WO1992012255A1 - Determination simultanee d'analytes multiples a l'aide d'une technique de chimioluminescence heterogene a resolution temporelle - Google Patents

Determination simultanee d'analytes multiples a l'aide d'une technique de chimioluminescence heterogene a resolution temporelle Download PDF

Info

Publication number
WO1992012255A1
WO1992012255A1 PCT/US1991/009714 US9109714W WO9212255A1 WO 1992012255 A1 WO1992012255 A1 WO 1992012255A1 US 9109714 W US9109714 W US 9109714W WO 9212255 A1 WO9212255 A1 WO 9212255A1
Authority
WO
WIPO (PCT)
Prior art keywords
analyte
lived
specific binding
analytes
chemiluminescent
Prior art date
Application number
PCT/US1991/009714
Other languages
English (en)
Inventor
Omar S. Khalil
Kevin R. Genger
Stephen M. Cotter
Yi-Her Jou
Noman A. Abunimeh
Robert G. Hiltibrand
Stephen D. Stroupe
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
Priority to CA002098020A priority Critical patent/CA2098020C/fr
Application filed by Abbott Laboratories filed Critical Abbott Laboratories
Priority to JP4502981A priority patent/JPH06504374A/ja
Publication of WO1992012255A1 publication Critical patent/WO1992012255A1/fr
Priority to KR1019930701951A priority patent/KR930703461A/ko

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
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • G01N33/582Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances with fluorescent label
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6816Hybridisation assays characterised by the detection means
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54313Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals the carrier being characterised by its particulate form
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54313Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals the carrier being characterised by its particulate form
    • G01N33/54326Magnetic particles

Definitions

  • This application relates generally to chemiluminescent labels and more particularly, relates to the simultaneous determination of multiple analytes using a heterogeneous chemiluminescent assay method.
  • acridinium compounds as labels for immunoassays and the subsequent generation of short-lived chemiluminescent signals from these labels has been described by I. Weeks et al., "Acridinium Esters as High Specific Activity Labels in Immunoassays," Clinical Chemistry 29:1474-1478 (1984).
  • Triggering chemiluminescent reactions in a porous matrix and the subsequent detection of signals resulting from immobilized microparticles is described in co- pending United States Patent Application Serial No. 206,645, which enjoys common ownership and is incorporated herein by reference.
  • the use of an ion capture separation method with chemiluminescent detection in a porous matrix is described in co-pending United States Patent Application Serial No. 425,643, which enjoys common ownership and is incorporated herein by reference.
  • the ability to perform simultaneous testing for multiple analytes in the same sample would offer several advantages. First, it would allow obtaining multiple assay results for the same sample processing time, thus it would increase the through-put of automated instruments. Second, it would decrease the cost per test, because the same position of a disposable test device would be used for several test results. Third, it would decrease the amount of volume of used disposable test devices and therefore, the amount of biohazard waste material. Fourth, it would allow simultaneous assays for analytes such as viral antigens or antibodies to viral antigens, thus offering better screening tests. Finally, it would allow simultaneous assays for different analytes such as drugs or for a drug and its metabolites, thus increasing the predictive value of the assay, which in turn would lead to more accurate diagnosis.
  • the present invention overcomes the problems of existing methods by allowing the simultaneous determination of analytes in a chemiluminescent assay by using time resolutions of the light emissions generated under the same reaction conditions.
  • the present invention provides a novel method in which the simultaneous measurement of analytes in a competitive or in a sandwich assay are achieved. This in turn improves the predictive value of the assay and leads to more accurate diagnosis.
  • the present invention provides a method for the determination of multiple analytes which may be present in a test sample comprising (a) incubating the test sample with a mixture of members of specific binding pairs attached to a solid phase for a time and under conditions sufficient for analyte/anti-analyte specific binding pairs to form; (b) incubating with the so-formed specific binding pairs a mixture of labeled members of specific binding pairs for each analyte wherein each analyte is bound to a different chemiluminescent compound (label) capable of generating a different short-lived or long-lived chemiluminescent signal upon contact with a triggering solution; (c) triggering the signal with a triggering solution; (d) measuring the chemiluminescent signal detected; and (e) determining the presence and the amount of each analyte present in the test sample by calculating the difference in time-profile of the signals generated from the chemiluminescent compounds (labels).
  • the solid phase can include a suspension of microparticles comprising a mixture of groups of particles, each group having attached thereto a member of a specific binding pair for one analyte, a tube coated with a mixture of members of specific binding pairs for the analytes, a suspension of magnetizable particles comprising a mixture of groups of particles wherein each group of particles has attached thereto members of a specific binding pair for an analyte, a plastic bead coated with a mixture of members of specific binding pairs for the analytes and a derivatized membrane having attached thereto by chemical binding members of specific binding pairs for the analytes which cover all the membrane or discrete regions of the membrane.
  • the method can further comprise a separation step.
  • the separation step can be performed by magnetic separation.
  • the present invention also provides a method for performing a simultaneous determination of multiple analytes in a test sample which may contain said analytes using a competitive binding chemiluminescence assay comprising (a) incubating the test sample with a known amount of chemiluminescent labeled analytes each capable of generating a short-lived and long-lived chemiluminescent signal and a solid phase which has a limited amount of a mixture of members of specific binding pairs for said analyte attached thereto for a time and under conditions sufficient for analyte/anti-analyte specific binding members to form; (b) adding a substrate specific for one of the labels and incubating to allow a long-lived chemiluminescence-generating reaction to occur; (c) triggering the resultant mixture with alkaline peroxide; and (d) integrating and time-discriminating the short-lived and the long-lived components of the chemiluminescence signal generated.
  • the analytes include haptens, macromolecu.es, metabolites and antibodies.
  • the assay comprises (a) incubating the test sample with a known amount of chemiluminescent labeled specific binding pair members each member capable of generating either a short-lived or a long-lived chemiluminescent signal and a solid phase which has a mixture of analyte derivatives attached thereto, for a time and under conditions sufficient for analyte/anti-analyte specific binding pairs to form; (b) adding a substrate specific for one of the labels and incubating to allow a long-lived chemiluminescence-generating reaction to occur; (c) triggering the resultant mixture with a triggering solution specific for the other label; and (d) integrating and time-discriminating the short-lived and the long
  • the present invention further provides a method for performing a simultaneous chemiluminescence assay for any of multiple analytes which may be present in a test sample, comprising: (a) incubating the test sample with a solid phase coated with a mixture of members of specific binding member pairs for the analytes to form analyte/anti-analyte specific binding pairs; (b) separating the solid phase; (c) adding a mixture of members of specific binding pairs for the analytes having attached thereto different chemiluminescent compounds (labels) capable of generating a different short-lived or long-lived chemiluminescent signal upon contact with a triggering solution and incubating same; (d) adding a substrate to one of the labels and incubating same to allow a long-lived chemiluminescence generating reaction to proceed; (e) triggering the resultant mixture with a triggering solution; and (f) integrating the generated chemiluminescence signal and time-discriminating the short-live
  • the present invention additionally provides a method for the determination of multiple analytes in a test sample which may contain any of the analytes, comprising: (a) incubating the test sample with mixture of members of specific binding pairs of each analyte attached to polymeric ionic molecules; (b) adding a mixture of chemiluminescent labeled members of specific binding pairs for each analyte wherein each analyte is bound to a different chemiluminescent compound (labels) capable of generating a different short-lived or long-lived chemiluminescent signal upon contact with a triggering solution and incubating same to form a reaction mixture of analyte/anti-analyte specific binding pairs; (c) transferring the reaction mixture to a porous membrane treated with a polymeric cationic compound; (d) triggering a chemiluminescent signal with a triggering solution; (e) detecting the chemiluminescent signal generated; and (f) determining the presence and
  • the present invention also provides a kit for performing a simultaneous determination of two analytes comprising containers containing members of specific binding pairs for each analyte wherein each analyte is bound to a different chemiluminescent compound (label) capable of generating a different short-lived or long-lived chemiluminescent signal upon contact with a triggering solution.
  • the short-lived chemiluminescent compound (label) in the kit is an acridinium sulfonamide compound (label); and the long-lived chemiluminescent compound is an alkaline phosphatase substrate or a ⁇ -galactosidase substrate.
  • acridinium compounds labels
  • immunoassays The chemiluminescent properties of acridinium compounds (labels) and their use for immunoassays has been described.
  • Immunochemical tracers with acridinium esters or acridinium sulfonamide levels can be triggered with an alkaline peroxide solution to produce a chemiluminescent signal that maximizes after approximately two (2) seconds. Light emission is completely extinguished after approximately ten (10) seconds.
  • Acridinium sulfonamide labeling chemistry may be employed according to this invention for making a stable tracer of high quantum yield. Such chemistry is described in co-pending U.S. Patent Application Serial No. 371 ,763 entitled "Chemiluminescent Acridinium Salts" filed June 23, 1989, which enjoys common ownership and is incorporated herein by reference.
  • chemiluminescent 1 ,2-dioxetanes can be generated in a variety of ways.
  • a siloxy-substituted dioxetane, 4-(6,tert- butyldimethylsiloxy-2-napthyl)-4-methoxyspiro[1 ,2-dioxetane-3,2'adamantane] is triggerable with tetrabutylammonium flori e solution to produce a chemiluminescent signal lasting for 20 minutes.
  • enzymes such as aryl esterase, alkaline phosphatase or ⁇ -galactosidase react with aryl dioxetane derivatives stabilized with an adamantane cage to produce similar long-lived chemiluminescent signals.
  • long-lived emissions from alkaline phosphatase catalyzed reactions of 3(2'-spiroadamantane)-4-methoxy-4-(3"- phosphoryloxy)-phenyl-1,2-dioxetane (AMPPD) and of a similar ⁇ -galactosidase substrate have been described in WO 881 00694, along with the use of these compounds (labels) in an immunoassay.
  • Alkaline phosphatase and ⁇ -galactosidase labeling techniques are known, and catalyzed dioxetane chemiluminescence can be used according to this invention to generate long-lived signals.
  • a short-lived chemiluminescent label can be any member of the acridinium and/or phenanthridinium compounds (labels) or any chemiluminescent compound (label), as long as it is capable of generating a short ⁇ lived signal.
  • Luminol derivatives also can be used with the chemiluminescent generating reaction performed, at a triggering condition of pH and catalysts, in such a way that the reaction is triggered and decays in a few seconds.
  • a long-lived chemiluminescent compound (label) can be one of the class of dioxetane compounds (labels) that can react with enzyme labels such as alkaline phosphatase or ⁇ -galactosidase to generate chemiluminescent signals of long durations. It is also contemplated that any compound which can be configured to generate a long-lived signal, such as luminol, or any compound which generates a long-lived signal can be used. Alkaline phosphatase catalyses the decomposition of AMPPD at pH 8-9 to generate a chemiluminescence signal of long duration. Increasing the pH to ca.
  • a trigger solution according to this invention is an alkaline peroxide solution.
  • This trigger solution can be prepared by the addition of hydrogen peroxide to a sodium hydroxide solution or by dissolving solid urea-hydrogen peroxide adduct in sodium hydroxide.
  • the alkaline peroxide trigger solution acts on the acridinium label to generate a short-lived chemiluminescence signal due to the decomposition of a high energy intermediate and the generation of an excited acridone derivative.
  • Alkaline peroxide also according to this invention, enhances the enolization of the enzyme reaction product and hence, the intensity of the chemiluminescence signal. This appears as a pH-jump-like change in the signal.
  • Alkaline peroxide also according to this invention, reacts with the non-luminescent reaction product of ⁇ -galactosidase with AMPGD to generate a chemiluminescence signal of long duration.
  • a solid phase according to the present invention is a mixture of polymeric microparticles with chemically or physically bound antigens or antibodies.
  • Microparticles that can be used include polystyrene, carboxylated polystyrene, polymethylacrylate or similar particles with radius in the range of from about 0.1 to 20 ⁇ m.
  • a preferred separation method for these particles is the use of microparticle capture on a glass fiber matrix followed by triggering a chemiluminescent reaction on this matrix.
  • Another preferred method of separation is that which is described in co- pending U.S. Patent Application Serial No. 150,278, and U. S. Patent Application Serial No. 375,029, both of which enjoy common ownership and both of which are incorporated herein reference.
  • These applications describe the use of ion capture separation, in which the antibodies or antigens for the assay in question are chemically attached to polyanionic acids such as polyglutamic acid or potyacrylic acid.
  • the fibrous pad is treated with a cationic polymer to render the fibers positively charged. Separation of the immunochemical reaction products are affected by the electrostatic interaction between the positively charged pad and the negatively charged poly-anion/immune complex.
  • Magnetizable microparticles that can be used preferably have ferric oxide or chromium oxide cores and polystyrene, carboxylated polystyrene, polymethylacrylate coating.
  • a preferred separation method for these particles is the use of constant or pulsed magnets, washing said particles, and then suspending the separated particles in a vessel where a chemiluminescent reaction can be generated and detected.
  • solid supports are known to those in the art and include the walls of wells of a reaction tray, test tubes, polystyrene beads, nitrocellulose strips, membranes, and others.
  • Microparticles with chemically or physically bound antigens or antibodies can be used according to the invention as capture phases in a binding reaction to make use of the fast diffusion rates of these particles in solution to yield rapid results.
  • Microparticles that can be used according to this invention include polystyrene, carboxylated polystyrene, polymethylacrylate or similar particles with radius in the range of from about 0.1 to 20 ⁇ m.
  • Test samples which can be tested by the methods of the present invention described herein include biological fluids such as human and animal body fluids.
  • biological fluids such as human and animal body fluids.
  • whole blood, serum, plasma, cerebrospinal fluid, urine, as well as cell culture supernatants, and the like may be used.
  • 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 including infectious disease antigens such as viral, bacterial, rickettsial antigens and also cancer markers such as CEA, also macromolecules, haptens and/or their metabolites, 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 as a member of a specific binding pair for the determination of Vitamin B12, or the use of lectin as a member of a specific binding pair for the determination of a carbohydrate.
  • the analyte can include a protein, a peptide, an amino acid, DNA or RNA probe sequences, 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 details for the preparation of such antibodies and the suitability for use as specific binding members are well-known to those skilled in the art.
  • a “capture reagent”, as used herein, refers to an unlabeled specific binding member which is specific either for the analyte as in a sandwich assay, for the indicator reagent or analyte as 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 .
  • the “indicator reagent” comprises a signal generating compound (label) which is capable of generating a measurable signal detectable by external means conjugated (attached) to a specific binding member for the analyte(s).
  • Specific binding member as used herein means 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.
  • the indicator reagent also can be a member of any specific binding pair, including either hapten-anti-hapten systems such as biotin or anti-biotin, avidin or biotin, a carbohydrate or a lectin, a complementary nucleotide sequence, an effector or a receptor molecule, an enzyme cofactor and an enzyme, an enzyme inhibitor or an enzyme, and the like.
  • hapten-anti-hapten systems such as biotin or anti-biotin, avidin or biotin, a carbohydrate or a lectin, a complementary nucleotide sequence, an effector or a receptor molecule, an enzyme cofactor and 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 an 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.
  • the various signal generating compounds (labels) contemplated in the practice of the invention include short-lived chemiluminescence signal generating compounds (labels) such as an acridinium sulfonamide, an acridinium ester or a phenanthrkJine compound (label), and long-lived chemiluminescence signal generating compounds (labels) whose signals result from action of enzymes or nucleophilic agents on dioxetane compounds containing an adamantane structure.
  • short-lived chemiluminescence signal generating compounds labels
  • labels such as an acridinium sulfonamide, an acridinium ester or a phenanthrkJine compound (label)
  • long-lived chemiluminescence signal generating compounds (labels) whose signals result from action of enzymes or nucleophilic agents on dioxetane compounds containing an adamantane structure.
  • the reagent(s) employed for the assay can be provided in the form of a kit with one or more containers such as vials or bottles, with each container containing a separate reagent such as a monoclonal antibody, or a cocktail of monoclonal antibodies, employed in the assay as capture phases or as indicator reagents which comprise chemiluminescent compounds (labels) as signal generating compounds.
  • a test sample which may contain any or all of the analytes of interest, a mixture of probes for the analytes labeled as described herein and a mixture of capture phases for the analytes are incubated for a period of time and under conditions sufficient to allow optimal immunochemical binding reactions for the analytes to take place.
  • the capture phase then is separated and washed.
  • a substrate specific to the alkaline phosphatase label then is added. This substrate is rendered chemiluminescent by the action of the enzyme label. After the enzyme/substrate reaction reaches an end point, the separated reaction mixture is triggered using an alkaline peroxide solution.
  • the signal is collected over a period of about four to ten seconds and is integrated to give the sum of the pH-jump enhanced alkaline phosphatase catalyzed chemiluminescence and the alkaline peroxide triggered acridinium sulfonamide chemiluminescence.
  • the residual signal is collected and integrated for an equal time interval to give the steady state enzyme catalyzed chemiluminescence. If two analytes are tested, the presence or absence of either of the two analytes is determined from the relative magnitude of the signals collected at the two time intervals.
  • a test sample which may contain any or all of the analytes of interest, a mixture of probes for the analytes labeled as described herein and a mixture of capture phases for the analytes are incubated for a period of time and under conditions sufficient to allow optimal immunochemical binding reactions for the analytes to take place.
  • the capture phase then is separated and washed.
  • a substrate specific to the alkaline phosphatase label then is added. This substrate is rendered chemiluminescent by the action of the enzyme label. After the enzyme/substrate reaction reaches an end point, the generated chemiluminescence signal corresponding to the enzyme label is integrated.
  • the separated reaction mixture is then triggered using an alkaline peroxide solution.
  • the signal is collected over a period of a few hundred milliseconds and corresponds to the alkaline peroxide triggered acridinium sulfonamide chemiluminescence. If two analytes are tested, the presence or absence of either of the two analytes is determined from the relative magnitude of the signals collected at the two time intervals.
  • a test sample which may contain any or all of the analytes of interest, a mixture of probes for the analytes labeled as described herein and a mixture of capture phases for the analytes are incubated for a period of time and under conditions sufficient to allow optimal immunochemical binding reactions for the analytes to take place.
  • the capture phase then is separated and washed.
  • a substrate specific to the ⁇ -galactosidase label then is added. This substrate is hydrolized by the action of the enzyme label. After the enzyme/substrate reaction reaches an end point, the separated reaction mixture is triggered using an alkaline peroxide solution.
  • the signal is collected over a period of about one second and is integrated to give the alkaline peroxide triggered acridinium sulfonamide chemiluminescence.
  • the residual signal is collected after a delay of four to five seconds and integrated for an equal time interval to give the steady state enzyme catalyzed product that is rendered chemiluminescent by the action of alkaline peroxide. If two analytes are tested, the presence or absence of either of the two analytes is determined from the relative magnitude of the signals collected at the two time intervals.
  • a microparticle-based one-step sandwich immunoassay for multiple analytes is performed according to this invention, as follows: A test sample which may contain any or all of the analytes of interest is contacted with a mixture of microparticles comprising two groups of particles, one group coated with a polyclonal antibody to the first analyte of interest, and the second group of particles coated with a polyclonal antibody to the second analyte of interest. A conjugate mixture of labeled antibodies then is added to the reaction vessel.
  • One antibody in this mixture is specific for the first analyte and is labeled with (bound to) a short ⁇ lived chemiluminescence signal generating compound (label) such as an acridinium sulfonamide, an acridinium ester or a phenanthridine compound.
  • label such as an acridinium sulfonamide, an acridinium ester or a phenanthridine compound.
  • label a short ⁇ lived chemiluminescence signal generating compound
  • the second antibody of the conjugate mixture is labeled with alkaline phosphatase.
  • the mixture is incubated for a time and under conditions sufficient for analyte-specific complexes, comprising analyte and anti-analyte antibodies, to form. Then, the microparticles are separated and washed.
  • a preferred method of separation is one which employs a glass fiber pad, using the fluid removal method described in co-pending U. S.
  • An alkaline phosphatase substrate such as that described in published EP 0 254 051 or WO 881 00694, capable of generating a long-lived chemiluminescence signal when reacted with the substrate, then is added to the separated particles and is allowed to react for a time and under conditions sufficient to approach steady state. This generally takes only a few minutes. The microparticles and substrate reaction product then is triggered with an alkaline peroxide solution in an apparatus such as that described in co-pending U. S. Patent Application 07/206,645.
  • Another method of the invention comprises contacting the test sample which may contain either or both of the analytes of interest with a mixture comprising two groups of microparticles.
  • a conjugate mixture of labeled antibodies is added into the reaction vessel.
  • One antibody of the conjugate mixture is specific to the first analyte and is labeled with a short-lived chemiluminescence signal generating label such as an acridinium sulfonamide, an acridinium ester, or a phenanthridine compound (label).
  • the second antibody of the conjugate mixture is specific for the second analyte and Is labeled with alkaline phosphatase.
  • the mixture is incubated for a time and under conditions sufficient to form analyte/anti- analyte/anti-anti-analyte complexes. Then, the microparticles are separated and washed, and the chemiluminescence signals triggered and measured as described hereinabove.
  • the microparticles are coated with polyclonal antibodies and the conjugate is a mixture of labeled polyclonal antibodies.
  • a microparticle-based two-step sandwich immunoassay for two analytes can be performed according to the invention, as follows.
  • the test sample which may contain either or both analytes of interest and a mixture of microparticles comprising two groups of particles, one group of particles coated with a polyclonal antibody specific to the first analyte, and the second group of particles coated with a polyclonal antibody to the second analyte, are contacted.
  • the test sample and microparticles are incubated for a time and under conditions sufficient to allow binding of the analytes to the particles.
  • the microparticles are separated and washed.
  • a preferred method of separation is on a glass fiber pad, using the fluid removal method described in co-pending application Serial No.
  • a conjugate mixture of labeled antibodies is added to the separated particles on the pad.
  • One antibody in the conjugate mixture is specific to the first analyte and is labeled with a short-lived chemiluminescent generating label such as an acridinium sulfonamide, an acridinium ester or a phenanthridine compound (label).
  • the second antibody of the conjugate mixture is specific for the second analyte and is labeled with alkaline phosphatase. This conjugate mixture is allowed to incubate with the separated microparticles for a time and under conditions sufficient for complexes to form. The excess conjugate mixture then is removed by washing.
  • An alkaline phosphatase substrate which is capable of producing long-lived chemiluminescence signal when reacted with the enzyme then is added to the separated particles. This mixture is allowed to react for a time and under conditions sufficient to approach steady state and thus to form a substrate reaction product. The amount of time to reach steady state is usually only a few minutes.
  • the microparticle and substrate reaction product then is triggered with an alkaline peroxide solution in a reaction vessel such as that described in co- pending U. S. Patent Application Serial No. 206,645 previously incorporated herein by reference.
  • the test sample which may contain any of the analytes of interest and a mixture comprising two groups of microparticles, one group of particles coated with a monoclonal antibody to an epitope of the first analyte and the second group of particles coated with a monoclonal antibody to an epitope of the second analyte, is contacted to form a mixture.
  • the mixture is incubated for a time and under conditions sufficient for analyte/anti- analyte antibody complexes to form. Then, the microparticles are separated and washed. A conjugate mixture of labeled antibodies then is added to the so-formed complexes.
  • One antibody of the conjugate mixture is specific to the first analyte and is labeled with a short-lived chemiluminescence-generating label such as an acridinium sulfonamide, an acridinium ester or a phenanthridine compound (label).
  • the second antibody of the conjugate mixture is specific for the second analyte and is labeled with alkaline phosphatase. After incubation for a time and under conditions sufficient for the conjugate to bind to either or both analyte/anti-analyte complexes, the excess conjugate mixture is washed off and the chemiluminescence signals are generated and measured.
  • the microparticles can be coated with polyclonal antibodies and the conjugate can be a mixture of labeled polyclonal antibodies.
  • Ion capture procedures for immobilizing an immobilizable reaction complex with a negatively charged polymer described in co-pending U. S. Patent Application Serial No. 150,278 filed January 29, 1988, which enjoys common ownership and is incorporated herein by reference, can be employed according to the present invention to effect a fast solution-phase immunochemical reaction.
  • 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 chemiluminescent signal measurements as described in co-pending U.S. Patent Application Serial No. 425,643, previously incorporated herein by reference.
  • An ion capture-based competitive chemiluminescent immunoassay for two haptens can be performed according to this invention, as follows.
  • the test sample which may contain any of the analytes of interest is contacted with a mixture of labeled antibodies.
  • One antibody in the mixture is specific to the first hapten, and is labeled with an acridinium sulfonamide, an acridinium ester or a phenanthridine compound (label).
  • the second antibody of the mixture is specific for the second hapten and is labeled with alkaline phosphatase.
  • the test sample and mixture of labeled antibodies are incubated for a time and under conditions sufficient for hapten/anti-hapten complexes to form a reaction mixture.
  • a mixture of capture phase is added to the reaction mixture.
  • the capture phase mixture comprises a mixture of two components each of which is a hapten bound to polyglutamic acid residue. The binding can be accomplished either directly or through a carrier
  • Another alternative way of performing these assays according to the present invention is to use a combination of microparticle capture and ion capture separation procedures.
  • a sample suspected of containing multiple analytes can be incubated with a mixture of microparticles having bound thereto a member of specific binding pair for one analyte, and polyionic residues having bound thereto members of specific binding pairs for the second analyte.
  • the reaction mixture is then transferred to a porous matrix that has been treated to carry an opposite charge to the polyionic residue.
  • microparticles captured analyte is retained on the porous matrix by hydrophobic interactions and the poly ⁇ ionic captured analyte is retained on the oppositely charged porous matrix by ionic interaction.
  • the retained reaction products are washed and a mixture of conjugates is added on the porous matrix.
  • One of these conjugates is labeled with a label that can be triggered to yield a short-lived chemiluminescence signal, the other is labeled with a label that yields a long lived chemiluminesce signal.
  • the signals are triggered and segregated by time resolution.
  • the phencyclidine (PCP) capture reagent was prepared by coupling the free acid form of polyglutamic acid with phenylcyclidine-4-chloroformate.
  • the free acid form of polyglutamic acid was prepared from the sodium salt of polyglutamic acid according to the following procedure: 1 gm of polyglutamic acid sodium salt (Sigma Chemical Company; St. Louis, MO) was stirred overnight with 7 gms of AG5OW-X8 cation exchange resin (Bio-Rad Laboratories, Richmond, CA) suspended in 20 mL water. The ion exchange resin was previously swelled and washed in distilled water. The supernatant was separated from the resin and lyophilized to give about 0.8 g of the free acid form of polyglutamic acid.
  • PCP-4-chloroformate was prepared by reacting 1.1 mg 4-
  • the reaction was carried out overnight at room temperature and then the reaction mixture was evaporated to dryness.
  • the dried product was dissolved in 1.5 mL 0.1 M sodium phosphate buffer, pH 7.0, and was dialyzed against a volume of the same buffer in a 3,500 molecular weight cut-off dialysis bag overnight at room temperature.
  • the precipitate was filtered.
  • the cloudy aqueous filtrate was extracted with methylene chloride (Fisher Scientific, Itasca, IL) until it was clear.
  • a fluorescence polarization immunoassay confirmed the presence of PCP on the PGA residue (performed on Abbott TD X ®, available from Abbott Laboratories, Abbott Park, IL 60064).
  • the aqueous layer was diluted in a solution containing 1% fish gelatin, 100 mM sodium chloride, 25 mM Tris, 1 mM magnesium chloride, 0.1 mM zinc chloride, 0.1% sodium azide, pH 7.2, to yield 1.875 ⁇ gm/mL of PCP-PGA capture reagent.
  • the mo ⁇ hine capture reagent was prepared by coupling the isothiocyanate derivative of the free acid form of polyglutamic acid with morphine-ovalbumin.
  • the free acid form of polyglutamic acid was activated by derivatizing it to an isothiocyanate (ITC-PGAFA).
  • ITC-PGAFA isothiocyanate
  • the isothiocyanate derivative was reacted with ovalbumin to form ovalbumin-PGA.
  • morphine was coupled to ovalbumin- PGA using isobutylchloroformate to yield the capture reagent: morphine- ovalbumin-PGA.
  • the procedure used was as follows. Ten (10) mg of the free acid form of polyglutamic acid was dissolved in 1 ml dimethylformamide (DMF). 0.01 ml of proton adsorbing agent 4-methylmorpholine and 4.8 mg of 1 ,4-phenylene diisothfocyanate (DITC)(available from the Aldrich Chemical Co., Milwaukee, Wl) in 0.2 ml of dimethylformamide were added to this solution (100 mole excess). This reaction mixture was stirred at room temperature overnight and then it was concentrated using a rotary evaporator. Twenty-five (25) ml of methylene chloride was added dropwise to precipitate the ITC-PGAFA.
  • DITC 1 ,4-phenylene diisothfocyanate
  • the flocculent precipitate was centrifuged, and methylene chloride and unreacted DITC were decanted. The precipitate then was suspended in 1 ml of dimethylformamide. The precipitation/centrifugation suspension process was repeated until no DITC was detectable in the supernatant using thin-layer chromatography (TLC) on silica slides. The remaining solid was vacuum dried to yield the ITC-PGAFA as a yellow powder.
  • TLC thin-layer chromatography
  • Ovalbumin-PGA was prepared according to the following procedure. Ten (10) mg ovalbumin (available from Sigma Chemical Co., St. Louis, MO) were dissolved in 0.5 ml of 0.2 M sodium phosphate buffer (pH 8.5) and filtered through a 0.45 ⁇ m syringe filter. This solution was reacted with 133.3 mg of ITC-PGA dissolved by sonication in 3 ml of 0.2 M sodium phosphate buffer at pH 9.0. The pH then was adjusted to 8.5 with 1 N sodium hydroxide.
  • the mixture was incubated overnight at 37°C, and then it was fractionated on an HPLC instrument using a TSK- 3000SWG preparative gel filtration column (available from Beckman Instruments, Arlington Heights, IL) run at 5 ml per minute with 0.1 M sodium phosphate, 0.3 M sodium chloride (pH 6.8). Fractions were monitored using an absorbance detector at 280 nm. Two (2) ml fractions were cut and six (6) fractions (12 ml) were grouped starting with the void volume.
  • One (1) mg morphine 3- ⁇ -D-glucuronide (Sigma Chemical Co., St. Louis, MO) MW 461.5, was activated by reacting it with 100 mole excess isobutylchloroformate (MW 136.15, available from Sigma Chemical Co., St. Louis, MO) and 100 mole excess 4-methylmorpholine in DMF at 0°C for one-half hour.
  • the activated morphine derivative was added to the dialyzed ovalbumin-PGA and kept in an ice bath for one (1) hour at 0-4°C. The reaction mixture then was kept at room temperature overnight.
  • the product solution was dialyzed against 0.1 M sodium phosphate buffer at pH 8.5, in 3,500 molecular weight cut-off dialysis bag to remove the uncoupled morphine.
  • the recovered dialysate was run in the fluorescence polarization immunoassay for morphine, using a commercial analyzer (TD ⁇ ® ( Abbott Laboratories, North Chicago, IL), which gave 3.5 morphine molecules per ovalbumin-PGA residue.
  • the capture reagent (morphine-ovalbumin- PGA) was diluted in a buffer solution containing 1% fish gelatin, 25 mM Tris, 100 mM sodium chloride, 1 mM magnesium chloride, 0.1 mM zinc chloride and 0.1% sodium azide at a pH of 7.2. The final concentration was 249.9 ng ovalbumin/ml.
  • the simultaneous assay capture reagent was prepared by mixing the two individual assay capture reagents. Solutions were diluted together to give 1.875 ⁇ g/ml of phenylcyclidine-polyglutamic acid and 0.25 ⁇ g/ml of morphine- ovalbumin-polyglutamic acid in a final solution which contained 1% fish gelatin, 25 mM Tris, 100 mM sodium chloride, 1 mM magnesium chloride, 0.1 mM zinc chloride and 0.1% sodium azide at a pH of 7.2.
  • Monoclonal anti-phenylcyclidine antibody was labeled with acridinium sulfonamide using EDAC coupling procedures known in the art. It then was diluted in a buffer which contained 1% fish gelatin, 25 mM Tris, 100 mM sodium chloride, 1 mM magnesium chloride, 0.1 mM zinc chloride, 10% normal mouse serum and 0.1 sodium azide at pH 7.2. The final antibody concentration was 118 ng/ml.
  • Monoclonal anti-morphine antibody was labeled with alkaline phosphatase using procedures known in the art. It was diluted in the same buffer used for the anti-phenylcyclidine-acridinium probe. Its final concentration was 48.2 ng/ml.
  • the simultaneous assay probe consisted of a monoclonal anti-phenylcyclidine antibody conjugated to acridinium (118 ng/ml) and a monoclonal anti-morphine antibody conjugated to alkaline phosphatase (48.2 ng/ml) diluted in the same buffer.
  • the alkaline phosphatase substrate solution was 0.4 mM of 3-(2'- spiroadamantane)-4-methoxy-4-(3"-phosphoryloxy) phenyl-l ,2,dioxetane,
  • SUBST AMPPD (available from Tropix Inc., Bedford, MA) disodium salt, in a solution of 0.05 M sodium bicarbonate buffer containing 1 mM magnesium chloride at pH 9.5.
  • the disposable reaction tray used for this assay was that described in co- pending U. S. Patent Application Serial No. 425,651 , which enjoys common ownership and which is incorporated herein by reference.
  • the device comprised a funnel-like structure, a porous element, and an absorbant material, which were assembled to provide intimate contact between the porous element and the absorbent material.
  • the porous element was made of fibrous glass material, Product No. 4111 glass fiber filter paper, which had a nominal thickness of 0.05 inches and which is available from Hollingsworth and Voss Co., East Walpole, MA.
  • Test samples used were phenylcyclidi ⁇ e calibrators from a commercially available fluorescence polarization kit ( TD X ® kit, Abbott Laboratories, North Chicago, IL) which contained 500, 250, 120, 60, 25 and 0 ng/ml PCP in human urine.
  • 80 ⁇ l of Tris buffer (Abbott Laboratories, North Chicago, IL) were dispensed on the fibrous glass matrix pad of the detection well of a disposable reaction tray. This was followed by 80 ⁇ l of a 0.5% CelquatTML-200 solution on each pad. Solutions were dispensed using two FMI-RH pumps (Fluid Metering Inc., Oyster Bay,
  • test sample was pipetted into the shallow reaction well of the disposable reaction tray, using an automated pipettor.
  • 50 ⁇ l of acridinium-labeled anti-PCP antibodies were dispensed into each incubation well.
  • the mixture was incubated for 9.6 minutes while the disposable reaction tray was moving on the track in a temperature controlled tunnel at 32°C.
  • the timing belt steps at the rate of 0.8 inches per minute, the reaction tray was stationary for 36 seconds after each step for a reaction step to take place.
  • 50 ⁇ l of a solution containing PCP- PGA capture reagent prepared as in Example 1 at a concentration of 118 ⁇ g PGA ml was dispensed into the incubation well through a tip centered on the well and connected to an FMI pump through a Teflon® line.
  • reaction mixture was further incubated while the disposable tray continued movement along the track. After 4.8 minutes, each quaternary ammonium polymer-treated glass fiber matrix was rinsed with 100 ⁇ l of IM X ® Tris buffer (available from Abbott Laboratories, North Chicago, IL) dispensed from a tip centered on the fibrous pad of the detection well. After 4.8 more minutes, the disposable reaction tray was located under the transfer device described in the co-pending Patent Application Serial No. 425,643, previously incorporated herein by reference. The 150 ⁇ l assay mixture then was transferred from the shallow incubation well onto the pre-treated glass fiber matrix in the detection well.
  • IM X ® Tris buffer available from Abbott Laboratories, North Chicago, IL
  • Transfer was effected using one 350 ⁇ l pulse of IM X ®Tris buffer injected from three adjacent nozzles at a linear rate of 210 cm/sec.
  • the transfer fluid was injected by a stepper-motor controlled syringe pump.
  • a valve directed the transfer solution to each side of the disposable reaction tray.
  • the transferred mixture then was allowed to drain through the fibrous pad for 12 seconds.
  • 50 ⁇ l of the AMPPD substrate solution was dispensed in each detection well to saturate the fibrous matrix.
  • the disposable tray was moved on the timing belt to allow subsequent well pairs to be located under the transfer device to effect transfer of the reaction mixture.
  • the disposable device then was moved to a detection position where a chemiluminescence detector (described in co-pending U. S. Patent Application Serial No. 425,643) was located.
  • the detection head had two photomultiplier tubes (PMT) and light pipes, each centered on a detection well.
  • PMT photomultiplier tubes
  • Each PMT was powered by a Bertan high voltage power supply and connected to a photon counting amplifier board and a counter/timer board constructed using components and methods known in the art.
  • the two trigger solution injectors associated with each PMT were connected to an FMI pump using a black Teflon® tube and a manifold. As the tray reached the detection position, the detector head was lowered to create a light-tight seal with the surface feature on the disposable.
  • the high voltage to the PMT was gated on, the two trigger solution pumps and the counter/timer boards were activated simultaneously by the Intel 310
  • SUBSTIT improve the dose response curve in the first window and eliminate it in the second window.
  • variations to the assay optimization steps such as wash solution volume, wash solution composition, addition of detergents to the transfer buffered solution, pretreatment of the glass fiber pad by addition of proteins such as fish gelatin, casein, etc., can be applied by those skilled in the art to improve the dose response curve, and these variations lie within the teachings of the invention.
  • the cut-off of this assay was considered to be 25 ng/ml.
  • the data from Table 1 indicate that all test samples which contained 25 ng/ml PCP or higher were well- differentiated from the negative control, which indicated the validity of the assay procedure.
  • Test samples used were calibrators from a commercially available opiates fluorescence polarization kit (TD X ® kit, Abbott Laboratories, North Chicago, IL) which contained 1000, 600, 350, 200, 100 and 0 ng/ml morphine in human urine.
  • 80 ⁇ l of IM ⁇ ® Tris buffer solution (available from Abbott Laboratories, North Chicago, IL) followed by 80 ⁇ l of CelquatTML-200 solution were dispensed on the glass fiber matrices of a disposable reaction tray previously described in Example 2. Solutions were dispensed using two FMI-RH pumps and controlled via a triac board by an Intel 310 Development System (Intel Inc., Sunnyvale, CA).
  • the tray was moved on a linear track using a timing belt and a stepper motor.
  • the stepper motor was controlled by a board employing components known to those skilled in the art.
  • 50 ⁇ l of alkaline phosphatase-iabeled anti-morphine antibody was dispensed into each incubation well.
  • the mixture was incubated for 9.6 minutes while the disposable reaction tray was moving on the track in a temperature controlled tunnel at 32°C.
  • the timing belt steps at the rate of 0.8 inches per minute, the reaction tray was stationary for 36 seconds after each step for a reaction step to take place.
  • a chemiluminescence detector (described previously) was located. After a 4.8 minute substrate incubation time had elapsed, 85 ⁇ l of 0.3% alkaline peroxide trigger solution was injected, and the resulting chemiluminescence signal was integrated for eight (8) seconds after an eight (8) seconds delay from the onset of trigger solution injection to give the intensity of the long-lived chemiluminescence signal.
  • the results of the assay are shown in Table 2.
  • the cut-off of this assay was considered to be 100 ng/ml.
  • the data in Table 2 indicate that all test samples which contained 100 ng/ml morphine or higher were well-differentiated from the negative control, which indicated the validity of the assay procedure.
  • Short -lived signal (convoluted)- short-lived signal - (long-lived signal x 0.655 )
  • Table 2 show that the chemiluminescence signal in the long-lived window is the dose response for morphine in the sample, while the corrected short-lived signal is independent of morphine concentration.
  • a different time window or a signal deconvolution scheme can be applied by those skilled in the art to improve the dose response curve in the first window and eliminate it in the second window.
  • variations to the assay optimization steps such as wash solution volume, wash solution composition, addition of detergents to the transfer buffered solution, pretreatment of the glass fibrous pad by addition of proteins such as fish gelatin, casein, etc. are variations which can also be applied by those skilled in the art to improve the dose response curve and lie within the teachings of this invention.
  • Test samples used were phenylcyclidine calibrators from a commercially available fluorescence polarization kit (TD X ® kit, Abbott Laboratories, North Chicago, IL) at 500, 250, 120, 60, 25 and 0 ng/ml, or TD ® opiate calibrators (commercially available from Abbott Laboratories, North Chicago, IL) at 1000, 600, 350, 200, 100 and 0 ng/ml prepared in human urine.
  • IM ⁇ ® Tris buffer solution available from Abbott Laboratories, North Chicago, IL
  • CelquatTML-200 solution were dispensed on glass fiber matrices of the disposable reaction tray previously described. Solutions were dispensed using two FMI-RH pumps and controlled via a triac board by an Intel 310 Development System (Intel, Inc., Sunnyvale, CA). The tray was moved on a linear track using a timing belt and stepper motor. The stepper motor was controlled by a board employing components known to those of ordinary skill in the art. After 4.8 minutes, 50 ⁇ l of test sample was pipetted into the shallow reaction well of the disposable reaction tray, using an automated pipettor.
  • This mixture was incubated for 9.6 minutes while the disposable reaction tray was moving on the track in a temperature controlled tunnel at 32°C.
  • the timing belt steps at the rate of 0.8 inches per minute, the reaction tray was stationary for 36 seconds after each step for a reaction step to take place.
  • 50 ⁇ l of the simultaneous assay capture reagent solution containing PCP-PGA at a concentration of 1.875 ⁇ g/ml and morphine-ovalbumin-PGA at a concentration of 0.25 ⁇ g/ml prepared as in Example 1 was dispensed into the incubation well through a tip centered on the well. The reaction mixture was further incubated while the disposable tray continued movement along the track.
  • each quaternary ammonium polymer- treated glass fiber matrix was rinsed with 100 ⁇ l of IM ⁇ ® Tris buffer dispensed from a tip centered on the detection well. After 4.8 more minutes of incubation, the disposable reaction tray was located under the transfer device described in co- pending U. S. Patent Application Serial No. 425,643. The 150 ⁇ i assay mixture then was transferred from the shallow incubation well onto the pre-treated glass fiber matrix in the detection well. Transfer was affected using one 350 ⁇ l pulse of IM X ® Tris buffer injected from three adjacent nozzles at a linear rate of 210 cm/sec.
  • the substrate solution was dispensed in each detection well to saturate the fibrous matrix.
  • the disposable tray was moved on the timing belt to allow subsequent well pairs to be located under the transfer device to effect transfer of the reaction mixture.
  • the disposable device then was moved to a detection position where a chemiluminescence detector (described previously) was located.
  • a chemiluminescence detector (described previously) was located.
  • 85 ⁇ l of 0.3% alkaline peroxide trigger solution was injected into the detection well.
  • the resulting chemiluminescence was integrated for eight (8) seconds immediately after the onset of trigger solution injection to give the intensity of the short-lived chemiluminescence signal.
  • the chemiluminescence signal was integrated for another eight (8) subsequent seconds to give the long-lived chemiluminescence signal.
  • the results of the assay are shown in Tables 3 and 4.
  • the cut-off of the PCP assay was considered to be 25 ng/ml.
  • the data in Tables 3 and 4 indicate that all test samples which contained 25 ng/ml PCP or higher were well-differentiated from the negative control.
  • the cut-off of the morphine assay was considered to be 100 ng/ml.
  • the data in Tables 3 and 4 indicate that the test samples which contained 100 ng/ml morphine or higher were well-differentiated from the negative control.
  • the data also indicates that the cut-off of either assay was not affected by using the simultaneous assay procedure as described herein.
  • a longer time window or a signal deconvolution scheme are variations which can be applied by those skilled in the art to improve the dose response curve in the first window and eliminate it in the second window.
  • variations of assay optimization steps such as wash solution volume, wash solution composition, addition of detergents to the transfer buffered solution, pretreatment of the glass fibrous matrix by addition of proteins such as fish gelatin, casein, etc., are variations which can also be applied by those skilled in the art to improve the dose response curve, and these variations lie within the teachings of this invention.
  • PCP [Morphine] Short-lived %l Long-lived %l ng/ml ng/ml Signal (0-8 sec.) Signal (8-16 sec.)
  • each analyte in the simultaneous assay resembles that in the individual assay of the analyte, which indicates the ability to detect either of the two analytes in a test sample using the novel assay method of the invention.
  • plastic tubes coated with mixtures of antibodies and detected in tube luminometers or plastic beads such as 1/4" polystyrene beads coated with mixtures of antibodies and transferred after the completion of the binding reaction to a test tube and then detected in a tube luminometer can be used as media for simultaneous assays based on chemiluminescence signal time resolution.
  • Microparticles can be made from any suitable paniculate material that are easily recognizable by those skilled in the art, such as polystyrene, polymethyl acrylate, derivatized cellulose fibers, polyacrylamide, and the like.
  • the assay method of this invention may be extended to other small molecules, macromolecules, or to nucleic acid probe assays.
  • the present invention has been described using acridinium sulfonamide-labeled and alkaline phosphatase-labeled compounds as tracers, it is contemplated that other acridinium compounds or their analogs, or other chemiluminescent compounds (labels) may be used.
  • the assay of the present invention may be employed for the detection of viral particles, such as HBsAg or HIV antigens, or specific fragments thereof. Simultaneous assays for macromolecular disease state markers, such as carcino- embryonic antigen (CEA) and alpha-fetoprotein (AFP) may also be performed, as well as nutritional status markers, such as simultaneous determination of Vitamin B-
  • CCA carcino- embryonic antigen
  • AFP alpha-fetoprotein
  • nutritional status markers such as simultaneous determination of Vitamin B-
  • hormones such as LH and FSH, bacteria (e.g., streptococci), nucleic acid species (e.g., DNA or RNA).
  • the present invention also is useful in small molecular competitive binding assays such as those for T3 and T4 and digoxin and its analogs.
  • Substances of abuse and their metabolites such as cocaine and benzolecogonine in serum, nicotine and cotinine, codeine and nor-cod ⁇ ine, diazepam and nor-diazepam may be detected using the method of the present invention.
  • Simultaneous assay of therapeutic drugs and simultaneous determination of two steroids also can be performed according to the method of the present invention.
  • Other combinations of analytes can be contemplated and assayed for, by those skilled in the art, using the method of this invention.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Analytical Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Pathology (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Investigating Or Analysing Materials By The Use Of Chemical Reactions (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

Techniques de détermination d'analytes multiples pouvant se trouver dans un échantillon de test consistant à utiliser différents traceurs chimioluminescents à durées de vie courte et longue, à intégrer le signal de chimioluminescence généré et à procéder à une discrimination temporelle des constituants à durées de vie courte et longue du signal généré. L'invention concerne également des matériels contenant ces composés chimioluminescents à durées de vie courte et longue.
PCT/US1991/009714 1990-12-28 1991-12-23 Determination simultanee d'analytes multiples a l'aide d'une technique de chimioluminescence heterogene a resolution temporelle WO1992012255A1 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CA002098020A CA2098020C (fr) 1991-01-02 1991-12-20 Composes antiviraux
JP4502981A JPH06504374A (ja) 1990-12-28 1991-12-23 時間分解不均一化学ルミネッセンスアッセイを用いた複数の分析対象物の同時決定
KR1019930701951A KR930703461A (ko) 1990-12-28 1992-12-23 시간-분리된 이종 화학발광 검정을 이용한 다중 분석물의 동시측정방법

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US63603890A 1990-12-28 1990-12-28
US636,038 1990-12-28

Publications (1)

Publication Number Publication Date
WO1992012255A1 true WO1992012255A1 (fr) 1992-07-23

Family

ID=24550134

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1991/009714 WO1992012255A1 (fr) 1990-12-28 1991-12-23 Determination simultanee d'analytes multiples a l'aide d'une technique de chimioluminescence heterogene a resolution temporelle

Country Status (6)

Country Link
EP (1) EP0566629A4 (fr)
JP (1) JPH06504374A (fr)
KR (1) KR930703461A (fr)
AU (1) AU9146491A (fr)
CA (1) CA2098617A1 (fr)
WO (1) WO1992012255A1 (fr)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0617286A2 (fr) * 1993-03-18 1994-09-28 Wallac Oy Support biospécifique en phase solide
EP0623821A2 (fr) * 1993-05-06 1994-11-09 Ciba Corning Diagnostics Corp. Essais utilisant des conjugués luminescents mixtes
US5395752A (en) * 1993-03-19 1995-03-07 Ciba Corning Diagnostics Corp. Long emission wavelength chemiluminescent compounds and their use in test assays
DE19500862A1 (de) * 1994-01-13 1995-07-20 Abion Ohg Reaktionssäulen für simultane Mehrfachmessung und Verfahren zur Bestimmung von Verbindungen
EP0617287A3 (fr) * 1993-03-16 1995-07-26 Wallac Oy Dosage biospécifique.
EP0709466A2 (fr) * 1994-10-28 1996-05-01 Gen-Probe Incorporated Compositions et procédés pour la détection et quantification simultanée de plusieurs séquences d'acide nucléique
WO1996019731A2 (fr) * 1994-12-22 1996-06-27 Abbott Laboratories Procede de detection, par mesure de temps differentielle, de plusieurs analytes dans un echantillon test
WO1998001578A1 (fr) * 1996-07-06 1998-01-15 Boehringer Mannheim Gmbh Detection d'analytes au moyen de deux marqueurs
WO1999056130A1 (fr) * 1998-04-29 1999-11-04 Queen Mary & Westfield College Procede de dosage de plusieurs analytes
EP1146338A1 (fr) * 1999-11-19 2001-10-17 Hitachi Software Engineering Co., Ltd. Lecteur de biopuce et reactif de marquage
US7211441B2 (en) 2000-01-18 2007-05-01 Council For The Central Laboratory Of The Research Councils Lipoprotein assay
US9414777B2 (en) 2004-07-13 2016-08-16 Dexcom, Inc. Transcutaneous analyte sensor
US9986942B2 (en) 2004-07-13 2018-06-05 Dexcom, Inc. Analyte sensor
US10610137B2 (en) 2005-03-10 2020-04-07 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US10813577B2 (en) 2005-06-21 2020-10-27 Dexcom, Inc. Analyte sensor
US11437141B2 (en) 2009-04-30 2022-09-06 Dexcom, Inc. Performance reports associated with continuous sensor data from multiple analysis time periods

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4761382A (en) * 1982-09-10 1988-08-02 The Welsh National School Of Medicine Immunological procedure for detecting or quantifying substances
US4824775A (en) * 1985-01-03 1989-04-25 Molecular Diagnostics, Inc. Cells labeled with multiple Fluorophores bound to a nucleic acid carrier
US4923819A (en) * 1987-03-27 1990-05-08 Chimerix Corporation Time-resolved fluorescence immunoassay
US4931223A (en) * 1986-07-24 1990-06-05 Tropix, Inc. Methods of using chemiluminescent 1,2-dioxetanes

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4761382A (en) * 1982-09-10 1988-08-02 The Welsh National School Of Medicine Immunological procedure for detecting or quantifying substances
US4824775A (en) * 1985-01-03 1989-04-25 Molecular Diagnostics, Inc. Cells labeled with multiple Fluorophores bound to a nucleic acid carrier
US4931223A (en) * 1986-07-24 1990-06-05 Tropix, Inc. Methods of using chemiluminescent 1,2-dioxetanes
US4923819A (en) * 1987-03-27 1990-05-08 Chimerix Corporation Time-resolved fluorescence immunoassay

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Clinical Chemistry, Volume 35, No. 8, issued 1989, ARNOLD Jr. et al., "Assay Formats Involving Acridinium-Ester-Labeled DNA Probes", pages 1588-1594, see the Abstract. *
Clinical. Chim. Acta, Volume 194, No. 1, issued 17 December 1990, NELSON et al., "Chemiluminescent DNA Probes: A Comparison of the Acridinium Ester and Dioxetane Detectron Systems and their use in Clinical Diagnostic Assays", Abstract No. 91160169, see the entire Abstract, especially the last 6 lines. *
See also references of EP0566629A4 *
Tetrahedron Letters, Volume 28, No. 11, issued 1987, SCHAAP et al., "Chemical and Enzymatic Triggering of 1,2-Dioxetanes. 3: Alkaline Phosphatase-Catalyzed Chemiluminescence from an Aryl Phosphate-Substituted Dioxetane", pages 1159-1162, see the first paragraph of page 1159. *

Cited By (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0617287A3 (fr) * 1993-03-16 1995-07-26 Wallac Oy Dosage biospécifique.
EP0617286A3 (fr) * 1993-03-18 1995-07-26 Wallac Oy Support biospécifique en phase solide.
EP0617286A2 (fr) * 1993-03-18 1994-09-28 Wallac Oy Support biospécifique en phase solide
US5395752A (en) * 1993-03-19 1995-03-07 Ciba Corning Diagnostics Corp. Long emission wavelength chemiluminescent compounds and their use in test assays
US5879894A (en) * 1993-03-19 1999-03-09 Chiron Diagnostics Corporation Long emission wavelength chemiluminescent compounds and their use in test assays
US5702887A (en) * 1993-03-19 1997-12-30 Chiron Diagnostics Corporation Long emission wavelength chemiluminescent compounds and their use in test assays
AU679008B2 (en) * 1993-05-06 1997-06-19 Chiron Diagnostics Corporation Mixed luminescent conjugate test assays
EP0623821A2 (fr) * 1993-05-06 1994-11-09 Ciba Corning Diagnostics Corp. Essais utilisant des conjugués luminescents mixtes
EP0623821A3 (fr) * 1993-05-06 1995-09-20 Ciba Corning Diagnostics Corp Essais utilisant des conjugués luminescents mixtes.
US5672475A (en) * 1993-05-06 1997-09-30 Chiron Diagnostics Corporation Mixed luminescent conjugate test
DE19500862A1 (de) * 1994-01-13 1995-07-20 Abion Ohg Reaktionssäulen für simultane Mehrfachmessung und Verfahren zur Bestimmung von Verbindungen
US5756709A (en) * 1994-10-28 1998-05-26 Gen-Probe Incorporated Compositions for the simultaneous detection and quantification of multiple specific nucleic acid sequences
EP0709466A2 (fr) * 1994-10-28 1996-05-01 Gen-Probe Incorporated Compositions et procédés pour la détection et quantification simultanée de plusieurs séquences d'acide nucléique
WO1996013612A3 (fr) * 1994-10-28 1996-07-11 Gen Probe Inc Compositions et procedes de detection et de quantification simultanees de sequences specifiques multiples d'acide nucleique
US5658737A (en) * 1994-10-28 1997-08-19 Gen-Probe Incorporated Compositions and methods for the simultaneous detection and quantification of multiple specific nucleic acid sequences
US5840873A (en) * 1994-10-28 1998-11-24 Gen-Probe Incorporated Labeled hybridization assay probes useful for the detection and quantification of multiple nucleic acid sequences
EP0709466A3 (fr) * 1994-10-28 1996-06-12 Gen Probe Inc
US5827656A (en) * 1994-10-28 1998-10-27 Gen-Probe Incorporated Compositions and methods for the simultaneous detection and quantification of multiple specific nucleic acid sequences
WO1996013612A2 (fr) * 1994-10-28 1996-05-09 Gen-Probe Incorporated Compositions et procedes de detection et de quantification simultanees de sequences specifiques multiples d'acide nucleique
WO1996019731A2 (fr) * 1994-12-22 1996-06-27 Abbott Laboratories Procede de detection, par mesure de temps differentielle, de plusieurs analytes dans un echantillon test
WO1996019731A3 (fr) * 1994-12-22 1996-09-19 Abbott Lab Procede de detection, par mesure de temps differentielle, de plusieurs analytes dans un echantillon test
WO1998001578A1 (fr) * 1996-07-06 1998-01-15 Boehringer Mannheim Gmbh Detection d'analytes au moyen de deux marqueurs
WO1999056130A1 (fr) * 1998-04-29 1999-11-04 Queen Mary & Westfield College Procede de dosage de plusieurs analytes
EP1146338A1 (fr) * 1999-11-19 2001-10-17 Hitachi Software Engineering Co., Ltd. Lecteur de biopuce et reactif de marquage
EP1146338A4 (fr) * 1999-11-19 2004-09-22 Hitachi Software Eng Lecteur de biopuce et reactif de marquage
US7211441B2 (en) 2000-01-18 2007-05-01 Council For The Central Laboratory Of The Research Councils Lipoprotein assay
US10918314B2 (en) 2004-07-13 2021-02-16 Dexcom, Inc. Analyte sensor
US10722152B2 (en) 2004-07-13 2020-07-28 Dexcom, Inc. Analyte sensor
US10524703B2 (en) 2004-07-13 2020-01-07 Dexcom, Inc. Transcutaneous analyte sensor
US11883164B2 (en) 2004-07-13 2024-01-30 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US11064917B2 (en) 2004-07-13 2021-07-20 Dexcom, Inc. Analyte sensor
US11045120B2 (en) 2004-07-13 2021-06-29 Dexcom, Inc. Analyte sensor
US11026605B1 (en) 2004-07-13 2021-06-08 Dexcom, Inc. Analyte sensor
US10709362B2 (en) 2004-07-13 2020-07-14 Dexcom, Inc. Analyte sensor
US10709363B2 (en) 2004-07-13 2020-07-14 Dexcom, Inc. Analyte sensor
US10993641B2 (en) 2004-07-13 2021-05-04 Dexcom, Inc. Analyte sensor
US9986942B2 (en) 2004-07-13 2018-06-05 Dexcom, Inc. Analyte sensor
US10918315B2 (en) 2004-07-13 2021-02-16 Dexcom, Inc. Analyte sensor
US10993642B2 (en) 2004-07-13 2021-05-04 Dexcom, Inc. Analyte sensor
US10799158B2 (en) 2004-07-13 2020-10-13 Dexcom, Inc. Analyte sensor
US10799159B2 (en) 2004-07-13 2020-10-13 Dexcom, Inc. Analyte sensor
US10813576B2 (en) 2004-07-13 2020-10-27 Dexcom, Inc. Analyte sensor
US10980452B2 (en) 2004-07-13 2021-04-20 Dexcom, Inc. Analyte sensor
US10827956B2 (en) 2004-07-13 2020-11-10 Dexcom, Inc. Analyte sensor
US10932700B2 (en) 2004-07-13 2021-03-02 Dexcom, Inc. Analyte sensor
US9414777B2 (en) 2004-07-13 2016-08-16 Dexcom, Inc. Transcutaneous analyte sensor
US10918313B2 (en) 2004-07-13 2021-02-16 Dexcom, Inc. Analyte sensor
US10716498B2 (en) 2005-03-10 2020-07-21 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US10709364B2 (en) 2005-03-10 2020-07-14 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US10918318B2 (en) 2005-03-10 2021-02-16 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US10918317B2 (en) 2005-03-10 2021-02-16 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US10918316B2 (en) 2005-03-10 2021-02-16 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US10925524B2 (en) 2005-03-10 2021-02-23 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US10856787B2 (en) 2005-03-10 2020-12-08 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US10610137B2 (en) 2005-03-10 2020-04-07 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US10743801B2 (en) 2005-03-10 2020-08-18 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US10898114B2 (en) 2005-03-10 2021-01-26 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US11000213B2 (en) 2005-03-10 2021-05-11 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US10617336B2 (en) 2005-03-10 2020-04-14 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US10610136B2 (en) 2005-03-10 2020-04-07 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US11051726B2 (en) 2005-03-10 2021-07-06 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US10610135B2 (en) 2005-03-10 2020-04-07 Dexcom, Inc. System and methods for processing analyte sensor data for sensor calibration
US10813577B2 (en) 2005-06-21 2020-10-27 Dexcom, Inc. Analyte sensor
US11437141B2 (en) 2009-04-30 2022-09-06 Dexcom, Inc. Performance reports associated with continuous sensor data from multiple analysis time periods

Also Published As

Publication number Publication date
EP0566629A1 (fr) 1993-10-27
AU9146491A (en) 1992-08-17
KR930703461A (ko) 1993-11-30
JPH06504374A (ja) 1994-05-19
EP0566629A4 (fr) 1995-01-25
CA2098617A1 (fr) 1992-06-29

Similar Documents

Publication Publication Date Title
JP3611579B2 (ja) 抗体検出用化学発光イムノアッセイ
EP0424634B1 (fr) Méthode et appareil pour des tests de chemiluminescence en phase hétérogène
US7867781B2 (en) Detection methods
Self et al. Advances in immunoassay technology
Yolken Enzyme-linked immunosorbent assay (ELISA): a practical tool for rapid diagnosis of viruses and other infectious agents.
JP4074335B2 (ja) 電子数の多い化学発光性アリール置換1,2−ジオキセタン
WO1992012255A1 (fr) Determination simultanee d'analytes multiples a l'aide d'une technique de chimioluminescence heterogene a resolution temporelle
NZ504710A (en) Chromatography assay for the detection of analytes in blood
JPH08240590A (ja) 特異的結合アッセイ用試薬およびそのキット
CN113156119A (zh) 一种采用血管紧张素转化酶ii(ace2)检测冠状病毒的方法
AU650503B2 (en) Amplified heterogeneous chemiluminescent immunoassay
EP0835451A1 (fr) Dosage par fixation specifique et par chimioluminescence, sans fractionnement
JPH10511460A (ja) 被験サンプル中の複数分析物の時差式検出方法
JP2001511820A (ja) サイクロスポリン誘導体およびその使用
EP0184701B1 (fr) Procédé pour déterminer un ligand
JP2509840B2 (ja) 免疫測定法及び免疫測定用試薬キット
WO2001044499A9 (fr) Composants du complement marque modifie pour immunodosages

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 IT LU MC NL SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 1992902803

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2098617

Country of ref document: CA

WWP Wipo information: published in national office

Ref document number: 1992902803

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1992902803

Country of ref document: EP