WO1996033408A1 - On-line coupling of biochemical detection to continuous-flow separation techniques - Google Patents

On-line coupling of biochemical detection to continuous-flow separation techniques Download PDF

Info

Publication number
WO1996033408A1
WO1996033408A1 PCT/EP1995/001523 EP9501523W WO9633408A1 WO 1996033408 A1 WO1996033408 A1 WO 1996033408A1 EP 9501523 W EP9501523 W EP 9501523W WO 9633408 A1 WO9633408 A1 WO 9633408A1
Authority
WO
WIPO (PCT)
Prior art keywords
affinity molecule
detection
effluent
ligand
binding
Prior art date
Application number
PCT/EP1995/001523
Other languages
French (fr)
Inventor
Jan Van Der Greef
Hubertus Irth
Erik Van Der Vlics
Aaike Oosterkamp
Johan Haggblad
Original Assignee
Rijksuniversiteit Leiden
Karo Bio Ab
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 Rijksuniversiteit Leiden, Karo Bio Ab filed Critical Rijksuniversiteit Leiden
Priority to AU24475/95A priority Critical patent/AU2447595A/en
Priority to PCT/EP1995/001523 priority patent/WO1996033408A1/en
Publication of WO1996033408A1 publication Critical patent/WO1996033408A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N30/84Preparation of the fraction to be distributed
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N30/84Preparation of the fraction to be distributed
    • G01N2030/8429Preparation of the fraction to be distributed adding modificating material
    • G01N2030/8435Preparation of the fraction to be distributed adding modificating material for chemical reaction
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography
    • G01N30/88Integrated analysis systems specially adapted therefor, not covered by a single one of the groups G01N30/04 - G01N30/86
    • G01N2030/8809Integrated analysis systems specially adapted therefor, not covered by a single one of the groups G01N30/04 - G01N30/86 analysis specially adapted for the sample
    • G01N2030/8813Integrated analysis systems specially adapted therefor, not covered by a single one of the groups G01N30/04 - G01N30/86 analysis specially adapted for the sample biological materials

Abstract

The present invention relates to an on-line detection method comprising the on-line coupling of a fractionation and a biochemical detection technique, which method comprises the addition of a controlled amount of an affinity molecule to the effluent of the fractionation step to react with analytes in the effluent, the subsequent addition of a controlled amount of a detectable ligand capable of binding to the affinity molecule, and detection of the affinity molecule/detectable ligand complex. Further, the invention is directed to an on-line method for the screening of compounds on their binding capability to a known affinity molecule, which method comprises a fractionation step providing an effluent, the addition of a controlled amount of said affinity molecule to the effluent of the fractionation step and effecting a contact time sufficient to allow a reaction with or interaction between the compounds in the effluent, the subsequent addition of a controlled amount of a detectable ligand capable of binding to the affinity molecule, and detection of the affinity molecule/detectable ligand complex.

Description

ON-LINE COUPLING OF BIOCHEMICAL DETECTION TO CONTINUOUS-FLOW SEPARATION TECHNIQUES
The present invention relates to the on-line coupling of biochemical detection to continuous-flow separation techniques. Preferably, this on-line detection method is used as a screening method.
It is well-known to the person skilled in the art that immunoassays are highly sensitive detection techniques which combine the selectivity of biospecific interactions between antibodies and antigens with the sensitive detection of labels used a reporter molecules.
However, it is well known that immunoassays suffer from the problem of cross- reactivity in which reactions of antibodies with more than one analyte lead to erroneous results. For this reason, immunoassays are frequently combined with a fractionation step, e.g., a separation step using HPLC or another type of liquid chromatography.
In the field to which the present invention relates, there is a need for on-line coupling of a fractionation step and an immunoassay detection system. Several approaches have been proposed and described for performing continuous-flow immunoassays. Most of these immunoassays are in the form of a post-column reaction detection system based on a sequential additional type immunoassay.
Cassidy et al., Anal. Chem. 64 (1992), 1973-1977 disclose a kinetically controlled immunoassay based on the sequential addition of antibody, sample and label on a protein A column, performing immunoassays for albumin and transferrin in less than 1 minute.
Nilson et al. , J. Chromatography, 597 (1992) 383-389 describe a continuous-flow
competitive assay involving horse-radish peroxidase-labelled antibodies. The system
described was coupled to a size-exclusion column to allow the monitoring of enzyme
activity.
These prior art detection techniques, which are of a sequential nature, are not suitable
for on-line coupling to liquid chromatographic or other fractionation systems since these
techniques do not allow the continuous monitoring of the fractionation effluent.
In the article of Irth et al. in the Journal of Chromatography 633 (1992) 65072 and in
the articles of Oosterkamp et al. in the Journal of Chromatography 633 (1993) 65-72 and
in the Journal of Chromatography 653 (1994) 55-61, a method for the on-line detection
of digoxin and its metabolites is described. The on-line detection process comprises the
direct injection of a sample containing digoxin and its metabolites, a liquid
chromatographic (LC) fractional separation step, the mixing of the effluent of the LC column with fluorescein-labelled antibodies against digoxigenin, one of the metabolites,
the removal of free labelled antibodies from the mixture via passage through a small column packed with an antigen-bound support, and detection of the strongly fluorescent
irnmunocomplexes.
The digoxigenin system described is based on association reactions of antibodies and antigens eluting from the analytical column. By the use of fluorescein-labelled antibodies detection limits in the nanomolar-range are obtained.
The immunoreagent in this prior art assay consists of fluorescein-labelled fragments of
anti-digoxigenin antibodies which were immimopurified and are commercially available.
The commercial availability of purified, labelled antibodies is however exceptional. In
almost all cases, antibodies are only available in unlabelled state in crudely purified
antiserum. Although labelling and purification schemes for antibodies (or other affinity
proteins) are known to the person skilled in the art, it is preferred to use antisera, which
may be commercially available, without any pretreatment.
Furthermore, it is difficult to selectively label only those antibodies in an antibody
preparation which are reactive with the analyte. In general, labelling techniques make
use of primary amine groups of antibodies resulting in the colabelling of all other
proteins present in the preparation. This leads to a drastic increase in the background
signal and, consequently, to an increase in detection limits.
The aim of the invention is to provide alternatives for the known technique, which uses
fluorescein-labelled antibodies. The present invention does not have the disadvantages
associated with the labelling of antibodies.
In more detail, the problem underlying the present invention is the provision of an on¬
line detection technique without requiring the use of labelled antibodies. This problem is overcome by the provision of an on-line detection comprising the on¬ line coupling of a fractionation step to a biochemical detection technique in which the
detection method comprises the addition of a controlled amount of an affinity molecule to an effluent of the fractionation step whereby the affinity molecules bind analytes in
the effluent and the subsequent addition of a controlled amount of a detectable ligand
capable of binding to the affinity molecule, and detection of the affinity
molecule/detectable ligand complex whereby reduction in the amount of detected affinity molecule/detectable ligand complex indicates the presence of an affinity molecule-
binding compound in the effluent.
In particular, in a first step, a suitable affinity molecule for analytes to be detected -
which affinity molecule may e.g be an affinity protein, such as an antibody or avidin - is
added to the effluent of a liquid chromatography or a capillary electrophoresis column to
react with ligands or analytes eluting from the column. Unbound affinity molecules
react in a second step with an excess of a detectable ligand, which may be the analyte to
be detected in labelled from, such as a fluorescein-labelled analyte to titrate the
remaining free binding sites; or as an alternative, a competition reaction occurs between
the affinity molecule, the analyte in the effluent and the detectable ligand. Normally, the
labelled ligand/affinity molecule complex is detected after a separation of free and bound
labelled ligand, preferably on the basis of the difference in molecular weight. Finally,
the labelled ligand/affinity molecule complex is detected.
The combination of the fractionation step with the biochemical detection step in accordance with the present invention greatly enhances the performance of both
techniques. The combined techniques provide an analytical method which is characterised by a high selectivity and a high sensitivity. Further, the problem
associated with cross-reactivity does no longer occur when using the method of the
present invention.
The method of the present invention uses bioaffinity molecules such as antibodies,
receptors, etc to detect any compounds showing high affinity for the ligand binding site of said affinity molecule. The compounds to be detected may be biological compounds
but are not in any way restricted thereto.
The method of the invention makes it possible to screen a mixture of different
compounds on their ability to bind to a certain affinity molecule, e.g., in order to find an inhibitor for said affinity molecule. Therefore, the present invention further relates to a
fast on-line method for the screening of compounds on their binding capability to a
known affinity molecule, which method comprises a fractionation strep providing an
effluent, the addition of a controlled amount of said affinity molecule to the effluent of the fractionation step and effecting a contact time sufficient to allow a reaction with or
an interaction between the compounds in the effluent, the subsequent addition of a
controlled amount of a detectable ligand capable of binding to the affinity molecule, and
detection of the affinity molecule/detectable ligand complex.
A comparison of the detection signal obtained when analytes are injected with the signal obtained when only the controlled amounts of affinity molecule and detectable ligand are introduced in the continuous-flow system, provides information in respect of the
percentage of the binding sites occupied by analytes present in the effluent of the
fractionation step. The person skilled in the art possesses the knowledge to process and
evaluate the data obtained from the detection method. The percentage binding sites
occupied by an analyte present in the effluent can be used to find new compounds which
show an interaction with a known affinity molecule. This information provides the
possibility to implement the on-line separation/affinity molecule detection process of the
present invention in e.g drug discovery.
A system of high-throughput screening to be used in drug discovery including the
method of the invention may, for instance, consist of the following steps. Complex
samples generated for example by an upstream combinational chemistry system, are
prefractionated in fractions containing compounds of similar polarity using, for example,
a solid-phase extraction technique or electrophoretic sample handling principles. Each
fraction may additionally be separated using, for example, either analytical or
preparative-scale liquid chromatographic separation columns. The compounds eluting
from said LC column are on-line detected using a suitable affinity molecule detection
technique. Where preparative-scale separation columns are applied, a post-column flow-
split will be made. One of the two flow streams is subjected to detection using the affinity molecule detection technique of the invention, the other stream is directed to a
fraction collector. Dependent on the signal obtained from the affinity molecule detector,
fractions containing compounds causing a positive response will be collected while fractions causing a negative response will be discarded. This complete screening method can be automated using valve-switching processes.
A suitable fractionation method to be used in the method of the present invention
comprises a liquid chromatography separation or a capillary electrophoresis step. Other
separation or fractionation techniques which are known to the person skilled in the art
and which allow a relatively continuous output stream can, however, be used as well.
In a preferred embodiment, the liquid chromatography separation step is a reversed
phase HPLC step.
The detection method which forms an integral part of the methods of the present
invention can be chosen dependent on a suitable detectable ligand. Examples of suitable
detection methods comprise methods based on all types of luminescence detection
principles, such as fluorescence, time-delayed fluorescence and chemiluminescence
spectroscopy; methods based on electrochemical detection and on radiometric detection.
In the methods of the present invention all molecules capable of interaction with or
binding to other molecules can be used as affinity molecule. In preferred embodiments,
the affinity molecule is selected from the group of cytosolic receptors, e.g. , the
estrogen, glucocorticoid receptors; solubilized membrane bound receptor, e.g. , a β-
receptor; affinity proteins, such as antibodies, enzymes, avidin; polynucleotides and
polysaccharides. Furthermore, the affinity molecule may be an "orphan receptor". Such molecules are thought to be nuclear receptors, as they are structurally related to characterised
receptors, but for which no ligand has been identified. The determination of ligands for orphan receptors is relatively slow. By using the method of the present invention, this determination can be accelerated.
The term "detectable ligand" as used in the present description and claims refers to a
ligand which is capable of interacting or reacting with the afore-defmed affinity
molecule, and which can be detected in a biochemical detection method. An example of
a detectable ligand is a labelled analyte.
If labelled antigens are used instead of the labelled antibodies used in the prior art
techniques described above, two major advantages are obtained. First, it is easier to
prepare and, particularly, purify low-molecular weight labelled antigens. Second, the
background noise signal problem obtained in the preparation step of labelled antibodies
does not occur when labelled antigens are used for immunodetection.
On-line coupling as used in the methods of the present invention requires fast reaction
times in order to minimize extra-column band broadening. This means that affinity
molecule-ligand interactions having reaction times in the order of minutes rather than
hours should be considered.
In one embodiment of the method of the invention, biotinylated DNA-fragments are determined by the continuous addition of avidin to the eluate of a HPLC column. The
avidin is allowed to react with the biotinylated DNA-fragments in a so-called reaction
coil. Subsequently, a continuous flow of a solution comprising fluorescein-labelled biotin is added in the flow leaving the reaction coil. In a following step in the
continuous flow process, free and avidin-bound fluorescein-labelled biotin are separated
from each other, e.g., by the use of a separation procedure making use of the difference
in molecular weight between free and bound biotin, such as passing a column packed
with Clg-silica restricted access support. The high-molecular weight labelled
ligan/protein complex passes the C18-column unretained and is subsequently detected by
means of fluorescence detection. For the biotin-avidin system a detection limit in the
order of 100-500 fmol was obtained for biotin.
The methods of the present invention can be applied as well to systems wherein a DNA
or RNA probe is used as a receptor for DNA and/or RNA derived analytes. In this
case, the detectable ligand may be at least part of complementary strand which can be,
e.g., labelled with a radioactive isotope.
In a very advantageous embodiment within the scope of the present invention the affinity
molecule used is the estrogen receptor and the detectable ligand is coumestrol or another
natively fluorescent estrogenic ligand. In this embodiment it is also possible to use the
steroid binding domain of the human estrogen receptor, which can, e.g., be prepared by
gene technology, instead of the complete estrogen receptor. This embodiment does not require a separation step wherein bound and free coumestrol are separated. Coumestrol O 96/33408 PC17EP95/01523
is a compound which provides a strong fluorescent signal when bound to the estrogen receptor, and provides a weak fluorescent signal in free state. Since coumestrol and the
estrogen receptor are introduced in the continuous-flow system of the present invention
in controlled amounts, a comparison of the fluorescence signals obtained will give
information as to how many binding sites of the estrogen receptor are occupied by
coumestrol and how many binding sites are occupied by competitive analytes. Although
this embodiment using the steroid binding domain of the estrogen receptor and
coumestrol or another natively fluorescent estrogenic ligand does not require a
separation step wherein bound and free coumestrol are separated, the fluorescence data
will provide more detailed qualitative and quantitative information if such a separation
step is carried out.
Methods in accordance with the present invention will be described in further detail by
way of example only with reference to the following Examples.
Example 1
Biotin was applied to a liquid chromatography (LC) column. The LC separation was
carried out on a 100 x 3.0 mm internal diameter stainless-steel column packed with
Nucleosil C18 (5μm particles, Macherey-Nagel, Dϋren, Germany) using methanol/aqueous triethylammonium acetate (10 mmol/1; pH 7.0) 19:90 (v/v) as a
mobile phase, which mobile phase was delivered by means of a Kratos-ABI (Ransey,
U.S.A.) Spectroflow 400 pump. In the effluent of the LC separation step, avidin was delivered in binding buffer consisting of sodium phosphate (10 mmol/1, pH 8.0; Merck, Darmstadt, Germany) containing 0.5 mol/1 sodium chloride (analytical grade; Merck,
Darmstadt, Germany), the avidin concentration being 34 nmol/1, by means of a
Pharmacia (Uppsala, Sweden) P3500 pump (flow rate 0.4 ml/min). The combined
avidin-biotin flow was introduced into a knitted 0.5 mm interanl diameter poly -
tetrafluoroefhylene reaction coil with an internal volume of 800 μl. Subsequently,
fluorescein-labelled biotin (Sigma, St. Louis, U.S. A) was delivered in the mobile phase
at a flow rate of 0.8 ml/min using a Kratos-ABI Spectroflow 400 pump. The flow
obtained was introduced into a knitted 0.5 mm internal diameter polytetrafluoroethylene
reaction coil with an internal volume of 400 μl. The reactions were performed at 20°C.
Separation of free and avidin-bound fluorescein-biotin was performed using a 10 x 40
mm interanl diameter column packed with Crg alkanediol silica (described in DE-A-41
30 475). After this column, the flow was introduced into a fluorescence detector (Merck
1080 fluorescence detector; excitation wavelength 486 ran; emission wavelength 520
nm).
The absolute detection limit obtained for biotin was 160 fmol. The detection system
provided a linear signal ranging from 8 to 200 nmol/1 (20 μl injections).
Example 2 A mixture consisting of T6-biotin (0.5 μM) and T3-biotin-T3 (0.5 μM) representing
biotinylated DNA-fragments was injected onto an LC column. The LC separation was
carried out on a 100 x 3.0 mm internal diameter stainless-steel column packed with Nucleosil Cg (5 μm particles, Macherey-Nagel, Dϋren, Germany) using
acetonitrile/aqueous triethylammonium acetate (10 mM, pH 7.0) 7:93 (v/v) as a mobile phase, which mobile phase was delivered by means of a Kratos-ABI (Ramsey, U.S. A) Spectroflow 400 pump. In the effluent of the LC separation step, avidin was delivered
in binding buffer consisting of sodium phosphate (10 nmol/1, pH 8.0; Merck,
Darmstadt, Germany) containing 0.5 mol/1 sodium chloride (analytical grade; Merck,
Darmstadt, Germany), the avidin concentration being 34 nmol/1, by means of a
Pharmacia (Uppsala, Sweden) P3500 pump (flow rate 0.4 ml/min). The combined
avidin-biotinylated compounds flow was introduced in a knitted 0.5 mm internal
diameter polytetrafluoroethylene reaction coil with an internal volume of 800 μl.
Subsequently, fluorescein-labelled biotin (Sigma, St. Louis, U.S. A) was delivered in the
mobile phase at a flow rate of 0.8 ml/min using a Kratos-ABI Spectroflow 400 pump.
The flow obtained was introduced into a knitted 0.5 mm internal diameter
polytetrafluoroethylene reaction coil with an internal volume of 400 μl. The reactions
were performed at 20°C.
Separation of free and avidin-bound fluorescein-biotin was performed using a 10 x 4.0
mm internal diameter column packed with C18 alkanediol silica (described in DE-A-41 30 475). After passing this column, the flow was introduced in a fluorescence detector
(Merck 1080 fluorescence detector; excitation wavelength 486 nm; emission wavelength
520 nm).
It appeared that the retention times of T3-biotin-T3 and T6 were 6.2 and 6.6 minutes, respectively.
EXAMPLE 3
A mixture of 17β-estradiol, estriol, diethylstilbestrol, zeranol, estrone, progestrone,
testosterone and dexamethasone (concentration: 100 nmol/1; injection 20 μl) was
applied to a LC column. The LC separation was carried out on a stainless-steel column
packed with Nucleosil C2 (5 μm particles, Macherey-Nagel, Dϋren, Germany) using
acetonitrile:methanol: potassium phosphate buffer (10 mM, pH 7.4) 20:20:60 (v/v) as a
mobile phase, which mobile phase was delivered by means of a Kratos-ABI (Ramsey,
U.S. A) Spectroflow 400 pump at a flow rate of 0.5 ml/min. In the effluent of the LC
separation step, estrogen receptor (human estrogen receptor steroid binding domain;
Karo-Bio, Huddinge, Sweden) was delivered in a potassium phosphate buffer (200
nmol/1, pH 7.4; Merck, Darmstadt, Germany), the estrogen receptor concentration being
5 nmol/1, by means of a Pharmacia (Uppsala, Sweden) P3500 pump (flow rate 0.5
ml/min). The combined estrogen-analytes flow was introduced into a knitted 0.5 mm
internal diameter polytetrafluoroethylene reaction coil with an internal volume of 900 μl.
Subsequently, coumestrol (Eastman-Kodak, Rochester, New York, U.S. A) in a
concentration of 111 nmol/1 in the potassium phosphate buffer was delivered in the
mobile phase at a flow rate of 0.5 ml/min using a Kratosd-ABI Spectroflow 400 pump.
The flow obtained was introduced into a knitted 0.5 mm internal diameter
polytetrafluoroethylene reaction coil with an internal volume of 570 μl. The reactions
were performed at 20°C. Subsequently, the flow was introduced into a fluorescence detector (Merck 1080 fluorescence detector; excitation wavelength 340 nm; emission wavelength 410 nm). It
was found that 17β-estradiol, estriol, diethylstilbestrol, zeranol and estrone could be detected in a concentration of 50 nmol/1, whereas progestrone, testosterone and
dexamethasone did not provide any response at concentrations of 100 nmol/1.

Claims

1. An on-line detection method comprising the on-line coupling of a
fractionation step to a biochemical detection technique in which the detection method
comprises the addition of a controlled amount of an affinity molecule to an effluent of
the fractionation step whereby the affinity molecules bind analytes in the effluent and the
subsequent addition of a controlled amount of a detectable ligand capable of binding to the affinity molecule, and detection of the affinity molecule/detectable ligand complex
whereby reduction in the amount of detected affinity molecule/detectable ligand complex
indicates the presence of an affinity molecule-binding compound in the effluent.
2. An on-line method for the screening of compounds for their binding
capability to a known affinity molecule, which method comprises adding a controlled
amount of said affinity molecule to the effluent of a fractionation step and effecting a
contact time sufficient to allow a reaction with or interaction between the compounds
and the affinity molecule in the effluent, the subsequent addition of a controlled amount
of a detectable ligand capable of binding to the known affinity molecule, and detection of
the affinity molecule/detectable ligand complex whereby reduction in the amount of
detected affinity molecule/detectable ligand complex indicates the presence of an affinity
molecule-binding compound in the effluent.
3. The method of claim 1 or 2, wherein the fractionation step is a liquid
chromatography separation or a capillary electrophoresis step.
4. The method of claim 3, wherein the liquid chromatography separation step is a HPLC or a reversed phase HPLC step.
5. The method of any one of the preceding claims, wherein the detection is
effected by fluorescence, time-delayed fluorescence or chemiluminescence spectroscopy,
or by a method based on electrochemical detection or radiometric detection.
6. The method of any one of the preceding claims, wherein the affinity
molecule is a cytosolic receptor, a solubilized membrane bound receptor, an antibody,
an enzyme, avidin, a polynucleotide or a polysaccharide.
7. The method of any one of the preceding claims, wherein the detectable
ligand is a labelled ligand or a ligand possessing native detection properties such as
fluorescence or electrochemical activity.
8. A method according to any preceding claim in which the affinity molecule is
an estrogen receptor or a portion thereof.
9. The method of claim 8, wherein the affinity molecule is an estrogen receptor
or the steroid binding domain thereof and the detectable ligand is coumestrol or another
natively fluorescent estrogenic ligand.
10. A method according to any preceding claim, in which the method is arranged downstream of a combinatorial chemistry system whereby the combinatorial chemistry system provides the fractionation step providing the effluent.
11. A method according to any preceding claim in which the affinity molecule is
an orphan receptor.
12. Compounds detected by the method according to any preceding claim.
13. The use of compounds detected by their method according to any one of
claims 1-11 as a ligand for the affinity molecule.
PCT/EP1995/001523 1995-04-21 1995-04-21 On-line coupling of biochemical detection to continuous-flow separation techniques WO1996033408A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU24475/95A AU2447595A (en) 1995-04-21 1995-04-21 On-line coupling of biochemical detection to continuous-flow separation techniques
PCT/EP1995/001523 WO1996033408A1 (en) 1995-04-21 1995-04-21 On-line coupling of biochemical detection to continuous-flow separation techniques

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/EP1995/001523 WO1996033408A1 (en) 1995-04-21 1995-04-21 On-line coupling of biochemical detection to continuous-flow separation techniques

Publications (1)

Publication Number Publication Date
WO1996033408A1 true WO1996033408A1 (en) 1996-10-24

Family

ID=8166007

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP1995/001523 WO1996033408A1 (en) 1995-04-21 1995-04-21 On-line coupling of biochemical detection to continuous-flow separation techniques

Country Status (2)

Country Link
AU (1) AU2447595A (en)
WO (1) WO1996033408A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991013354A1 (en) * 1990-02-23 1991-09-05 The Government Of The United States Of America, As Represented By The Secretary Of The Department Of The Navy Flow immunosensor method and apparatus

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991013354A1 (en) * 1990-02-23 1991-09-05 The Government Of The United States Of America, As Represented By The Secretary Of The Department Of The Navy Flow immunosensor method and apparatus

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
A. J. OOSTERKAMP ET AL.: "Bioanalysis of digoxin and its metabolites using direct serum injection combined with liquid chromatography and on-line immunochemical detection.", JOURNAL OF CHROMATOGRAPHY B, vol. 653, pages 55 - 61 *
H. IRTH ET AL.: "On-line immunochemical detection in liquid chromatography using fluorescein-labelled antibodies.", JOURNAL OF CHROMATOGRAPHY, vol. 633, 24 February 1993 (1993-02-24), pages 65 - 72 *
M. NILSSON ET AL.: "Process monitoring by flow-injection immunoassay. Evaluation of a sequential competitive binding assay.", JOURNAL OF CHROMATOGRAPHY, vol. 597, 24 April 1993 (1993-04-24), pages 383 - 389 *
S. A CASSIDY ET AL.: "Kinetic chromatographic sequential addition immunoassays using protein A affinity chromatography", ANALYTICAL CHEMISTRY, vol. 64, pages 1973 - 1977 *

Also Published As

Publication number Publication date
AU2447595A (en) 1996-11-07

Similar Documents

Publication Publication Date Title
Oosterkamp et al. Reversed-phase liquid chromatography coupled on-line to receptor affinity detection based on the human estrogen receptor
AU660480B2 (en) Method and apparatus for detecting trace contaminants
Burnina et al. A cost-effective plate-based sample preparation for antibody N-glycan analysis
Irth et al. Strategies for on-line coupling of immunoassays to high-performance liquid chromatography
Oosterkamp et al. Bioanalysis of digoxin and its metabolites using direct serum injection combined with liquid chromatography and on-line immunochemical detection
Todoroki et al. Bioanalysis of bevacizumab and infliximab by high-temperature reversed-phase liquid chromatography with fluorescence detection after immunoaffinity magnetic purification
US5491096A (en) Antigen detection with affinity chromatography and parallel processing a control
Miller et al. Affinity chromatography with immunochemical detection applied to the analysis of human methionyl granulocyte colony stimulating factor in serum
Law et al. Confirmation of cannabis use by the analysis of Δ9-tetrahydrocannabinol metabolites in blood and urine by combined HPLC and RIA
Lensmeyer et al. Liquid-chromatographic procedure for simultaneous analysis for eight benzodiazepines in serum.
Van Bommel et al. High-performance liquid chromatography coupled to enzyme-amplified biochemical detection for the analysis of hemoglobin after pre-column biotinylation
EP1367133A1 (en) Continuous-flow enzyme assay with mass spectrometry detection
EP1048951A1 (en) Mass spectrometry-based technologies for continuous flow biossays using known ligands
Anguizola et al. Peak decay analysis and biointeraction studies of immunoglobulin binding and dissociation on protein G affinity microcolumns
US6080590A (en) On-line coupling biochemical detection to continuous flow separation techniques
Wang et al. Noncompetitive immunoassays using protein G affinity capillary chromatography and capillary electrophoresis with laser-induced fluorescence detection
Pfaunmiller et al. Use of protein G microcolumns in chromatographic immunoassays: a comparison of competitive binding formats
WO1996033408A1 (en) On-line coupling of biochemical detection to continuous-flow separation techniques
EP1048950A1 (en) Identification of ligands for orphan receptors using on-line coupling of mass spectrometry to continuous-flow separation techniques
EP0821790B1 (en) On-line coupling of biochemical detection to continuous-flow separation techniques
Nakamura et al. Two-cycle liquid-chromatographic quantitation of cortisol in urine.
Tang et al. Coupling immunoassays with chromatographic separation techniques
EP1330648B1 (en) Continuous-flow homogeneous biochemical assays based on mass spectrometric detection of known ligands
Oosterkamp et al. Gradient reversed-phase liquid chromatography coupled on-line to receptor-affinity detection based on the urokinase receptor
Ramm et al. Investigation of native and aggregated therapeutic proteins in human plasma with asymmetrical flow field-flow fractionation and mass spectrometry

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA FI JP KR NO US

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref country code: CA