WO1988005537A1 - Conjugate of biological molecules and a chelating agent - Google Patents

Conjugate of biological molecules and a chelating agent Download PDF

Info

Publication number
WO1988005537A1
WO1988005537A1 PCT/FI1987/000163 FI8700163W WO8805537A1 WO 1988005537 A1 WO1988005537 A1 WO 1988005537A1 FI 8700163 W FI8700163 W FI 8700163W WO 8805537 A1 WO8805537 A1 WO 8805537A1
Authority
WO
WIPO (PCT)
Prior art keywords
conjugate
antibody
dtpa
fragment
conjugate according
Prior art date
Application number
PCT/FI1987/000163
Other languages
French (fr)
Inventor
Pirkko Vihko
Marja SÖDERVALL
Reijo Vihko
Original Assignee
Innofarm Oy
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 Innofarm Oy filed Critical Innofarm Oy
Publication of WO1988005537A1 publication Critical patent/WO1988005537A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/531Production of immunochemical test materials
    • G01N33/532Production of labelled immunochemicals
    • G01N33/533Production of labelled immunochemicals with fluorescent label
    • 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
    • G01N33/573Immunoassay; Biospecific binding assay; Materials therefor for enzymes or isoenzymes

Definitions

  • the present invention concerns conjugates of biological molecules labeled with a fluorescent agent, especially labeled antibody conjugates.
  • Radioimmunoassays are the most generally used assay methods. Research has been carried out in order to find substitutes for the radioactive labels due to their detrimental effect on health and also to their often short storage time.
  • One alternative is a fluorescent label.
  • Some of the lanthanide ions, specifically Eu 3+ and Tb 3+ form strongly fluorescing chelates with suitable organic, multiteeth ligands, e.g. ethylenediaminetetraacetic acid (EDTA) and diethylenetriaminepentaacetic acid (DTPA) .
  • EDTA ethylenediaminetetraacetic acid
  • DTPA diethylenetriaminepentaacetic acid
  • WO 84/03698 Eu 3+ cheiating DTPA derivatives have been described, which can be coupled to antibodies for in vitro diagnostic purposes.
  • the cheiating compounds for example N 1 - and N 2 -(p- isothiocyanatobenzyl)diethylenetriaminetetraacetic acid, are synthesized by means of a laborious four stage process, wherein use is made of reagents which from the point of view of safety of the working environment are irritating and toxic to the working environment (bromoacetic acid and thiophosgene).
  • reagents which from the point of view of safety of the working environment are irritating and toxic to the working environment (bromoacetic acid and thiophosgene).
  • a mixture of isomers is formed.
  • the isomers are separated by means of HPLC and the synthesis sequence is continued using the pure isomers.
  • Eu3 + is chelated in the third stage of the synthesis series (I. Hemmilä et al., Anal. Biochem. 197 (1984) 335), also in a multistage process.
  • the Eu 3+ -chelate of the DTTA isocyanate derivative is reacted with the antibody.
  • the unbound chelate thus has to be removed by gel filtration. At this ratio the conjugate contains 7 Eu 3+ /lgG.
  • the claims of the US patent 4,454,106 comprise coupling of DTPA to an antibody using the process of mixed anhydrides and the labeling of the conjugate obtained i.a. with a fluorescent metal, e.g. Eu 3+ . in the method according to the patent appr.
  • 1.5 DTPA/IgG is obtained.
  • This binding ratio has been obtained by testing with a 1 11 in-label.
  • the patent does not, however, contain an example describing the use of Eu 3+ as a label.
  • the literature contains no example of Eu 3+ -labeling with DTPA anhydrides, and thus the labeling cannot be considered to have been carried out with any prior described method.
  • the TR-FIA sensitivity is not sufficient enough with the ratio 1.5 Eu 3+ /igG, but the withinassay and betweenassay variations are large.
  • DTPA has been coupled to proteins using also the cyclic anhydride of DTPA (cDTPAA) (I) of the formula
  • cDTPAA has several advantages. It is a substance that is easily synthesized in a single step. It is also available commercially (Sigma, Aldrich). It is an unam- biguous, under dry conditions stable substance as opposed to the mixed anhydride (US-patent 4,454,106), which always has to be prepared shortly before use. The re- producibility of the process is because of the unstability poor, and there is no reliable analytical method for the anhydride formed.
  • cDTPAA is poorly soluble substance. Hnatowich has used for the administration of small amounts of cDTPAA a chloroform suspension. With this method, administration from an unhomogenous mixture is not very precise.
  • the DTPA-conjugate of the antibody is prepared with the mono- or bicyclic anhydride of DTPA and the conjugate obtained is labeled for diagnostic use, for example with Eu 3+ (TR-FIA).
  • a DTPA-conjugate is formed from the protein with the mono- or bicyclic anhydride.
  • the bicyclic an- hydride is either a commercially available product or prepared according to Eckelman (Eckelman et al., J. Pharm. Sci. 64 (1975) 704).
  • the monocyclic anhydride is prepared according to Halpern (Radioimmunoimaging and Radioimmunotherapy, ed .
  • the mono- or bicyclic anhydride is dissolved in dry DMSO, as opposed to Hnatowich, whereby due to complete dissolution, the pipetting accuracy is good. From this homogenous solution a suitable amount is pipetted into a bicarbonate solution of the antibody. The reaction time is 1 hour at room temperature.
  • the antibody DTPA-derivative thus obtained may be used for different diagnostic purposes in vitro and in vivo.
  • an unsymmetrical conjugate (II) is formed containing four cheiating acid groups, and having a structure corresponding to that of the N'-benzyl-DTTA-conjugate (III). Only the group of DTTA and the bond connecting to the protein are different. ( III )
  • the antibody DTPA-derivative prepared with a cyclic anhydride is comparable to the N-benzyl-DTTA- derivative for use in TR-FIA. It is easier and much faster to work with the cyclic anhydride.
  • the commercial DTPA-anhydride or the DTPA-anhydride prepared in one simple synthesis step is reacted with the antibody for 1 hour at room temperature.
  • Excess DTPA is removed by dialyzing while simultaneously exchanging the buffer to one suitable for labeling.
  • the cheiating substance is added in the form of the commercially easily available chloride. There is no need to use an excess of EuCl 3 , but a tested suitable amount, wherefore no gel filtration is needed.
  • the reagent disclosed in the patent publication WO 84/03698 is laboriously prepared in four synthesis steps. In the synthesis two isomers are formed of which only one forms a rapidly dissociating Eu- chelate. From this follows that the yield of usable chelate after the separation of the isomers is poor. In addition, as the degree of conjugation is low, the consumption of a laboriously prepared reagent is high.
  • the same antibody DTPA-conjugate may be used for different diagnostic purposes, both in vitro and in vivo, by choosing a suitable metal for each purpose.
  • the purified F(ab') 2 -fragment of PAP Prostatic Acid Phosphatase
  • purified whole antibody as well as SHBG (Sex Hormone Binding Globulin) purified monoclonal antibody
  • the bicyclic DTPA is manufactured by Sigma.
  • the mono-cDTPA we have manufactured our.
  • the dimethylsulphoxide (DMSO) is manufactured by SNEA or Aldrich.
  • Purified monoclonal PAP-antibody is used as such or is split with pepsin to F(ab') 2 , which is purified using protein-A-Sepharose-4B-affinity chromatography.
  • the F(ab') 2 -fraction obtained (0.02 M PBS buffer, pH 8.0) or the whole monoclonal antibody (IgG) is concentrated in an Amicon concentration device and dialyzed into a NaHCO 3 -buffer, pH 8.0. The dialyzed sample is lyo- philized and deep frozen.
  • F(ab') 2 is dissolved in water shortly before use so that the protein concentration is 10 mg/ml and the concentration of NaHCO 3 is 0.1 M.
  • the SHBG antibody is used as whole (IgG).
  • cDTPAA is dissolved in dry DMSO, from which a desired amount is pipetted into the protein solution.
  • the solution is mixed for 30 s to 1 min and incubated at room temperature for one hour.
  • the pH of the solution is checked.
  • the sample is dialyzed into a 0.1 M Na-citrate buffer, pH 5, to remove free DTPA.
  • cDTPA/F(ab') 2 /IgG 5 the cDTPA- DMSO-solution (17.85 mg/ml), 5 /ul, is pipetted into the protein solution, wherein 5 mg F(ab')2 or IgG/0.5 ml.
  • the labeling was carried out using 10 M Eu with respect to the antibody-DTPA-conjugate.
  • labeling is carried out with the Eu-DTPA-derivative chelate as described in the patent WO 84/03698, it is used in an excess of 160 M with respect to the antibody.
  • EuCl 3 (Fluka 46130) was dissolved in 0.1 M sodium citrate buffer, pH 5 to 6, 2.75 mg EuCl 3 /mI, 15 minutes before use.
  • the antibody DTPA-conjugate was prepared using a ratio of DTPA/IgG of 20:1.
  • EuCl 3 (Fluka 46130) was dissolved in 0.1 M sodium citrate buffer at pH 6. Labeling was carried out using Eu at a ratio of approximately 10 M with respect to the IgG-DTPA-derivative at room temperature for one hour.
  • the SHBG-antibody was labeled with the Eu-chelate described in the patent specification WO 84/03698. Thereby the Eu-chelate was used in an excess of 160 M with respect to the antibody.
  • the remaining labeling conditions were: 50 mM K 2 HPO 4 , pH 9.4, incubation over night at +4°C. Separation of the labeled antibody was carried out using gel filtration in order to remove excess chelate.
  • reaction mixture was poured into a gel filtration column (Sepharose 6B 1.5 cm x 30 cm, at the top Sephadex 6-25 1.5 cm x 5 cm). Elution was carried out using 0.05 M tris-HCl, 0.9% NaCl, 0.1% NaN 3 (pH 7.75, 25°C) as elution buffer.
  • TR-FIA of SHBG functioned in the same way with both labels and their distinction with respect to the different standards was as good.
  • the invention also concerns a method for the determination of the degree of conjugation between a cheiating agent and a biological molecule.
  • the degree of conjugation is meant the molar ratio cheiating agent/biological molecule.
  • the degree of conjugation is usually determined by coupling to the cheiating groups in the conjugate a radioactive isotope. This mode of determining is, however, cumbersome and unprecise due to the short half-life of the isotopes.
  • Eu is coupled to the cheiating groups and the fluorescence is measured, the degree of conjugation may be determined very precisely and in a simple manner.

Abstract

The conjugate of a biological molecule, especially a protein, such as a monoclonal antibody or its fragment and of mono- or bicyclic DTPA, which conjugate is labeled with a fluorescent agent. For the preparation of the labeled conjugate the protein and the chelating agent are first combined, whereafter the conjugate is labeled with the fluorescent agent. The invention also concerns a method for the determination of the degree of conjugation of conjugates.

Description

Conjugate of biological molecules and a cheiating agent.
The present invention concerns conjugates of biological molecules labeled with a fluorescent agent, especially labeled antibody conjugates.
Immunological methods have become standard methods in clinical chemistry, especially for the determination of biologically active compounds in low concentrations. Radioimmunoassays are the most generally used assay methods. Research has been carried out in order to find substitutes for the radioactive labels due to their detrimental effect on health and also to their often short storage time. One alternative is a fluorescent label. Some of the lanthanide ions, specifically Eu3+ and Tb3+, form strongly fluorescing chelates with suitable organic, multiteeth ligands, e.g. ethylenediaminetetraacetic acid (EDTA) and diethylenetriaminepentaacetic acid (DTPA) .
In the patent publication WO 84/03698 Eu3+ cheiating DTPA derivatives have been described, which can be coupled to antibodies for in vitro diagnostic purposes. The cheiating compounds, for example N1- and N2-(p- isothiocyanatobenzyl)diethylenetriaminetetraacetic acid, are synthesized by means of a laborious four stage process, wherein use is made of reagents which from the point of view of safety of the working environment are irritating and toxic to the working environment (bromoacetic acid and thiophosgene). In the second stage of the synthesis a mixture of isomers is formed. The isomers are separated by means of HPLC and the synthesis sequence is continued using the pure isomers. Eu3+ is chelated in the third stage of the synthesis series (I. Hemmilä et al., Anal. Biochem. 197 (1984) 335), also in a multistage process. The Eu3+-chelate of the DTTA isocyanate derivative is reacted with the antibody. The reaction is slow (overnight, 4°C) and the degree of conjugation low, about 10%, wherefore the reagent has to be used in a large excess (chelate:antibody = 60:1). The unbound chelate thus has to be removed by gel filtration. At this ratio the conjugate contains 7 Eu3+/lgG.
The claims of the US patent 4,454,106 comprise coupling of DTPA to an antibody using the process of mixed anhydrides and the labeling of the conjugate obtained i.a. with a fluorescent metal, e.g. Eu3+. in the method according to the patent appr. 1.5 DTPA/IgG is obtained. This binding ratio has been obtained by testing with a 111in-label. The patent does not, however, contain an example describing the use of Eu3+ as a label. Also the literature contains no example of Eu3+-labeling with DTPA anhydrides, and thus the labeling cannot be considered to have been carried out with any prior described method. For example the TR-FIA sensitivity is not sufficient enough with the ratio 1.5 Eu3+/igG, but the withinassay and betweenassay variations are large.
DTPA has been coupled to proteins using also the cyclic anhydride of DTPA (cDTPAA) (I) of the formula
CD
Figure imgf000004_0001
(e.g. Hnatowich, US 4,479,930 and Hnatowich et al.. Int. J. Appl. Radiat. Isot. 33 (1982) 327). The use of cDTPAA has several advantages. It is a substance that is easily synthesized in a single step. It is also available commercially (Sigma, Aldrich). It is an unam- biguous, under dry conditions stable substance as opposed to the mixed anhydride (US-patent 4,454,106), which always has to be prepared shortly before use. The re- producibility of the process is because of the unstability poor, and there is no reliable analytical method for the anhydride formed.
On the other hand, cDTPAA is poorly soluble substance. Hnatowich has used for the administration of small amounts of cDTPAA a chloroform suspension. With this method, administration from an unhomogenous mixture is not very precise.
According to the present invention the DTPA-conjugate of the antibody is prepared with the mono- or bicyclic anhydride of DTPA and the conjugate obtained is labeled for diagnostic use, for example with Eu3+ (TR-FIA).
In time-resolved fluoroimmunoassay use is made of the difference between the duration of the fluorescence of a specific signal and that of a non-specific background, whereby the signal-noise ratio is improved. Almost in all published methods use is made of Eu3+- labeled antibody, wherefrom the bound Eu3+ is determined quantitatively subsequent to dissociative fluorescence development. Wallac-LKB has patented DEL- FIA (dissociative Eu fluorescence enhancement FIA) (EP 64484 (1984)).
This invention simplifies the method described in the patent publication WO 84/03698, according to which the Eu3+-DTTA-chelate obtained in several synthesis stages is coupled to the antibody using a reaction time of 12 hours at +4°C. According to the present invention, initially a DTPA-conjugate is formed from the protein with the mono- or bicyclic anhydride. The bicyclic an- hydride is either a commercially available product or prepared according to Eckelman (Eckelman et al., J. Pharm. Sci. 64 (1975) 704). The monocyclic anhydride is prepared according to Halpern (Radioimmunoimaging and Radioimmunotherapy, ed . S W Burchiel & B A Rhodes, Elsevier 1983, p. 198-9). Typically when preparing the derivative, the mono- or bicyclic anhydride is dissolved in dry DMSO, as opposed to Hnatowich, whereby due to complete dissolution, the pipetting accuracy is good. From this homogenous solution a suitable amount is pipetted into a bicarbonate solution of the antibody. The reaction time is 1 hour at room temperature. The antibody DTPA-derivative thus obtained may be used for different diagnostic purposes in vitro and in vivo.
In TR-FIA, the Eu3+-chelate has to be sufficiently stable but easily dissociated in acid environment. The more teeth the complex former has, the more stable a complex it forms. Also the dissociation from such complexes is slow. It has, however, been discovered (I Hemmila, Lanthanides as Probes for Time-resolved Fluorometric Immunoassays, Thesis 1986) that from the unsymmetrical N'-benzyl-DTTA containing four cheiating acid groups, Eu3+ dissociates faster (1 min) than from the corresponding symmetrical N2-benzyl-DTTA (20 min).
When using a cyclic anhydride of DTPA for the preparation of the antibody DTPA-derivative, an unsymmetrical conjugate (II) is formed containing four cheiating acid groups, and having a structure corresponding to that of the N'-benzyl-DTTA-conjugate (III). Only the group of DTTA and the bond connecting to the protein are different. ( III )
< ID
Figure imgf000007_0001
Thus the antibody DTPA-derivative prepared with a cyclic anhydride is comparable to the N-benzyl-DTTA- derivative for use in TR-FIA. It is easier and much faster to work with the cyclic anhydride. The commercial DTPA-anhydride or the DTPA-anhydride prepared in one simple synthesis step is reacted with the antibody for 1 hour at room temperature.
Excess DTPA is removed by dialyzing while simultaneously exchanging the buffer to one suitable for labeling. The cheiating substance is added in the form of the commercially easily available chloride. There is no need to use an excess of EuCl3, but a tested suitable amount, wherefore no gel filtration is needed. On the other hand, the reagent disclosed in the patent publication WO 84/03698 is laboriously prepared in four synthesis steps. In the synthesis two isomers are formed of which only one forms a rapidly dissociating Eu- chelate. From this follows that the yield of usable chelate after the separation of the isomers is poor. In addition, as the degree of conjugation is low, the consumption of a laboriously prepared reagent is high. Also, as the cheiating metal according to the present invention is added in the last stage, the same antibody DTPA-conjugate may be used for different diagnostic purposes, both in vitro and in vivo, by choosing a suitable metal for each purpose.
For the preparation of the derivatives, the purified F(ab')2-fragment of PAP (Prostatic Acid Phosphatase) monoclonal antibody and purified whole antibody as well as SHBG (Sex Hormone Binding Globulin) purified monoclonal antibody was used. The bicyclic DTPA is manufactured by Sigma. The mono-cDTPA we have manufactured ourselves. The dimethylsulphoxide (DMSO) is manufactured by SNEA or Aldrich.
Example 1
Coupling of DTPA to a monoclonal antibody
1) Treatment of the protein
Purified monoclonal PAP-antibody is used as such or is split with pepsin to F(ab')2, which is purified using protein-A-Sepharose-4B-affinity chromatography. The F(ab')2-fraction obtained (0.02 M PBS buffer, pH 8.0) or the whole monoclonal antibody (IgG) is concentrated in an Amicon concentration device and dialyzed into a NaHCO3-buffer, pH 8.0. The dialyzed sample is lyo- philized and deep frozen. F(ab')2 is dissolved in water shortly before use so that the protein concentration is 10 mg/ml and the concentration of NaHCO3 is 0.1 M. The SHBG antibody is used as whole (IgG).
2) Preparation of the derivative
A. cDTPA/F(ab')2-derivative/IgG-derivative
For the conjugation reaction cDTPAA is dissolved in dry DMSO, from which a desired amount is pipetted into the protein solution. The solution is mixed for 30 s to 1 min and incubated at room temperature for one hour. The pH of the solution is checked. The sample is dialyzed into a 0.1 M Na-citrate buffer, pH 5, to remove free DTPA. For example when cDTPA/F(ab')2/IgG = 5 the cDTPA- DMSO-solution (17.85 mg/ml), 5 /ul, is pipetted into the protein solution, wherein 5 mg F(ab')2 or IgG/0.5 ml.
When adding DMSO to the protein solution one has to take into account that the amount of DMSO does not exceed 5% of the volume of the protein solution.
B. mono cDTPA/F(ab')2-derivative/IgG-derivative
Preparation in the same way as for the cyclic derivative. Mono-cDTPAA is added in a DMSO-solution to the protein solution.
Example 2
Labeling of the DTPA-conjugate of the PAP-antibody or its F(ab')2-fragment with EuCl3
The labeling was carried out using 10 M Eu with respect to the antibody-DTPA-conjugate. When labeling is carried out with the Eu-DTPA-derivative chelate as described in the patent WO 84/03698, it is used in an excess of 160 M with respect to the antibody.
EuCl3 (Fluka 46130) was dissolved in 0.1 M sodium citrate buffer, pH 5 to 6, 2.75 mg EuCl3/mI, 15 minutes before use. The DPTA-conjugate of the antibody or its fragment dissolved in 0.1 M sodium citrate buffer (10 mg conjugate/ml, pH 5 to 6) was mixed with a suitable amount of EuCl3 - sodium citrate solution and incubated for one hour at room temperature. The percentage of labeling is 98. Example 3
Labeling of the DTPA-conjugate of a SHBG-antibody with EuCl3
The antibody DTPA-conjugate was prepared using a ratio of DTPA/IgG of 20:1. EuCl3 (Fluka 46130) was dissolved in 0.1 M sodium citrate buffer at pH 6. Labeling was carried out using Eu at a ratio of approximately 10 M with respect to the IgG-DTPA-derivative at room temperature for one hour.
Example 4
For reason of comparison the SHBG-antibody was labeled with the Eu-chelate described in the patent specification WO 84/03698. Thereby the Eu-chelate was used in an excess of 160 M with respect to the antibody. The remaining labeling conditions were: 50 mM K2HPO4, pH 9.4, incubation over night at +4°C. Separation of the labeled antibody was carried out using gel filtration in order to remove excess chelate.
After incubation the reaction mixture was poured into a gel filtration column (Sepharose 6B 1.5 cm x 30 cm, at the top Sephadex 6-25 1.5 cm x 5 cm). Elution was carried out using 0.05 M tris-HCl, 0.9% NaCl, 0.1% NaN3 (pH 7.75, 25°C) as elution buffer.
The fluorescence was measured by adding a sample of 10/ul into 200/ul Delfia Enhancement-solution. After shaking the fluorescence was measured with an Arcus fluorimeter. The Eu-concentration was calculated using as the reference value 0.2 pmol Eu = 106 cps. Comparison results:
Using the EuCl3-label, the following numbers of Eu- molecules attached to antibodies were obtained:
Eu-molecules/protein molecules cDTPA/F(ab')2 (5:1) 1.9-2.0 cDTPA/F(ab')2 (10:1) 2.7 cDTPA/IgG (10:1) 2.4 cDTPA/IgG (20:1) 3.7 (Ex. 3) cDTPA/IgG (50:1) 6.9 m-cDTPA/IgG (300:1) 11.0
In the Example of the patent publication W0 84/03698 when labeling using an excess of 60 M of Eu-chelate 7 Eu-molecules/IgG is obtained and when raising the ratio to 160- 500 M excess Eu-chelate the ratio does not exceed 10 Eu-molecules/IgG. With the ratio 160 M of Eu- chelate in excess, 9.3 Eu-molecules per one antibody molecule were obtained (Example 4).
The labels according to the Examples 3 and 4 were tested with the SHBG Delfia kit-method using approximately the same protein concentration in the dilutions of both labels. This means in brief that the wells of the micro- titer plates were coated with polyclonal anti-SHBG- antibodies. The comparison solutions and the labels were pipetted into the wells and incubated, washed and measured using Delfia Enhancement-solution. SHBG-comparison solns EuCl3-label Eu-chelate label nmol/1 (Example 3) cps (Example 4) cps
0 3197 4281
6.25 43535 81940
12.5 77255 147983
25 146026 263334
50 238831 442793
100 389177 841566
200 787518 1579440
TR-FIA of SHBG functioned in the same way with both labels and their distinction with respect to the different standards was as good.
According to one embodiment the invention also concerns a method for the determination of the degree of conjugation between a cheiating agent and a biological molecule. By the degree of conjugation is meant the molar ratio cheiating agent/biological molecule. The degree of conjugation is usually determined by coupling to the cheiating groups in the conjugate a radioactive isotope. This mode of determining is, however, cumbersome and unprecise due to the short half-life of the isotopes. When, however, as in this invention, Eu is coupled to the cheiating groups and the fluorescence is measured, the degree of conjugation may be determined very precisely and in a simple manner.

Claims

Claims:
1. Conjugate of a biological molecule and a cheiating agent and labeled with a fluorescent agent, c h a r a c t e r i z e d in that the cheiating agent is mono- or bicyclic DTPA.
2. Conjugate according to the Claim 1, c h a r a c t e r i z e d in that the biological molecule is a poly- or monoclonal antibody or a fragment thereof, a pepti- de, nucleic acid, hapten, enzyme, synthetic probe or cDNA.
3. Conjugate according to the Claim 1, c h a r a c t e r i z e d in that the biological molecule is a monoclonal antibody or a fragment thereof.
4. Conjugate according to the Claim 3, c h a r a c t e r i z e d in that the monoclonal antibody or its fragment is the antibody of SHBG-antigen or a fragment thereof.
5. Conjugate according to the Claim 3, c h a r a c t e r i z e d in that the monoclonal antibody or its fragment is the antibody of PAP-antigen or a fragment thereof.
6. Conjugate according to the Claims 1-5, c h a r a c t e r i z e d in that the fluorescent agent is Eu.
7. Process for the preparation of a conjugate according to the Claims 1-6, c h a r a c t e r i z e d in that the biological molecule and the cheiating agent are first combined, whereafter the labeling of the conjugate is carried out with the fluorescent agent.
8. In vitro-diagnostic immunological method, c h a r a ct e r i z e d in that a conjugate according to the claims 1-6 is used for determining the antigen of an analyte.
9. Method for the determination of the degree of conjugation between a cheiating agent and a biological molecule, c h a r a c t e r i z e d in that the chelate mixed in a solution buffered to pH 5-6 and a EuCl3- buffer solution are incubated, whereafter the fluorescence is determined in a manner known.
PCT/FI1987/000163 1987-01-14 1987-12-08 Conjugate of biological molecules and a chelating agent WO1988005537A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
FI870122 1987-01-14
FI870122A FI80801C (en) 1987-01-14 1987-01-14 KJJUGAT AV EN MONOCLONAL ANTIKROPP OCH ANVAENDNING. ELLER ETT FRAGMENT DAERAV OCH ETT KELATERANDE AEMNE, DESS FRAMSTAELLNING

Publications (1)

Publication Number Publication Date
WO1988005537A1 true WO1988005537A1 (en) 1988-07-28

Family

ID=8523765

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/FI1987/000163 WO1988005537A1 (en) 1987-01-14 1987-12-08 Conjugate of biological molecules and a chelating agent

Country Status (2)

Country Link
FI (1) FI80801C (en)
WO (1) WO1988005537A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0709100A1 (en) * 1994-09-30 1996-05-01 Nihon Medi-Physics Co., Ltd. Contrast agent, containing several detection groups per molecule
US5792444A (en) * 1989-05-09 1998-08-11 The General Hospital Corporation Labeled chemotactic peptides to image focal sites of infection or inflammation

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE898708A (en) * 1983-01-21 1984-05-16 Schering Ag DIAGNOSTIC MEDICINE
US4479930A (en) * 1982-07-26 1984-10-30 Trustees Of The University Of Massachusetts Amines coupled wth dicyclic dianhydrides capable of being radiolabeled product
DD229595A1 (en) * 1984-12-14 1985-11-13 Adw Ddr METHOD FOR THE PRODUCTION OF CHELATE FIXED BIOMOLECULARS
EP0186947A1 (en) * 1984-11-01 1986-07-09 Nycomed As Paramagnetic contrast agents for use in "in vivo" diagnostic methods using NMR, and their preparation
US4647447A (en) * 1981-07-24 1987-03-03 Schering Aktiengesellschaft Diagnostic media
US4668503A (en) * 1982-07-26 1987-05-26 Trustees Of University Of Massachusetts Process for labeling amines with 99m Tc
WO1987007955A1 (en) * 1986-06-17 1987-12-30 Baxter Travenol Laboratories, Inc. Homogeneous fluoroassay methods employing fluorescent background rejection and water-soluble rare earth metal chelate fluorophores

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4647447A (en) * 1981-07-24 1987-03-03 Schering Aktiengesellschaft Diagnostic media
US4479930A (en) * 1982-07-26 1984-10-30 Trustees Of The University Of Massachusetts Amines coupled wth dicyclic dianhydrides capable of being radiolabeled product
US4668503A (en) * 1982-07-26 1987-05-26 Trustees Of University Of Massachusetts Process for labeling amines with 99m Tc
BE898708A (en) * 1983-01-21 1984-05-16 Schering Ag DIAGNOSTIC MEDICINE
EP0186947A1 (en) * 1984-11-01 1986-07-09 Nycomed As Paramagnetic contrast agents for use in "in vivo" diagnostic methods using NMR, and their preparation
DD229595A1 (en) * 1984-12-14 1985-11-13 Adw Ddr METHOD FOR THE PRODUCTION OF CHELATE FIXED BIOMOLECULARS
WO1987007955A1 (en) * 1986-06-17 1987-12-30 Baxter Travenol Laboratories, Inc. Homogeneous fluoroassay methods employing fluorescent background rejection and water-soluble rare earth metal chelate fluorophores

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, Vol. 101 (1984), Abstract No. 101:216407q; & BE,A,898708. *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5792444A (en) * 1989-05-09 1998-08-11 The General Hospital Corporation Labeled chemotactic peptides to image focal sites of infection or inflammation
EP0709100A1 (en) * 1994-09-30 1996-05-01 Nihon Medi-Physics Co., Ltd. Contrast agent, containing several detection groups per molecule

Also Published As

Publication number Publication date
FI80801C (en) 1990-07-10
FI870122A0 (en) 1987-01-14
FI80801B (en) 1990-03-30
FI870122A (en) 1988-07-15

Similar Documents

Publication Publication Date Title
US4808541A (en) Determination method utilizing reagents covalently labelled with essentially non-fluorescent lanthanide chelates in combination with time-resolved fluorescence spectroscopy and the reagents to be used in the method
Mukkala et al. The synthesis and use of activated N-benzyl derivatives of diethylenetriaminetetraacetic acids: alternative reagents for labeling of antibodies with metal ions
US4279992A (en) Specific binding assay employing an enzyme-cleavable substrate as label
JP5133066B2 (en) An immunoassay for measuring the release of neurotensin into the circulating fluid.
CA1213579A (en) Digoxigenin immunogens, antibodies, labeled conjugates, and related derivatives
US4404366A (en) Beta-galactosyl-umbelliferone-labeled hapten conjugates
US4331590A (en) β-Galactosyl-umbelliferone-labeled protein and polypeptide conjugates
JPS60252684A (en) Fluorescent assembly
EP0493745A1 (en) Fluorescent compound, complex, reagent, and specific binding assay employing said reagent
JPH0137693B2 (en)
JPH08504751A (en) Biotinylated chemiluminescent label, conjugate, assay and assay kit
JPH07502045A (en) Novel biotinylation reagent
US4261974A (en) Valproic acid immunogen conjugates and antibodies thereto
Xu et al. Novel solution-phase immunoassays for molecular analysis of tumor markers
US4478934A (en) Determination of adenosine by immunoassay involving acylation of the adenosine
US4292425A (en) βGalactosyl-umbelliferone valproic acid conjugates
EP0104926A2 (en) Assay processes and materials therefor
CN102183635A (en) In vitro detection kit for TAFI content
CN102103142B (en) In-vitro assay method for thrombin-activatable fibrinolysis inhibitor (TAFI) content
JPH0717672B2 (en) Cobalamin-acid hydrazide, its production method and cobalamin conjugate
US6190923B1 (en) Diethylenetriamine-N,N′,N″-triacetic acid derivatives
WO1988005537A1 (en) Conjugate of biological molecules and a chelating agent
Gonzalez et al. Direct solid-phase time-resolved fluoroimmunoassay of 17 alpha-hydroxyprogesterone in serum and dried blood spots on filter paper
JP3360174B2 (en) A homobifunctional reagent that binds enzymes, etc., to antibodies, etc.
JPS61250558A (en) Immunological assaying method

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): JP US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE FR GB IT LU NL SE