CA1200760A - Immunological reagents employing polymeric backbone possessing reactive functional groups - Google Patents

Immunological reagents employing polymeric backbone possessing reactive functional groups

Info

Publication number
CA1200760A
CA1200760A CA000413624A CA413624A CA1200760A CA 1200760 A CA1200760 A CA 1200760A CA 000413624 A CA000413624 A CA 000413624A CA 413624 A CA413624 A CA 413624A CA 1200760 A CA1200760 A CA 1200760A
Authority
CA
Canada
Prior art keywords
polymer
immunological
substance
homolog
water
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired
Application number
CA000413624A
Other languages
French (fr)
Inventor
Arm Azad
Michael C. Brown
Stefan J. Kirchanski
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ortho Clinical Diagnostics Inc
Original Assignee
Ortho Diagnostic Systems Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ortho Diagnostic Systems Inc filed Critical Ortho Diagnostic Systems Inc
Application granted granted Critical
Publication of CA1200760A publication Critical patent/CA1200760A/en
Expired legal-status Critical Current

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
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2474/00Immunochemical assays or immunoassays characterised by detection mode or means of detection
    • G01N2474/20Immunohistochemistry assay
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/962Prevention or removal of interfering materials or reactants or other treatment to enhance results, e.g. determining or preventing nonspecific binding
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/823Immunogenic carrier or carrier per se

Abstract

Abstract An immunological substance detecting reagent is provided which, in the preferred embodiment, employs an immunological homolog specific for the immunological sub-stance to be detected coupled to a water-soluble polymer having a net charge not greater than zero. The water-soluble polymer further has associated a plurality of marker substances such as fluorophore molecules thereby providing a reagent of increased sensitivity for the detection of an antigen-antibody immunological reaction.

Description

q ~Z(~ 7~

IMMUNOI-OGICAL REAGENTS EMPLOYING POLYMERIC ~ACKBONES

~iel~ ~r ~e ~n-en~i-n This invention relates to the field of immunoassays and more specifically, describes an immunoassay reagent formed by the coupling of an immunological homolog specific for the immunological substance ~rO be detected, to a water soluble polymeric backbone capable of ~urther attachment to marker substances~

The deteetion oE foreign subs~ances in body fluids is often essential to the proper diagnosis of a diseased state and selection of the appropriate treatment thereor.
The foreign substances, generally termed antigens, may have associated therewith the capacity to stimulate the formation of a corresponding antibody which reacts specifically with tha~ antigen. The antibody itself i5 a protein that is formed in response to the presence of an antigen for reaction with that ~pecific antigen.
Antibodies comprise a special group of serum proteins called immunoglobulins. Although ~he group of antibodies comprises a r~stricted group of proteins that are capable of specifically reacting with antigens, there is an enormous variety of macromolecules capable of behaving as antigens, including proteins, many polysaccharides, neucleoproteins, lipoproteins, numerous synthetic polypeptides as well as many other small molecules, called haptens, when they are suitably linked to proteins or synthetic polypeptides~

The specificity of antibody-antigen reactions has been utilized in ~he diagnosis of pathological sta~es or physiolo~ical conditions and more particularly, in the OR~-33 ~3t)~

detection of antigenic determinants As used her~in~ the phrase "immunological 3ubstancen ~hall be defined as either an antibody or an antigen while the phrase ~immunological homolog~ ~hall be defined as the complement of the immunological substance which is capable of specific reaction therewith. Conse-quently, if he immunological substance being discussed is an antibody, then the immunological homolog would be the antigen for which that antibody is specif iCo The converse is equally contemplated.

In accordan~e with the knowledge of those skilled in the art, antigen~antibody reactions can be manifested by enzyme immunoassay, radioimmunoassay, or immunofluores-cence techniques with high sensitivity; however, in largepart~ these techniques are limited to the sensitivity of typically comple~ instrumentation de~igned to locate and quantify the marker substances employed.

A well known class of prior art techniques for detecting an antigen~antibody reaction involve!d labeling the antibody with a tag or marker substance. This te¢hnique, however, possesses ~everal disadvantages. The antibody protein is a ve~y sensitive protein whose reactivity, the capability of selectively reacting with its immunological homolog present even in small concentrations, can be easi-ly destroyed by the chemical addition of marker substances to ~he protein, i~e., denaturation. Further problems include the inability to attach a sufficient concentration of weakly fluorescent but desirable dye molecules such as the red excited fluorescent dye in order to get into a detectable range~ On the other hand, if too many dye molecules are attached, then, even though denaturation ORI;30 7~7 of the antibody may not occur, nonspecific staining is likely because of the hydrophobic nature of the antibody-dye complex. Additionally, the loss of specificity occasioned by the presence of cationic charges, present on many dyes, makes such a system undesirable.

The concentration at the site of an immunological reaction of a marker substance (and thus the sensitivity of detec-tion) may be increased by employing an indirect staining technique wherein a second immunoglobulin, directed against the first immunoglobulin, carries several dye molecules attached in normal fashion. Since the second imm~noglobulin is typically heterospecific, i.e., it binds to several sites, the attachment of several second immuno-globulins to the first is possible. Consequently, theattachment of significantly greater numbers of dye mole-cules onto the first immunoglobulin, specific against the antigen to be detected, increases the antigen detection sensitivity. Unfortunately, such a procedure involves two reaction steps making it unsuitable for facile use in automated instrumentation. Additionally, while the second antibody system does increase detection sensitivity, it still suffers from the same limitations present in the directly labelled antibody system first described.
It is an object of the present invention to minimize ~he loss of reactivity and sensitivity due to nonspecific binding occasioned by the above methods.

Another class of well-known prior art techniques employs a biotin-avidin complex which binds by physical adsorption.
Although biotin-avidin does not result in a covalent bond, it nonetheless exhibits a very high binding constant. The biotin is, in relation to the antibody, a comparatively small molecule so that the antibody is capable of carrying a number of biotin molecules on its surface. The addition of several labelled avidins to biotinylated antibody results in specific adsorption thereby yielding labelled antibody. Because the avidin is also a small molecule, less than half the size of an antibody, the amount of dye that can be attached to the avidin is limited. It is an object of the present invention to eliminate this limitation and still enjoy the advantages presented by employment of the biotin-avidin technique for linking an antibody to another substance.

Further attempts to increase the sensitivity of immunolog-ical reaction detection systems have employed the substi-tutions of radioisotopes for dyes. Radioisotopes are physically small labels and thereby minimize steric hindrances and sensitivity losses and permit the most sensitive level of detection. The use of radioisotopes, however, presents numerous disadvantages including the relatively short life of gamma emitting isotopes, e.g.
125I, the impairment of immunological reactivity by gamma radiation of the isotope, health hazards involved in the use of dangerous radioisotopes necessitating the use of procedures complying with federal standards, as well as requiring precise safety controls in addition to expensive, complex detection instrumentation. Further, the radioimmunoassay while suitable for soluble antigens is no~ readily adaptable on a cell by cell basis since it is really an averaging technique. To make the radio-immunoassay sensitive to a single cell for automated flow cytometry instrumentation would require advanced radio-graphic techniques. Such techniques would be expected to be slow and procedurally very complex. As a result, radioisotopes are generally nonsuitable for individual cell analysis. It is an object of the present invention to provide a reagent which will increase the sensitivity of autoradiographic techniques and radioimmuhoassays and to decrease the possibility of radiation damage normally caused by direct labelling due to close physical proximity of the radioactive label to the antibody.

Enzymes, used in substitution for dye marker substances, advantageously provide an improvement in sensitivity due to the great turnover of substrate. ~lthough such a system requires additional steps beyond the use of the typical dye mar~er substance, there is no photobleaching problem as evidenced with the use of fluorescent dyes.
Furthermore, propitious choice of the enzyme permits the production of a colored compound or an acid readily dis-cerned and quantified by automated equipment. Disadvan-tages include the large and sticky nature of enzymes whichoften results in denaturation of the antibody, and a loss of specificity due to the inherent nature of enzymes for nonspecific attachment or steric hindrance from homolog binding. Furthermore, enzymes present shelf life problems which reduce their effectiveness for practical use in clinical environments. If specific structures on the cell are to be localized or measured, then the substrate must precipitate directly on the cell in a conveniently detectable form, e.g., a fluorescent compound. A problem generally encountered with enzyme systems is a limitation in sensitivity because of a fixed and limited enzymatic rate. Compensation can be made for this by simply waiting longer for the enzyme to continue to act upon the substrate~ This, of course, reduces the efficiency of such a technique in clinical applications and its usefulness in automated procedures where system throughput capability is of paramount importance. It is an object of the present invention to provide a reagent, compatible with enzymes, for detecting immunological reactions with increased sensitivity.

ï(}~

Electron opaque stains have found useful application in electron microscopy, however, such a system is far more complex and requires lengthy procedures thereby effective-ly eliminating its clinical value. To some degree, this disadvantage is compensated by an increase in resolutionO
Electron opaque stains include ferritic compounds, i.e., proteins containing iron or colloidal gold, however, it is noted that these compounds may be larger than the antibody and may therefore be expected to deleteriously affect the antibody's reactivity. It is an object of this invention to reduce such an effect on the antibody as well as to provide a reagent that is electronically dense and opaque to electron scanning.

Traditional attempts to overcome many of the above-described problems include the use of polymer particles or microspheres containing fluorescein or other marker type substances. Generally these polymer par~icles are constructed in the range of 50-2000 nanometers and conse quently, the number that can be attached to a cell through an antibody is sterically limited. Typicallyr the particles are coated with large numbers of antibodies;
however, due to the physically large nature of the reagent, there is a greatly reduced chance of the antibody approaching an antigenic site on the cell in an appropriate orientation to result in the immunological reaction. Traditionally, the polymer particles have been made from styrenes and vinyl monomers by emulsion polymerization, however, the resulting hydrophobic particles disadvantageously exhibit nonspecific binding as well as colloidal instability. The latter instability is aggravated by the attachment of antibodies on the particle's surface. Of further disadvantage is the surface denaturation efect on the antibody following attachment of the antibody on the surface of the microsphere.

Attempts have been made to overcome the hydrophobic nature of polymeric particles by the incorporation of hydrophilic monomers into the particlesl however, these attempts have been limited to emulsion polymerization procedures.
Although alteration of the composition of the particles can improve nonspecific binding and colloidal instability problems, these undesirable characteristics cannot be completely eliminated since a minimal amount of hydropho-bic monomer is required in order to synthesize the poly-meric particle. One such polymeric particle detectionsystem is described in U.S. Patent No. 4,254,096 to Monthony et al wherein an assay method is described using antibodies covalently bound to hydrophilic polymeric par-ticles which are water-insoluble, an undesirable characteristic. Even in the case of completely hydrophilic monomers, cross-linking agents are needed in order to make a hydrophilic particle as described in U.S~
Patent No. 3,853,987 to DreyerO It is an object of the present invention to eliminate the requirement for a cross-linking agent.

Hirschfeld and Eaton describe another technique to increase sensitivity using a polymeric backbone in U.S.
Patent No. 4,169,137. They specifically describe an anti-gen detecting reagent consisting of a primary amine con-taining, polyfunctional polymeric bac~bone, i.e., polyly-sine, having coupled therewith a plurality of fluorescent dye molecules~ The polyfunctional polymeric backbone was covalently bound through the primary amine- on the polylysine to a primary amine moiety on the antibody by the use of a dialdehyde such as glutaraldehyde. Although such a method results in greater sensitivity per antigen site, serious limitations are presented by the described methodology. Specifically, the polymer is restricted to a primary amine containing polymer which, because of its : , ;

~.~r~ 7~

cationic charges, exhibits strong nonspecifi~ bi~ding thereby greatly reducing its specificity. In fact, it was found that the type of polymer described by Hirschfeld attached to cells even without an antibody. O further disadvantage is the requirement of a dialdehyde coupling reagent. These restrictions serve to drastically limit the advantageous attachment of various fluorescent dyes to the polymeric backbone~ Also, the employment of a dialdehyde does not permit the attachment of nonprimary amine containing polymeric bac~bones to an antibody. When employed within the pH ranges of interest for biological applications, the described system tends to be cationical-ly charged and therefore attaches nonspecifically, by electrostatic interaction, to anionically charged cell and tissue surfaces. Due to the unstable nature of the dialdehyde formed, covalent linkage and the reversible dialdehyde reaction, it is expected that Hirschfeld's reagent will contain much disassociated polymer and antibody capable of competitively reacting with antigenic sites and thereby reducing the sensitivity of the assay.

It is an object of the present invention to provide an immunological substance detection assay employing a water-soluble polymer to carry many detectors, and to avoid nonspecific attachment due to hydrophobic and cationic charge characteristics as well as to avoid the need for dialdehyde linking agents.

It is another object of the present invention to provide a reagent capable of detecting an immunological substance with increased sensitivity. It is a further object to avoid the problems associated with hydrophobic polymers and particles and cationically charged complexes which disadvantageously result in nonspecific binding by provid-ing properly charged polymers having minimum hydrophobiccharacteristics. It is an object to provide a water-~, ~ d~ 7 ~ ~
~9_ soluble polymer, capable of carrying a pluraiity of marker substances, which does not have a net positive charge.

It is yet another object to provide a reagent capable of simple storage and effective use in automatea instrumenta-tion. It is a still further object to provide a reagent capable of employing the desirable~ but weakly 1uores-cent, red excited fluorescent dyes in concentrations and amounts capable of being detected with the technology presently employed in automated cytology instrumentation.

It is a yet further object to employ a water-soluble polymer in combination with avidin-biotin in order to utilize the individual advantages of each within a single reagent.

Summary of the Invention The principles of the present invention broadly employ direct and indirect marker techniques to aid in the detec-tion of an immunological reaction wherein an immunologic homolog, specific for the substance to be detected, is bound, through a variety of intermediate mechanisms, to water-soluble polymers serving as or carrying detectable marker mechanismsO It is to be understood that the term water-soluble polymer, as used herein, encompasses that meaning as it is used in the conventional organic chemistry sense and does not include what is typically known in the art as microspheres which require covalent cross-linkages or particles which are water-insoluble (hydrophobic) polymers. Consequently, a covalent cross~
linking agent is not needed in the present invention.
Typically, the invention embraces single chain polymers having a weight up to approximately 1 X 10/ daltons and up to approximately 1 x 10~ daltons for branched polymers.

7~

Branched polymers encompass water-soluble polymers which contain a polymeric 'backbone' chain having a plurality of additional polymeric chains, composed of ~he same or different monomers, covalently attached anywhere along the length of the 'backbone' chain. ~ useful analogy for a branched polymer would be to a wire bristle brush which may or may not have missing bristles.

Thus, there is provided an immunological substance detecting reagent comprising:
a) an immunological homolog specific for the substance to be de~ected;
b) a water-soluble, substantially noncross-linked and nonprimary amine containing pol~ner having a net charge not greater than zero; and c) means ~or attaching substantially only one homolog to each water-soluble polymer.

In accordance with the stated objects, the principles of the present invention embrace several embodiments of immunological substance detecting reagents employing a water-soluble polymer.
All embodiments provide for charge specification but differ whether the charge of the polymer, the charge of the polymer having a marker substance attached to form a polymer-marker substance complex, or the charge of the polymer-marker substance complex with means for attaching to the antibody is specified.
All embodiments include means for attaching the polymer to the homolog.

Additional embodiments have the attaching means selected from the group consisting of activated groups on the immunological homologs, activated coupling reagents, activated groups on the polymer, activatable groups on the polymer, and biotin~avidin complex wherein the avidin is bonded to the polymer and the biotin is bonded to the immunological homolog. Additional embodiments may further comprise a marker substance attached to the polymer. Still other embodiments provide for a marker ',~`~ ' 6~1 --lOa-substance selected fro~ the group consisting of nonfluorescent dyes, fluorescent dyes, radioisotopes, electron opaque sub-stances, enzymes, a second immunological homolog of differing specificity than the first immunological homolog and micro-spheres. Preferred embodiments provide for a water-soluble polymer selected from the group consisting of polyacrylic acid, polymethacrylic acid, polyacrylamide, polyvinylalcohol, polyallylalcohol, polymer combinations of the foregoing, hydroxyeth~yl cellulose, hydroxypropyl cellulosel natural water-soluble polymers and synthetic water-soluble polymers.
S

Further objectives and understanding of the present invention will become apparent upon reference ~o the drawings wherein:

Fig~re 1 is a styliæed view of the preferred embodiment showing an immunoglobulin having attached thereto a water-soluble polymer carrying a marker substance other than a microsphere;

Figure 2 illustrates an alternative embodiment of the present invention employing a microsphere as a marker substance;
Figure 3 shows a stylized view of an immunoglobulin attached by the use of biotin-avidin to a water-soluble polymer having microsphere marker substances:

Figure 4 is a stylized view of an embodiment of the present invention for the detection of a cell surface antigen;

~igure 5 is a stylized view of a solid-phase antigen detection system for poly-valent antigens using an embodiment of the present invention; and Figure 6 is another stylized view of a solid-phase antigen detection system using the water-soluble polymer having a marker substance and attached to the antigen of interest.

Detailed Description of the Invention and Pr _erred Embodiments In accordance with the stated objects~ the preferred immunological substance detecting reagent as shown in Figure 1 is provided having an immunological homolog, here an immunoglobulin 1, specific for an immunological substance, here an antigen, to be detected 30. As will be noted by those skilled in the art, the stylized component representations in the Figures have been significantly altered as to relative proportions as well as appearance for the sake of clarity. Also to be noted is that discussion pertaining to one figure may be equally applicable to other figures with the exception of different marker substances present or applica~ion in different assay systems. The immunoglobulin 1 is attached to a water-soluble polymer 2 having attached thereto a plurality of marker substances 3. Marker substances 3 are preferably fluorescent dyes although other mar~er sub-stances such as nonfluorescent dyes, radioisotopes, elec-tron opaque substances, enzymes! or a second homolog of differing specificity than the first homolog, may be used in substitution. Still alternatively, fluorescent-containing microspheres 4 could be used as shown in Figure
2. Also shown in Figure 2 is use of the reagent in a solid-phase immunoassay wherein the antigen 30 to be detected is immobilized on the surface of a substrate 51.

With reference to Figures 1 and 2, the water-soluble polymer 2 is advantageously chosen so as to avoid nonspecific hinding to the cell, thereby permitting greatest sensitivity and immunological response.
Consequently, those polymers which are cationically charged and therefore tend to attach nonspecifically are 3S preferably avoided. Typical of such preferably avoided, ~ (}~

cationically charged polymers are the primary amine-containing polymers.

The preferred water-soluble polymers have an anionic (negative) or zero charge and are ideally chosen from the group consisting o polyacrylic acid, polymethacrylic acid, polyacrylamide, polyvinylalcohol, polyallylalcohol, any of the various possible polymer combinations of these, hydroxyethyl cellulose, hydroxypropyl cellulose, natural water-soluble polymers as well as synthetic water-soluble polymers. It is noted, however, that many of the marker substances 3, 4 tend to be cationically charged, especially the dye-type marker substances. Since this tends to increase unwanted nonspecific binding, the water-soluble polymer may be advantageously adjusted to increase its anionic character and thereby compensate for the O increased nonspecific 'staining' characteristic detrimental to desired sensitivity.

The water-soluble polymer, typically having a molecular weight in the range of 1,000 to 100,000,000 daltans, provides the capability of attaching numerous dye-type substances to an antibody having a desired specificity.
Of great importance is that the attachment is at a sufficient distance from the antibody in order to avoid deleterious effects on the antibody's reactivity. All these considerations become particularly important when dealing with the long wavelength or red light excited fluorescent dyes which characte~istically often exhibit diminished fluorescence in comparison to other dyes such as fluorescein or acridine orange~ Consequently, by loading up the water-soluble polymer 2 with numerous dye molecules 3, a gain in ser.sitivity is accomplished since there is provided a greater concentration of fluorescence thereby enabling presently available automated ~(~7 instrumentation to record more readily the presence of an immunological reaction.

The water-soluble polymer additionally exhibits advantages when used with an enzyme-marker system. Enzymes, like immunoglobulins, are large proteins with an active site which is sensitive to steric hindrance. Direct attachment between an immunoglobulin and an enzyme, although permitting the detection of an immunc,logical reaction between an immunological substance and its homolog following the addition of th~ substrate, has a dlsadvantageously low sensitivity due to steric hindrance.
The advantageous employment o a water-soluble polymer to connect the immunoglobulin to single or multiple enzyme molecules permits each protein to act in the desired fashion unhindered by the presence of the other. As a result, the immunoassay reagent exhibits increased sensitivity in the detection of the immunological substance.
The invention shows additional utility in conjunction with monoclonal antibodies directed against specific tissue or tumor cell associated antigens. The polymer is capable of carrying an increased load of therapeutic treatment substances or agents above that possible only by employment of direct attachment of the agent to the immunoglobulin itself and further, decreases the chance of denaturation of the immunoglobulin by attachment of the agent. As a result, a monoclonal antibody, attached to a water-soluble polymer carrying chemical, vitamins, drugs, radiochemical treatment molecules or other material desired to be delivered to a specific or targeted site, will be able to localize and specifically attack tumor cells and advantageously provide increased concentrations of the treatment material directly to the targeted site.

It is preferred that a biocompatible polymer be employed in the present invention. Biocompatible means that the polymer is chosen, to avoid producing adverse toxic ef-fects, and to avoid nonspecific binding to cells or the formation of lattice-type structures yielding agglutination-type aggregates. Consequently, preferred embodiments employ polyacrylic acid and polyacrylamide, both polymers being formed from water-soluble monomers polymerized by a free-radical process although anionic/
cationic polymerization and condensation methods may also be used. Additionally, it has been found advantageous to employ monomers having functional groups to produce the polymer so that the functional groups may be later utilized to react wi~h a market substance such as a dye, or ~nological subs'.ance 1~ Examples of such monomers havlng functional groups include acryloyl chloride. The polyacryloyl chloride produced has an active chlorine atom capable of reacting with a primary amine to form an amide linkage. To be noted, however, i5 that the functional groups are not restricted and other groups including, but not limited to, hydroxyl thiocyanate, isothiocyanate, sulfonyl chloride, succinimide, and dichlorotriazinyl may be employed in substitution.

In accordance with organlc procedures well known, it may be necessary to warm the dye and/or put it in the presence of a catalyst in order to stimulate its reaction with the functional group on the polymer. The amount of dye added to the polymer may be advantageously regulated by controlling the concentration ratios or by utili~ing different functional groups, one for the dye and one for the antibody. This, of course, is made possible by the fact that the dye typically has only one functional group reactive site to react with the polymer.

?(~

In accordance with known skills, other methods which may be advantageously employed to attach the marker substances to the polymer include a) utilizing an activate~ group on the marker substance, b) employing an external activating reagent, c~ in those marker substances having a first - portion and a second portion, polymerizing the monomer having the first portion attached to form the polymer, and then adding the second portion for attachment to the first portion, and d) copolymerization of the marker substance with the monomer.

Figure 3 demonstrates the utilization of avidin 6-biotin 5 complex in order to effectuate the linking of immunoglobu-lin 1 to polymer 2 carrying, in this case, microspheres 4.
The polymer could just as easily be the carrier of a different marker-type substance such as a dye or other member selected from the marker substance group previously described. ~iotin-avidin may be advantageously employed ~ecause of its unusually strong binding characteristics, its chemical stability, and sensitivity amplification.
The biotin 5 is easily attached to the immunoglobulin by methods well known, and because of its small si~e, has little or no effect upon the reactivity of the lmmunoglobulin and further, several may be at~ached to the antibody to yield increased sensitivity. In contrast to biotîn, the avidin molecule is considerably larger and in the past has been utilized to directly carry fluorescein and other dye-type materials. However, the present invention advantageously employs avidin merely to couple the polymer to the antibody. Since the polymer is capable of carrying far more fluorescein or other dye-type material than avidin, the result is a greater concentra-tion of dye associated with the immunological homolog which in t~rn provides greater sensitivity. Although it is difficult to ensure that only one polymer molecule is ~,7~1r~(~7~

attached per avidin molecule, in compensation, the bio~i-nilation of antibodie~ can be controlled more precisely.

-Figure 4 demonstrates the utili~ation o~ the present invention in the determination and localization of an immunological substance or antigenic site 21 on a cell surface 20 by the use of the immunological homolog or immunoglobulin 1 having attached thereto biotin 5. The biotin physically adsorbs to avidin 6 attached to the water-soluble polymer 2 which, in turn, is capable of carrying marker substances 3. With those reagents employing biotin-avidin~ it is preferred that just the biotinylated antibody is first reacted with the antigens and then khe avidin coupled portion of the reagent is added.

The flexibility of the present invention is further illustrated in Figure S wherein a stylized view of a solid-phase immunoassay is demonstratedO An immunoglobu-lin 1, sensitive for the antigen to be detected 71, isimmobili~ed on a surace 51 and the sample containing the antigen is contacted with the immobili~ed immunoglobu-lin surface. Subsequently the reagent of the present invention, having an immunoglobulin 1 sensitive for the antigen and attached to a polymer 2 having marker substances 3 i5 washed over the surface. Quantifying the fluorescence or presence of other marker substance will permi~ direct calculation as to the quantity of antigen present and its precise localization. In the expanded view, the marker substance is an enzyme 40 which is reactive with substrate 41 and breaks it down to 42 and 4 3 which may be individually detected.

Figure 6 further demonstrates the enormous flexibility of
3 5 the present invent ion and shows the attachment of a water-. ~ . ~
soluble polymer 2 carrying marker substances 3 to an antigen 80 which thereupon may be ontacted with an immunoglobulin 1 specific for the antigen and which immunoglobulin may be immobilized on the surface 51 or may in turn be attached to another polymer carrying marker substances. Further, there may be present unlabelled antigens 80 and an inhibition-type assay employed utilizing methods well-known in the art.

It is to be understood that, as implied by the use of ~he ~erms immunological substance and immunological homolog, in many cases, the polymer carrying the marker substances can be attached to the anti~en to form a reagent capable of detecting the presence of an antibody immobilized on the surface of a cell or substrate or present in an 1~ aqueous solution.

It is further to be understood that many other alterna-tives or combinations of components will occur to those skilled in the art without departing from the spirt or scope of the invention.

A copolymer, of approximately 50~ (by weight) acrylic acid and 50% (by weight) acrylamide and containing one fluores-cent monomer per 100 nonfluorescent monomers, was produced by dissolvmg the monomers in dImethylfonm~mide (DMFj and adding 2.5% (by weight of m~ncmer) 4,4-A2Obis(4-cyanovaleric acid), as an initiator~ and polymerizing for 16 hours At 65C under nitrogen. The fluorescent monomer was produced by reacting fluoresceinamine and acryloylchloride in equi-molor amounts at room temperature. After polymerization, the nonpolymerized components were removed by gel filtra-tion on Biogel P-10 in anhydrous fonE~de . Fifteen mg of fluorescent polymers were activated by treating the poly-mer with 34 mg of carbonyl diimidazole ~amount equal to 2X

?(~

the available number of carboxyl groups) and allowing it to react for 30 minutes at room temperature. Sixty mg of N-Hydroxysuccinimide (5X the available number of carboxyl groups) were added and allowed to react a~ room tempera-ture for 30 minutes. The resulting polymer containingsuccinimide ester was concentrated by precipitation with anhydrous acetone and, after washing with acetone, was stored in acetone.

For the monoclonal antibody experiments, 1 mg of activated p~lymer was dried with nitrogen. One-half ml of sodium borate (0~125 M~ and 200 ~9 of OKT3 (T-cell directed monoclonal antibody available from Ortho Pharmaceutical Corporation, Route 202, Raritan, N.J.) in 100 ~1 phosphate buffered saline were added to the water-soluble polymer and the mixture was incubated for 30 minutes at room temperature. The resulting OKT3-polymer was assayed using the Ortho protocol for T-cell enumeration (this protocol is incorporated by reference and available from Ortho Diagnostic Systems Inc., Route 202, Raritan, N.J.).
Essentially the procedure comprises the following: whole blood is reacted with fluorescently labelled antibody~
Following lysis of the red cells with an ammonium chloride based lysing reagent, lymphocytes are distinguished from other leucocytes by their forward and 90 light scatter characteristics. The ORTHO SPECTRUM III~ flow cytometer (available from Ortho Diagnostic Systems) collects fluorescence data and identifies lymphocytes by their characteristic light scatter as specified above. This provides a histogram display in which labelled versus unlabelled lymphocytes can be distinguished and enumerated. Assayed in this manner, polymer-OKT3 showed 64~ OKT3 positive lymphocytes while conventional OKT3 showed 66% positive lymphocytes (not significantly different). At the same time polymer-OKT3 showed enhanced labelling in that the mean fluorescence was higher and there were many more counts in the highest fluorescence channels.

Exam~le 2 ____ Two hundred ~g of OKT3 ( 1. 3 X 10~9 moles) was reacted with 2~6 X 10 8 moles of biotinyl N-hydroxysuccinimide (2.6 ~1 in DMF) in a inal volume of 200 ~1 of 0.1 M sodium borate for two hours at room temperature. This treatment introduced a calculated 10 biotin groups per antibody assuming S0% labelling. Meanwhile, 680 ~g of purified avidin was added to 2.5 mg of dried, activated fluorescein-polymer prepared as in Example 1 in 200 ~1 of 0.125 M sodium bora~e and reacted at room temperature for 1 hour. The avidin was then diluted to 1 ml total volume with phosphate buffered saline~ Ten ~1 of biotinylated OKT3 was mixed with 50 ~1 of buffy coat (5 X 105 leucocytes) and incubated for 30 minutes at room temperature. Following 3 washes with 1.5 ml phosphate buffered saline each to remove non-bound OKT3~biotin, 20 ~1 of avidin-polymer were added (fluorescein-avidin was also used for comparison purposes) and the mixture ~as incubated for 30 minutes at room temperature. The samples were washed twice wi~h 1.5 ml phosphate buffered saline each, red cells were lysed as in Example 19 and then analyzed with the ORTHO SPECTR~M III~ as in Example 1.
The results showed enhancement of labelling compared to conventionally labelled avidin. This demonstrates that the fluorescent polymer is readily detectable and became covalently bound to avidin which~ in turn, then bound successfully to the biotinylated O~T3 on the cell surface.

7~

The skilled man will readily appreciate that other marker substances may be used in su~stitution for fluorescein under appropriate conditions readily discernible by those skilled in the art.

Claims (17)

What is Claimed is:
1. An immunological substance detecting reagent comprising:
(a) an immunological homolog specific for the substance to be detected;
(b) a water-soluble, substantially noncross-linked and nonprimary amine containing polymer having a net charge not greater than zero; and (c) means for attaching substantially only one homolog to each water-soluble polymer.
2. An immunological substance detecting reagent comprising:
(a) an immunological homolog specific for the substance to be detected;
(b) a water-soluble polymer-marker substance complex having in combination a net charge not greater than zero, said polymer being nonprimary amine containing and substantially noncross-linked; and (c) means for attaching substantially only one homolog to each water-soluble polymer.
3. An immunological substance detecting reagent comprising:
(a) an immunological homolog specific for the substance to be detected; and (b) a water-soluble polymer having a marker substance associated and means for attaching substantially only one homolog to each polymer, the entire polymer-marker-attaching means complex having a net charge not greater than zero, said polymer being nonprimary amine containing and substantially noncross-linked.
4. An immunological substance detecting reagent as provided in claims 1 or 2 wherein the attaching means is selected from the group consisting of activated groups on the homolog, activated coupling reagents, activated groups on the polymer, activatable groups on the polymer, covalent bonds, and biotin-avidin complex.
5. An immunological substance detecting reagent as provided in claim 1 wherein the reagent further comprises a marker substance attached to the polymer.
6. An immunological substance detecting reagent as provided in claims 2, 3 or 5 wherein the marker substance is selected from the group consisting of nonfluorescent dyes, fluorescent dyes, radioisotopes, electron opaque substances, enzymes, a second immunological homolog of differing specificity than the first homolog and microspheres.
7. An immunological substance detecting reagent as provided in claims 1, 2 or 3 wherein the water-soluble polymer is selected from the group consisting of polyacrylic acid, polymethacrylic acid, polyacrylamide, polyvinylalcohol, polyallylalcohol, polymer combinations of the foregoing, hydroxyethyl cellulose, hydroxypropyl cellulose, natural water-soluble polymers and synthetic water-soluble polymers.
8. An immunological substance detecting reagent comprising:
(a) a first immunological homolog specific for the substance to be detected;
(b) a water-soluble, nonprimary amine containing and substantially noncross-linked polymer having a charge not greater than zero and selected from the group consisting of polyacylic acid, polymethacrylic acid, polyacrylamide, polyvinylalcohol, polyallyalcohol, polymer combinations of the foregoing, hydroxyethyl cellulose, hydroxypropyl cellulose, natural water-soluble polymers and synthetic water-soluble polymers; and the polymer further having in association therewith a marker substance selected from the group consisting of nonfluorescent dyes, fluorescent dyes, radioisotopes, electron opaque substances, enzymes, a second immunological homolog of differing specificity than the first homolog and microspheres; and (c) means for attaching substantially only one first homolog to each polymer-marker complex.
9. An immunological substance detecting reagent comprising:
(a) a first immunological homolog specific for the substance to be detected;
(b) a water-soluble, nonprimary amine containing and substantially noncross-linked polymer selected from the group consisting of polyacrylic acid, polymethacrylic acid, polyacrylamide, polyvinylalcohol, polyallylalcohol, polymer combinations of the foregoing, hydroxyethyl cellulose, hydroxypropyl cellulose, natural water-soluble polymers and synthetic water-soluble polymers, and having in association therewith a marker substance selected from the group consisting of nonfluorescent dyes, fluorescent dyes, radioisotopes, electron opaque substances, enzymes, a second immunological homolog of differing specificity than the first homolog and microspheres, the polymer-marker complex having a net charge not greater than zero; and (c) means for attaching substantially only one first homolog to each polymer-marker complex.
10. An immunological substance detecting reagent comprising:
(a) a first immunological homolog specific for the substance to be detected; and (b) A water-soluble, nonprimary amine containing and substantially noncross-linked polymer, selected from the group consisting of polyacrylic acid, polymethacrylic acid, polyacrylamide, polyvinyl alcohol, polyallylalcohol, polymer combinations of the foregoing, hydroxyethyl cellulose, hydroxypropyl cellulose, natural water-soluble polymers and synthetic water-soluble polymers, the polymer having in association therewith a marker substance selected from the group consisting of nonfluorescent dyes, fluorescent dyes, radioisotopes, electron opaque substances, enzymes, a second immunological homolog of differing specificity than the first homolog and microspheres, and the polymer further having in combination therewith means for attaching substantially only one first homolog to each polymer, the entire polymer-marker-attaching means complex having a net charge not greater than zero.
11. An immunological substance detecting reagent as provided in claims 8, 9 or 10 wherein the attaching means is selected from the group consisting of activated groups on the homolog, activated coupling reagents, activated groups on the polymer, activatable groups on the polymer, covalent bonds, and biotin-avidin complex.
12. An immunological substance detecting reagent as provided in claims 2, 3 or 5 wherein the attaching means is a complex of biotin-avidin and the marker substance is a fluorescent dye.
13. A reagent as provided in claim 5 wherein:
(a) the immunological substance is an antigen present at a site associated with tissue;
(b) the immunological homolog is an antibody specific for the antigen; and (c) the marker substance is substituted with a thera-peutic substance selected from the group consisting of chemotherapeutic substances, radiotherapeutic substances, vitamins, tumor-inhibiting drugs, and tissue affecting materials desired to be delivered to the antigenic site.
14. A treatment agent specific for tissue having an associated antigen comprising:
(a) a water-soluble, nonprimary amine containing and substantially noncross-linked polymer having a net charge not greater than zero;
(b) an antibody, specific for the antigen, bound to the polymer on a substantially one antibody per polymer basis; and (c) a therapeutic substance bound to the polymer and selected from the group consisting of chemotherapeutic substances, radiotherapeutic substances, vitamins, tumor-inhibiting drugs, and tissue affecting materials desired to be delivered to the antigenic site.
15. A treatment agent as provided in claim 14 wherein the water-soluble polymer is selected from the group consisting of polyacrylic acid, polymethacrylic acid, polyacrylamide, polyvinyl alcohol, polyallylalcohol, polymer combinations of the foregoing, hydroxyethyl cellulose, hydroxypropyl cellulose, natural water-soluble polymers and synthetic water-soluble polymers.
16. A treatment agent as provided in claim 15 wherein the antibody is bound to the polymer by bonding means selected from the group consisting of activated groups on the antibody, activated coupling reagents, activated groups on the polymer, activatable groups on the polymer, covalent bonds, and biotin-avidin complex.
17. An immunological substance detecting reagent as provided in claims 8, 9 or 10 wherein the attaching means is a complex of biotin-avidin and the marker substance is a fluorescent dye.
CA000413624A 1981-10-19 1982-10-18 Immunological reagents employing polymeric backbone possessing reactive functional groups Expired CA1200760A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US313,019 1981-10-19
US06/313,019 US4434150A (en) 1981-10-19 1981-10-19 Immunological reagents employing polymeric backbone possessing reactive functional groups

Publications (1)

Publication Number Publication Date
CA1200760A true CA1200760A (en) 1986-02-18

Family

ID=23214037

Family Applications (1)

Application Number Title Priority Date Filing Date
CA000413624A Expired CA1200760A (en) 1981-10-19 1982-10-18 Immunological reagents employing polymeric backbone possessing reactive functional groups

Country Status (5)

Country Link
US (1) US4434150A (en)
EP (1) EP0077671B1 (en)
JP (2) JPS5879162A (en)
CA (1) CA1200760A (en)
DE (1) DE3271843D1 (en)

Families Citing this family (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5258041A (en) * 1982-09-29 1993-11-02 Bio-Metric Systems, Inc. Method of biomolecule attachment to hydrophobic surfaces
US4722906A (en) * 1982-09-29 1988-02-02 Bio-Metric Systems, Inc. Binding reagents and methods
US4973493A (en) * 1982-09-29 1990-11-27 Bio-Metric Systems, Inc. Method of improving the biocompatibility of solid surfaces
US5217492A (en) * 1982-09-29 1993-06-08 Bio-Metric Systems, Inc. Biomolecule attachment to hydrophobic surfaces
US5512329A (en) * 1982-09-29 1996-04-30 Bsi Corporation Substrate surface preparation
US4663277A (en) * 1983-05-20 1987-05-05 Profile Diagnostic Sciences Inc. Virus detection method and materials
US4687732A (en) * 1983-06-10 1987-08-18 Yale University Visualization polymers and their application to diagnostic medicine
US4582810A (en) * 1983-09-30 1986-04-15 Becton, Dickinson And Company Immuno-agglutination particle suspensions
EP0155224A3 (en) * 1984-03-15 1987-05-06 CHROMAGENICS, Inc. Solid-borne complex bearing chromagen responsive functionality for antibody, antigen, receptor, or ligand detection
US4797363A (en) * 1984-03-19 1989-01-10 Board Of Trustees, University Of Illinois Bacteriophages as recognition and identification agents
EP0164889A3 (en) * 1984-05-07 1986-12-17 HSC Research Development Corporation Surface-treated polymeric substrates
GB8423228D0 (en) * 1984-09-14 1984-10-17 Unilever Plc Specific binding materials
FI850481A0 (en) * 1985-02-06 1985-02-06 Labsystems Oy FOERFARANDE FOER BESTAEMNING AV MOTMEDEL ELLER ANTIGENER.
US4735907A (en) * 1985-03-18 1988-04-05 Eastman Kodak Company Stabilized fluorescent rare earth labels and labeled physiologically reactive species
US4824659A (en) * 1985-06-07 1989-04-25 Immunomedics, Inc. Antibody conjugates
US4863875A (en) * 1985-08-26 1989-09-05 Gia Research Company, L.P. Dye labelled antibodies as reagents for use in immunoassay systems
US4806311A (en) * 1985-08-28 1989-02-21 Miles Inc. Multizone analytical element having labeled reagent concentration zone
DK112986D0 (en) * 1986-03-11 1986-03-11 Svenska Sockerfabriks Ab PROCEDURE FOR THE DETECTION OF COMPONENTS IN A TEST
US5263992A (en) * 1986-10-17 1993-11-23 Bio-Metric Systems, Inc. Biocompatible device with covalently bonded biocompatible agent
SE454781B (en) * 1986-10-17 1988-05-30 Wallac Oy HYBRIDIZATION PROCEDURE FOR THE DETECTION OF POLYNUCLEOTIDE SEQUENCE
JPS63131064A (en) * 1986-11-13 1988-06-03 プロフイル ダイアグノステイツク サイエンシス インコ−ポレイテツド Method of detecting protein and virus
US5206178A (en) * 1986-11-19 1993-04-27 Genetic Systems Corporation Membrane affinity concentration immunoassay
US4975532A (en) * 1986-11-28 1990-12-04 Sclavo, Inc. Method to derivatize dextran
US4959306A (en) * 1986-11-28 1990-09-25 Sclavo, Inc. Labeling design for a binding assay reagent
JPS6447952A (en) * 1987-05-06 1989-02-22 Saibaafuruua Inc Immunoassay, reagent used therefor and making thereof
US5024935A (en) * 1987-12-18 1991-06-18 Eastman Kodak Company Dye-providing composition, diagnostic test kit and their use in method for ligand determination using peroxidase labeled-receptor
US4968742A (en) * 1987-11-09 1990-11-06 Miles Inc. Preparation of ligand-polymer conjugate having a controlled number of introduced ligands
CA1312277C (en) * 1987-12-18 1993-01-05 Richard C. Sutton Avidin- and biotin-immobilized reagents, analytical elements and methods of use
US4813973A (en) * 1988-01-11 1989-03-21 The University Of Toronto Innovations Foundation Modified, water-soluble polymers containing amide groups reacted with xanthydrol and other dyes
AU609241B2 (en) * 1988-01-29 1991-04-26 Abbott Laboratories Ion-capture assays and devices
AU2684488A (en) * 1988-06-27 1990-01-04 Carter-Wallace, Inc. Test device and method for colored particle immunoassay
CA1308022C (en) * 1988-08-11 1992-09-29 Eleftherios P. Diamandis Multiple fluorescence labelling with europium chelators
US5053054A (en) * 1988-09-12 1991-10-01 Ortho Pharmaceutical Corporation Methods and reagents for staining intracellular components
SE466521B (en) * 1988-10-03 1992-02-24 Kurt G I Nilsson REAGENTS OF ANALYZE CONSISTING OF AN ENZYM AND ANOTHER LIGAND, WHICH ARE COVALENTLY BONDED TO A WATER INSULATED PARTICLE WITH A DIAMETER OF LESS THAN 500 AA
EP0520979B1 (en) * 1988-10-17 1999-01-13 Hemasure, Inc. Process for the covalent surface modification of hydrophobic polymers and articles made therefrom
FR2638462B1 (en) * 1988-10-28 1992-05-22 Oris Ind THREE-DIMENSIONAL COMPLEXES BASED ON STREPTAVIDINE (OR AVIDINE) AND POLYBIOTINYL LUMINESCENT COMPOUNDS
DE3840605A1 (en) * 1988-12-02 1990-06-07 Behringwerke Ag MEDIUM FOR IMMUNCHEMICAL TESTS CONTAINING CARBOXYL GROUP POLYMERS
US6010867A (en) * 1989-04-19 2000-01-04 Ibiden Co., Ltd. Reagent for biomaterials assay, preparation method thereof, and assay method
JP2619549B2 (en) * 1989-06-29 1997-06-11 日本商事株式会社 Antigen quantification method and solid phase used for it
GB8926407D0 (en) * 1989-11-22 1990-01-10 Gen Electric Plc Polymers
US5019521A (en) * 1990-03-12 1991-05-28 Photest Diagnostics, Inc. Paired polypeptides
JP2626654B2 (en) * 1990-03-31 1997-07-02 科学技術振興事業団 Targeting high molecular weight pharmaceutical compounds and intermediates thereof
US6120805A (en) * 1990-04-06 2000-09-19 Rhone-Poulenc Rorer Sa Microspheres, process for their preparation and their use
US5410016A (en) * 1990-10-15 1995-04-25 Board Of Regents, The University Of Texas System Photopolymerizable biodegradable hydrogels as tissue contacting materials and controlled-release carriers
US5462990A (en) * 1990-10-15 1995-10-31 Board Of Regents, The University Of Texas System Multifunctional organic polymers
US5380536A (en) * 1990-10-15 1995-01-10 The Board Of Regents, The University Of Texas System Biocompatible microcapsules
US5626863A (en) * 1992-02-28 1997-05-06 Board Of Regents, The University Of Texas System Photopolymerizable biodegradable hydrogels as tissue contacting materials and controlled-release carriers
GB9028038D0 (en) * 1990-12-24 1991-02-13 Nycomed Pharma As Test method and reagent kit therefor
DK130991D0 (en) * 1991-07-04 1991-07-04 Immunodex K S POLYMER CONJUGATES
DE4140142A1 (en) * 1991-12-05 1993-06-09 Boehringer Mannheim Gmbh, 6800 Mannheim, De MULTIVALENT DEXTRANREAGENT FOR USE IN PRECIPITATION TESTS
US5217743A (en) * 1992-02-07 1993-06-08 Paradigm Biotechnologies Partnership Biomaterials of enhanced biocompatibility
EP1958646A1 (en) * 1992-02-11 2008-08-20 Henry M. Jackson Foundation For The Advancement Of Military Medicine Dual carrier immunogenic construct
US5573934A (en) * 1992-04-20 1996-11-12 Board Of Regents, The University Of Texas System Gels for encapsulation of biological materials
WO1993017669A1 (en) * 1992-02-28 1993-09-16 Board Of Regents, The University Of Texas System Photopolymerizable biodegradable hydrogels as tissue contacting materials and controlled-release carriers
EP0660933A1 (en) * 1992-09-18 1995-07-05 Gec-Marconi Limited Separation method
GB9326277D0 (en) * 1993-12-23 1994-02-23 Marconi Gec Ltd Labelling
US5750357A (en) * 1994-05-18 1998-05-12 Microquest Diagnostics, Inc. Method of rapid analyte detection
GB0029154D0 (en) 2000-11-30 2001-01-17 Lee Helen Signal enhancement with multiple labelled-antibodies
DE10064827A1 (en) * 2000-12-22 2002-06-27 Dade Behring Marburg Gmbh Sandwich assay for detecting analyte, useful e.g. for hormones, with detection or correction of the hook effect by measuring detection signals twice
US6887377B2 (en) * 2001-02-01 2005-05-03 Sigma-Aldrich Co. Affinity matrices with enhanced visibility for molecular pull-down and immunoprecipitation applications
WO2004106890A2 (en) * 2003-05-22 2004-12-09 Ciphergen Biosystems, Inc. Trace incorporation of fluorescent monomer facilitating quality control of polymerization reactions
US20050118727A1 (en) * 2003-12-01 2005-06-02 Carsten Schelp Conjugates and their use in detection methods
CA2547353A1 (en) * 2003-12-01 2005-07-07 Dade Behring Marburg Gmbh Conjugates, and use thereof in detection methods
JP2007512543A (en) * 2003-12-01 2007-05-17 デイド・ベーリング・マルブルク・ゲゼルシヤフト・ミツト・ベシユレンクテル・ハフツング Homogeneous detection method
CN1925871A (en) * 2004-01-27 2007-03-07 南加州大学 Polymer-bound antibody concer therapeutic agent
EP1717584B1 (en) * 2005-04-25 2013-03-20 Fujirebio Inc. Polymeric fluorescent label
JP5034302B2 (en) * 2005-04-25 2012-09-26 富士レビオ株式会社 Fluorescent label
JP4848988B2 (en) * 2007-03-26 2011-12-28 セイコーエプソン株式会社 Detection kit
WO2010101931A2 (en) 2009-03-03 2010-09-10 Access Medical System Co., Ltd. Detection system and method for high sensitivity fluorescent assays
EP2504702A4 (en) * 2009-11-24 2014-12-17 Argos Inc Devices for detection of analytes
US9733242B2 (en) * 2012-10-07 2017-08-15 Sevident, Inc. Devices for capturing analyte
US9910040B2 (en) * 2012-07-09 2018-03-06 Sevident, Inc. Molecular nets comprising capture agents and linking agents
JP6010110B2 (en) 2011-04-20 2016-10-19 アクセス メディカル システムズ,リミティド Repeated amplification of light-emitting polymers.
US10641768B2 (en) * 2011-07-08 2020-05-05 Abbott Japan Co. Ltd. Methods and kits for decreasing interferences from leukocytes in specific binding assays
JP5071575B2 (en) * 2011-07-29 2012-11-14 セイコーエプソン株式会社 Detection kit
US9568431B2 (en) 2012-04-16 2017-02-14 Access Medical Systems, Ltd. Luminescent immunoassays for quantitating analytes having a wide concentration range
KR20220025946A (en) 2014-03-21 2022-03-03 애브비 인코포레이티드 Anti-egfr antibodies and antibody drug conjugates
CN109641962A (en) 2016-06-08 2019-04-16 艾伯维公司 Anti- B7-H3 antibody and antibody drug conjugates
BR112018075630A2 (en) 2016-06-08 2019-03-19 Abbvie Inc. anti-cd98 antibodies and antibody drug conjugates
JP2019526529A (en) 2016-06-08 2019-09-19 アッヴィ・インコーポレイテッド Anti-B7-H3 antibody and antibody drug conjugate
WO2017214335A1 (en) 2016-06-08 2017-12-14 Abbvie Inc. Anti-b7-h3 antibodies and antibody drug conjugates
JP6894747B2 (en) * 2017-04-19 2021-06-30 キヤノン株式会社 Polymer
CA3064697A1 (en) 2017-04-19 2018-10-25 Bluefin Biomedicine, Inc. Anti-vtcn1 antibodies and antibody drug conjugates
US11154919B2 (en) 2017-07-17 2021-10-26 Titanium Metals Corporation Planetary reform roller and method of reforming a vessel cavity
CN112433048A (en) * 2020-11-17 2021-03-02 深圳上泰生物工程有限公司 Kit for chemiluminescence immunoassay, and preparation method and application thereof
UY39610A (en) 2021-01-20 2022-08-31 Abbvie Inc ANTI-EGFR ANTIBODY-DRUG CONJUGATES

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3853987A (en) * 1971-09-01 1974-12-10 W Dreyer Immunological reagent and radioimmuno assay
GB1470074A (en) * 1973-07-30 1977-04-14 Block Engineering Radiant energy emitting analytical reagent
GB1533410A (en) * 1974-12-20 1978-11-22 Block Engineering Antigen detecting reagents
DK143689C (en) * 1975-03-20 1982-03-15 J Kreuter PROCEDURE FOR THE PREPARATION OF AN ADVERTISED VACCINE
AU528246B2 (en) * 1978-10-14 1983-04-21 Teikoku Hormone Mfg. Co., Ltd. Stable immunoassay reagent for hapten determination
JPS596389B2 (en) * 1978-10-14 1984-02-10 帝国臓器製薬株式会社 Stable immunochemical measurement reagent and measurement method for hapten
CA1121345A (en) * 1979-03-05 1982-04-06 Robert A. Yoshida Method for competitive protein binding assays inhibiting non-specific interference
JPS5718727A (en) * 1980-07-07 1982-01-30 Teijin Ltd Reactive polymer and its preparation

Also Published As

Publication number Publication date
JPH03236331A (en) 1991-10-22
EP0077671A1 (en) 1983-04-27
US4434150A (en) 1984-02-28
JPS5879162A (en) 1983-05-12
DE3271843D1 (en) 1986-07-31
JPH0324627B2 (en) 1991-04-03
EP0077671B1 (en) 1986-06-25

Similar Documents

Publication Publication Date Title
CA1200760A (en) Immunological reagents employing polymeric backbone possessing reactive functional groups
EP0195624B1 (en) Fluorescent labels and labeled species and their use in analytical elements and determinations
US3966897A (en) Medium for use in bioassay and method of using same
JP3075427B2 (en) Use of 1- (1-pyrrolidinylcarbonyl) pyridinium salt to bind compound to carboxylated particles
CA1289877C (en) Water-insoluble particle and immunoreactive reagent, analytical elementsand methods of use
US4735907A (en) Stabilized fluorescent rare earth labels and labeled physiologically reactive species
EP0462644B1 (en) Biologically active reagents prepared from carboxy-containing polymer, analytical element and methods of use
EP0496473B1 (en) Method of preparing biologically active reagents from succinimide-containing polymers, analytical element and methods of use
JPS60226900A (en) Nucleic acid-protein composite body
US5397695A (en) Attachment of compounds to polymeric particles using carbamoylonium compounds and a kit containing same
EP1221052A1 (en) Conjugated polymer tag complexes
JP2579972B2 (en) Membrane affinity concentrated immunoassay
GB2123146A (en) Dual parameter flow immunoassays
EP0308235B1 (en) Attachment of compounds to polymeric particles using carbamoylonium compounds
AU649397B2 (en) Immunoreactant carriers having a novel biocompatible intermediate coating and process of making same
US6010867A (en) Reagent for biomaterials assay, preparation method thereof, and assay method
EP0152254A2 (en) Chromogenic support immunoassay utilizing labeled complement components
EP0136347A1 (en) Insoluble surfaces treated to inhibit non-specific protein binding
EP0411711B1 (en) Attachment of compounds to polymeric particles using dication ethers and a kit containing same
JPH06160387A (en) Measuring method for antigen-antibody reaction
US5635605A (en) Method for detecting the presence of ring shaped particle tumor marker
Singer The use of polystyrene latexes in medicine
EP0537826B1 (en) Diagnostic test kit and specific binding assay using a peroxidase label and a modulator of signal
JPH07151756A (en) Method for measuring antibody
JPS6148764A (en) Method and kit for testing aqueous sample for specific substance

Legal Events

Date Code Title Description
MKEX Expiry