトープ分析と抗原及び抗体組み合わせ測定による結合アッセイの改良

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP KR

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 1993908748

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2133826

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 1993 518431

Country of ref document: JP

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 1993908748

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1993908748

Country of ref document: EP