WO1994023301A1 - Methode diagnostique de dosage de renine/angiotensine i - Google Patents

Methode diagnostique de dosage de renine/angiotensine i Download PDF

Info

Publication number
WO1994023301A1
WO1994023301A1 PCT/US1994/003595 US9403595W WO9423301A1 WO 1994023301 A1 WO1994023301 A1 WO 1994023301A1 US 9403595 W US9403595 W US 9403595W WO 9423301 A1 WO9423301 A1 WO 9423301A1
Authority
WO
WIPO (PCT)
Prior art keywords
component
raa
angiotensin
antibody
fluorescent
Prior art date
Application number
PCT/US1994/003595
Other languages
English (en)
Inventor
R. Tyler Mccabe
Bryan R. Wilson
Christopher A. Rhodes
Original Assignee
Pharmaceutical Discovery Corporation
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 Pharmaceutical Discovery Corporation filed Critical Pharmaceutical Discovery Corporation
Priority to AU65289/94A priority Critical patent/AU6528994A/en
Publication of WO1994023301A1 publication Critical patent/WO1994023301A1/fr

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • G01N33/743Steroid hormones
    • 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

  • This invention relates to a highly sensitive method for assaying components of the renin-angiotensin- aldosterone hormonal axis to aid in the diagnosis and treatment of hypertension, renal disease, adrenal gland disease, and cardiovascular disorders.
  • the renin-angiotensin-aldosterone (“RAA") hormonal axis contributes to the regulation of blood pressure, tissue perfusion, and sodium/potassium balance.
  • the mechanism is initiated by a drop in arteriolar pressure, sodium intake, or blood volume, any of which stimulates the kidney to secrete renin into the blood.
  • renin cleaves the liver-derived plasma protein angiotensinogen to form angiotensin I, which is further cleaved by converting enzymes from the lungs to form angiotensin II.
  • Angiotensin II directly raises blood pressure by arteriolar vasoconstriction and indirectly affects blood pressure and volume by causing sodium retention in the kidney and secretion of aldosterone from the adrenal cortex. Aldosterone then raises blood pressure and volume through action on the kidney distal tubule resulting in sodium retention coupled with potassium excretion.
  • Elevated levels of the major components of the RAA hormonal axis may lead to cardiovascular disorders such as hypertension and are excellent diagnostic indicators.
  • a health practitioner usually obtains a blood sample from the patient and submits it to an outside, site-licensed testing facility that identifies and measures the quantity of specific components of the blood, such as renin or angiotensin I.
  • Levels of components of the RAA hormonal axis can be measured by radioimmunoassay or bioassay.
  • Assay methods and clinical uses are reviewed in Sealey, J.E., et al . f "Hormone Assays: Renin, Aldosterone, Peripheral Vein, Renal Vein, and Urinary Assays," Hypertension: Pathophvsiology, Diagnosis, and Management, 1443-60, J.H. Laragh and B.M. Brenner, ed. , Raven Press, New York (1990) .
  • a radioimmunoassay uses the competition between radiolabeled and unlabeled substances in an antigen- antibody reaction to determine the concentration of the unlabeled substance, which may be an antibody or a substance against which specific antibodies can be produced.
  • a bioassay determines the activity of a component in a sample by comparing its effects on a live animal, cells or isolated tissue with those of a reference standard. The available assay methods are capable of detecting RAA components in the blood sample by radiometric or spectrophotometric analysis.
  • radioimmunoassays and bioassays are plagued with the following problems, among others, that hinder effective diagnosis relating to disorders involving RAA components: (1) Many radioimmunoassays and bioassays are sensitive only to nanogram levels of peptide, whereas measurement of at least picogram levels of material are necessary for effective, reliable diagnosis. (2) Radioimmunoassay and bioassay systems with high non-specific binding that distorts measured values. (3) Typically, the use of radioactive testing reagents necessitates a site-licensed facility.
  • Radioimmunoassays and bioassays typically employ two antibodies, a primary and secondary antibody, instead of one, which increases incubation time as well as background noise in the diagnostic system.
  • these assays consist of passing a sample through an immobilized phase consisting of labelled antibody bound to immobilized antigen (for example, passing plasma through a column containing labelled antibody to bound angiotensin I conjugated onto activated CH SepharoseTM 4B) , separating the immobilized phase from the biological sample (plasma) , washing and stripping the labelled antibody from the immobilized phase, and determining how much label was bound prior to passage of the sample versus after passage of the sample.
  • This type of displacement assay may also be useful for construction of standards.
  • a simple, rapid, sensitive, highly selective assay for measuring RAA components in blood and screening individuals in the health practitioner's facility would result in quick, accurate diagnosis. It would also be useful to be able to measure more than one RAA component at a time to determine ratios of one to another for diagnosis of various disorders.
  • an object of the present invention to provide a simple, rapid, sensitive method for measuring levels of RAA components in biological samples.
  • a method and assay for the rapid, sensitive, accurate measurement of a component of the renin- angiotensin-aldosterone ("RAA") hormonal axis in a biological sample using spectrofluorometric measurement of a fluorescent labeled antibody to an RAA component, or fluorescent labeled RAA component, is disclosed.
  • RAA renin- angiotensin-aldosterone
  • a known quantity of a fluorescent labelled antibody immunoreactive with a specific RAA component is incubated with known volumes of the RAA component conjugated with an inert substrate and a biological sample.
  • the amounts of bound and unbound fluorescent antibody are determined by spectrofluorometry, either after or during the incubation.
  • the amount of RAA component in the sample is then quantified after construction of a standard curve based on spectrofluorometric measurements of fluorescent antibody bound to increasing amounts of RAA component in a second incubation mixture.
  • plasma renin activity PRA is measured using fluorescently labeled angiotensin I, which is the RAA component. This is performed after generation of plasma angiotensin I from renin using an enzyme kinetic assay.
  • this method allows measurement of RAA components in picogram amounts in minutes and is useful for diagnosis of hypertension, renal disease, adrenal gland disease, and cardiovascular disorders.
  • Figure 1 is a graph of spectrofluorometric measurement of plasma angiotensin I (ng/ml) versus radioimmunoassay measurement, using the method of Sealey et al., Hypertension: Pathophysiology Diagnosis, and Management, 1990; Sealey, Clin. Che . , (1991), of plasma angiotensin I (ng/ml) .
  • Figure 2 is a graph showing a standard curve of spectrofluorometric (B/B 0 ) /1-(B/B 0 ) versus angiotensin I (ng) , where B represents specific counts per second (CPS) for the known standard quantities of angiotensin I, and B 0 is cps in the absence of angiotensin I.
  • B represents specific counts per second (CPS) for the known standard quantities of angiotensin I
  • CPS specific counts per second
  • the method and spectrofluorometric assay described below can be used to measure in minutes, with accuracy and sensitivity, a component of the renin-angiotensin- aldosterone ("RAA") hormonal axis in a biological sample, including, for example, whole blood, serum, plasma or urine, using a fluorescent label bound to an antibody immunoreactive with the specific RAA component to be measured or an RAA component, and spectrofluorometric technology.
  • RAA renin-angiotensin- aldosterone
  • the method uses disposable columns that contain immobilized angiotensin I along with active sites. Samples of 0.1, 0.25, 0.5, 0.75 and 1 ml of plasma are passed through a rack of disposable columns. Columns are washed with a blocking agent to inactivate unused sites for angiotensin I. A constant concentration of fluorescent antibody is passed over the columns. The eluent is collected and the fluorescent content measured. A ten to one hundred fold increase in the amount of plasma assayed compensates for not carrying out the time consuming step of angiotensin I generation. In order to calibrate the activity of the fluorescent antibody the column matrix has a fixed amount of angiotensin I pre-attached.
  • the column is standardized by passing buffer and blocking agent over the column, followed by the assay volume of fluorescent antibody to be used for the standard.
  • fluorescent labelled RAA component is introduced into a well, which contains a plasma sample, of a 96 well plate that has been coated with polyclonal antibody raised against angiotensin I. The plates are then evaluated for their free labelled RAA component.
  • a radioactive method described in Clin. Chem. 37/10(B) 1811-1819 (1991) is modified to use fluorescent labeled antibody, and results in sensitive, quantitative measurement of the RAA component in the absence of radioactivity.
  • the amount of angiotensin I is evaluated by placing 10 ⁇ l of sample in an assay tube followed by 200 ⁇ l of labelled angiotensin I. Labelled angiotensin I is added to empty tubes for the measurement of nonspecific binding. 20 ⁇ l of antibody is added to all tubes. The tubes are then incubated for between two and 18 hours. Two ml of polyethyleneglycol reagent is added to all tubes except the control tube and the tubes are centrifuged. Pellets are redissolved and measured spectrofluorometrically.
  • the RAA components that can be measured in a biological sample include angiotensin I, renin, angiotensin II, angiotensinogen, and aldosterone, or any other components to which antibodies can be raised. Antibodies to most components are commercially available or can be generated using standard immunization procedures.
  • the immunoglobulin fraction (IgG fraction) is isolated from the immunized animal using Fast Protein, Peptide and Polynucleotide Liquid Chromatography (FPLC) , as described below with reference to the purification of polyclonal IgG raised against angiotensin I.
  • FPLC Polynucleotide Liquid Chromatography
  • the RAA component should exhibit specific and reversible binding affinity along with a chemically modifiable group which can bind to the mixture.
  • the RAA components are available from commercial sources including Sigma Chemical Co. (St. Louis, MO), Peninsula (Belmont, CA) , and Bachem/Cal (Torrance, CA) .
  • an RAA component For use as a reagent in the assay, a known amount of an RAA component is provided.
  • the RAA components are found in the blood in amounts for PRA in normal subjects (male) of 2.4 ⁇ 0.2 (female) 2.3 ⁇ 0.2 ng/ml hr "1 ; Prorenin (male) 17.5 ⁇ 1.4 (female) 14.0 ⁇ 1.3 ng/ l hr "1 ; and Renin substrate (male) 1615 ⁇ 40 (female) 1750 ⁇ 60 ng/ml "1 hr "1 , as reported by Sealey et al., 1990.
  • Angiotensin I in the standard assay typically ranges between 0.01 ng and 100 ng ml "1 .
  • Antibodies to the RAA component are selected based on their immunoreactive specificity for the RAA component to be measured.
  • the antibodies are purified from immunoglobulin obtained from commercial sources such as Berkeley Antibody Company, Berkeley, California, using procedures known to one skilled in the art, such as affinity chromatography.
  • Fluorescent labels and methods of conjugation are Fluorescent labels and methods of conjugation.
  • RAA Reactive Component As used herein, the labelled antibody or labelled RAA component is referred to jointly as "RAA Reactive Component" .
  • the RAA reactive component is bound to a fluorescent label.
  • the criteria for selecting an appropriate fluorescent label are that the components of the reaction media (i.e., buffer composition, temperature) do not interfere with the emission properties of the label.
  • Fluorescent labels are selected for various reasons, including the reactivity of the compound, e.g., amine reactive or sulfhydryl reactive; its excitation and emission wavelengths, and other reasons such as the ability to quench the emission upon binding of the compound.
  • fluorescent labels examples include fluorescein, 7-chloro-4-nitrobenzo-oxo-l,3-diazole (“NBD chloride”) , rhodamine, Texas red, dichlorothrazinylamino fluorescein (“DTAF”), tetramethyl rhodamine isothiocyanate (“TRITC”) , isothiocyanate (“FITC”) , other fluorescein and rhodamine derivatives, as well as additional commercially available fluorescent moieties from sources such as Molecular Probes (Eugene, OR) , Research Organics (Cincinnati, OH) , and Aldrich Chemical Co. (Milwaukee, WI) or Pierce Chemical Co. (Rockford, IL) .
  • the preferred label is DTAF for labeling antibodies, based on its label- antibody conjugate stability.
  • the fluorescent label and the antibody are each dissolved in an appropriate buffer and then dialyzed together to produce the conjugate, which is then purified to remove unbound label by procedures known in the art, such as dialysis, gel filtration, or hydroxylamine treatment.
  • the fluorescent RAA component such as angiotensin I
  • sufficient concentrations of the label and RAA component are incubated together at a selected temperature and for an appropriate period of time to allow formation of the conjugate.
  • the reaction is monitored by thin layer chromatography for consumption of starting material.
  • the conjugate in the incubation mixture is then concentrated, for example, by lyophilization to remove water, and the conjugate is purified by procedures known in the art, including thin layer chromatography, HPLC, recrystallization, FPLC, and LC.
  • the ratio of label to RAA reactive components in the labeled conjugate is calculated based on the ratio of label and compound concentrations (expressed in moles/liter) , by procedures known in the art, which are determined by using ultraviolet and visible spectrophotometry.
  • a known amount of the RAA component to be measured is bound to an inert substrate, resulting in solid phase (immobilized) component.
  • the criteria for selecting an appropriate inert substrate are the ability to bind antigen and chemical stability of matrix.
  • suitable inert substrates are activated CH-SepharoseTM 4B and BioradTM immunobead reagent, using the manufacturers' procedures. These inert substrates are available from various commercial sources, for example Pharmacia Fine Chemicals (Piscataway, N.J.).
  • the preferred inert substrate is CH-SepharoseTM 4B, which is suitable for use with all compounds.
  • Binding of the inert substrate to the RAA component to be measured is accomplished by methods known in the art.
  • the inert substrate is exposed to the dissolved protein in a basic environment to couple the protein to the substrate, usually over a period of between two hours and 24 hours.
  • the excess active groups are blocked at the end of the incubation.
  • the matrix is then washed to remove unbound protein.
  • the labelled reactive component, immobilized component and biological sample mixtures are incubated in microcentrifuge tubes or other suitable containers.
  • the control incubation mixture for measurement of background fluorescence contains known amounts of fluorescent labelled RAA reactive components, control biological sample (defined as containing no RAA component) or egg albumin, and buffer and is measured spectrofluorometrically against "blank" incubation mixtures (defined as containing control biological sample and buffer but no fluorescent reactive component) .
  • the excitation and emission wavelengths are determined empirically for a given labelled RAA component preparation. These wavelengths are then used for the measurements.
  • the second control mixture placed in a series of microcentrifuge tubes and used for construction of a standard fluorescence curve, contains known amounts of labelled RAA component, control biological sample, and immobilized component, such as angiotensin I bound to CH- SepharoseTM 4B, in all tubes, and increasing known amounts of free angiotensin I in the series of tubes.
  • the sample incubation mixture contains the same known amounts of labelled reactive component, immobilized component, in the same amount as the control mixture, and a known amount of biological sample, such as blood, plasma, or serum, to be tested.
  • the mixtures are incubated with shaking at an appropriate temperature, such as room temperature (25°C) and for an appropriate time (30-90 minutes) to react the labelled reaction compound, RAA component to be measured and immobilized component.
  • an appropriate temperature such as room temperature (25°C) and for an appropriate time (30-90 minutes) to react the labelled reaction compound, RAA component to be measured and immobilized component.
  • Unbound labelled reactive component is removed in the supernatant after centrifugation of the mixtures. Fluorescence of unbound labelled reactive component is measured spectrofluorometrically. The amount of RAA component in the test sample is calculated from the standard fluorescence curve constructed from the control incubation mixtures. Methods of measuring fluorescence.
  • the excitation and emission spectra of the fluorescent label are measured using commercially available instrumentation, for example, a SPEX Fluorolog Model 1680 0.22-m Double Spectrometer with DM3000 software (SPEX Industries, Edison, N.J.). Quantitation of fluorescent label bound to the selected compound is accomplished by creating a standard line relating fluorescence intensity values to known amounts of RAA component, such as angiotensin I.
  • the renin/angiotensin I spectrofluorometric assay method focuses on measurement of angiotensin I and includes the following steps: (1) Antibody immunoreactive with angiotensin I, i.e., anti-angiotensin I, is purified from commercially available immunoglobulin G and conjugated with a fluorescent label, such as dichlorotriazinylamino fluorescein ("DTAF”) or tetramethyl rhodamine isothiocyanate (“TRITC”) . The amounts of label and antibody in the conjugate are determined by spectrophotometric measurement of visible and ultraviolet absorption.
  • DTAF dichlorotriazinylamino fluorescein
  • TRITC tetramethyl rhodamine isothiocyanate
  • Angiotensin I (Bachem/Cal, Torrence, CA.) is conjugated to an inert substrate, such as activated CH- SepharoseTM 4B.
  • an inert substrate such as activated CH- SepharoseTM 4B.
  • a known amount of the angiotensin I- substrate conjugate is incubated with known amounts of the fluorescent antibody and biological test sample.
  • the reaction mixture is centrifuged to separate the pellet containing fluorescent antibody bound to the angiotensin I in the test sample and to the angiotensin I-substrate conjugate, from the supernatant, containing the unbound fluorescent antibody.
  • the emission intensities of the bound and unbound fluorescent antibody are determined by spectrofluorometric measurement.
  • the centrifugation step can be omitted and the incubation reaction monitored directly in the spectrofluorometer.
  • This alternative step is possible when the fluorescent antibody has the ability to change spectral properties or emission intensity (spectral shift or quench, DNBC, and NBD, respectively) on complexing with the RAA component, allowing not only measurement of the amount of the antibody-RAA component complex, but also measurement of the kinetic rate of formation in real time, which obviates separation of bound from unbound fluorescent antibody using a centrifugation step. The result is a more rapid and convenient assay.
  • the amount of angiotensin I in the test sample is then quantified, based on comparison of the spectrofluorometric values to a standard straight line correlation graph constructed from a spectrofluorometric measurements of fluorescent anti-angiotensin I bound to increasing amounts of angiotensin I in a series of control samples.
  • Fluorescence measurement is used to confirm that the attachment of the fluorescent label to the antibody did not alter the antibody/antigen binding affinity, as described by (McCabe et al. , FASEB J.. 4:2934-2940 (1990), the teachings of which are incorporated herein as representative of methods now known to those skilled in the art.
  • RAA component such as angiotensin I peptide
  • a fluorescent probe such as 7-chloro-4-nitrobenzo-2-oxa- 1,3-diazole (“NBD chloride")
  • NBD chloride 7-chloro-4-nitrobenzo-2-oxa- 1,3-diazole
  • an inert substrate such as activated CH- SepharoseTM 4B
  • the principles and end result would be the same, with only the components of the assay system differing slightly.
  • Fluorescently labelled angiotensin I would be displaced by unlabelled angiotensin I found in the plasma. The greater the amount of angiotensin I in the plasma, the lower the value of the measured fluorescent intensity.
  • Example l Preparation of anti-angiotensin I labeled with DTAF and TRITC for measurement of angiotensin I in biological samples.
  • the IgG fraction of polyclonal antibodies was extracted from whole rabbit serum (Berkeley Antibody Company (BABCO) , Berkeley, CA) by FPLC by passing the IgG fraction in a 1 M Tris buffer pH 8 over an XK16/20 column (Pharmacia) containing Protein A Sepharose Fast FlowTM in 100 mM Tris.
  • the IgG fraction is stripped from the column using low pH, 50 mM Na citrate pH 3.5.
  • Fractions are collected from the column utilizing a UV-1 monitor and a FRAC 100TM fraction collector. The pH of the fractions are raised to physiologic values (in the range of pH 7-8) upon collection.
  • the non-specific IgG content is then passed to another XK16/20 column containing activated CH Sepharose 4BTM conjugated to angiotensin I.
  • the specific IgG are eluted from the column in similar fashion as to that of the non-specific IgG fraction.
  • the specific antibody undergoes gel filtration and buffer exchange through a Sephacryl S-100TM column.
  • Specific polyclonal anti-angiotensin I antibodies were further purified by i munoaffinity chromatography using angiotensin I (10 mg/ml, Bachem/Cal, Torrance, CA) conjugated to activated CH-SepharoseTM 4B (Pharmacia Fine Chemicals, Piscataway, N.J.). Fractions were evaluated by dot blot analysis for the detection of peptide antibody. Specific antibodies were diluted with 50 mM sodium phosphate, 27.6 mM maleic acid, 3 mM EDTA, 1.4 mM phenyl ethyl sulfonyl-fluoride, 0.01% neomycin sulfate, 0.9% NaCl, pH 7.8.
  • polyclonal anti-angiotensin I antibody (Berkeley Antibody Company (BABCO) ) was prepared at a concentration of approximately 14 mg/ml in 0.1 M sodium phosphate buffer (pH 7.4) based on the absorbance at 280 nm.
  • Polyclonal anti-angiotensin I was brought to a concentration of 2 mg/ml by dilution of 1 ml aliquots with
  • the label was conjugated to the protein by dialysis of the protein samples against aqueous solutions of the labels.
  • Dialysis was carried out in membrane tubing (Spectrum Medical Industries, Inc., Los Angeles) with a molecular weight cut-off of 3500 g/mole. This allowed low molecular weight label and buffer to diffuse in and out of the membrane, which retained the high molecular weight protein.
  • Each of the three protein-label conjugate samples for each label was subjected to a different purification procedure: dialysis, gel filtration, or hydroxylamine treatment followed by gel filtration.
  • Dialysis removes unbound label or any low molecular weight (less than 3500 g/mol) impurities and changes the buffer.
  • Gel filtration differentially elutes unbound label and conjugate from an ion-exchange resin.
  • Treatment with hydroxylamine removes unstable covalent linkages between protein and label.
  • Samples 1 and 4 were dialyzed against 0.1 M phosphate buffer (pH 7.4) for 24 hours at 4°C in the dark. The one liter dialysis solutions were changed several times during this period.
  • Samples 3 and 6 were treated with 0.1 M hydroxylamine hydrochloride by transferring the volume remaining in the dialysis membranes (approximately 2 ml) to 5.0 ml graduated test tubes, adding 0.25 ml 1.0 M hydroxylamine in 0.05 M borate buffer (resulting in pH 5.4), and bringing the volume to 2.5 ml with borate buffer. These samples were kept at 4°C for two hours until purification.
  • Samples 3 and 6 (after hydroxylamine treatment) and samples 2 and 5 were subjected to gel filtration (2.5 ml SephadexTM G-25 columns, Pharmacia Fine Chemicals, Piscataway, N.J.) to change the buffer to saline.
  • the protein volumes were diluted to 2.5 ml with 0.1 M phosphate buffer (pH 7.4).
  • the SephadexTM columns were equilibrated by passing 25 ml phosphate buffer (pH 7.4) through the column.
  • the 2.5 ml samples were added to the top of each of four columns.
  • the first fraction (approximately 2.5 ml) was collected until the sample volume had eluted just to the top of the column.
  • the protein was removed from the column by adding 3.5 ml 0.1 M phosphate buffer (pH 7.4) to the top and collecting until this volume reached the top of the column bed volume.
  • the label to protein ratio (“1/p") in the conjugate was calculated from the ratio of the label and protein concentrations.
  • the label concentration in the conjugate was approximated by using the extinction coefficient of the free label at its wavelength maximum.
  • the protein concentration was approximated from the absorbance at 280 nm using a correction factor for the label contribution to the absorbance at 280 nm; this procedure is known to those skilled in the art and is described in Brinkley, M. , "A Brief Survey of Methods for Preparing Protein Conjugates with Dyes, Haptens, and Cross-linking Reagents," 3(1) Bioconiu ate Chem. 2-13 (1992) .
  • the absorbance at 280 nm is a standard method for determining protein concentration.
  • the properties of free label needed in the calculations are the wavelength maximum, the extinction coefficient, and the ratio of the label absorbance at 280 nm to that at the maximum wavelength. These values were determined for DTAF and TRITC and are shown in Table 1.
  • the extinction coefficient of the protein at 280 nm and the molecular weight used in the calculation were those for human immunoglobulin G (molecular weight 153,000 g/mol, extinction coefficient 212,000 M'cm "1 ) . Practical Handbook of Biochemistry and Molecular Biology 265-268, G.D. Fasman, ed. , CRC Press, Boca Raton, Fla. (1989).
  • the data required from each protein sample was the absorbance at the label wavelength maximum (A ⁇ ) and the absorbance at 280 nm (A 2 g 0nm ) •
  • the label concentration was calculated directly from the absorbance at its wavelength maximum.
  • the protein concentration was calculated from the absorbance at 280 nm minus the label contribution at
  • the fluorescent conjugates were evaluated using radioimmunoassay according to the method of Sealey et al., (1990) ; Sealey, (1991) .
  • a binding affinity for anti- angiotensin I labeled with fluorescent label was obtained which was comparable to untreated anti-angiotensin I antibody.
  • Preservatives such as Tween 80TM at a concentration of 0.001%, can also be added to the aqueous buffer in which the conjugates are stored, to stabilize the antibodies.
  • the conjugates can be lyophilized and stored as a powder.
  • the best conjugate purification method is dialysis or hydroxylamine treatment followed by gel filtration.
  • Example 2 Conjugation of anti-angiotensin I antibody with DTAF.
  • This method is a modification of the procedure used for conjugation of DTAF with antibody in Example 1.
  • Three samples of label were prepared differently to optimize 1/p.
  • Sample A was identical to that used in Example 1.
  • Sample B was conjugated in a label solution twice as concentrated as in Example 1.
  • Sample C was at a lower pH for the conjugation.
  • buffer and label solutions were prepared and stored in the dark at 4°C: (1) buffer A, 0.05 M sodium borate, 0.04 M sodium chloride, pH 9.3; (2) buffer B, 0.05 M sodium bicarbonate, pH 8.2; (3) buffer C, 0.1 M phosphate, 0.2% neomycin, 0.01% sodium azide; (4) label solution A, 6 mg DTAF dissolved in 200 ml borate buffer A resulting in a label concentration of 5.28xl0 "5 M; (5) label solution B, 12 mg DTAF dissolved in 200 ml borate buffer A resulting in a label concentration of 1.06x10 ⁇ M; and (6) label solution C, 6 mg DTAF dissolved in 200 ml bicarbonate buffer B resulting in a label concentration of 5.28xl0 "5 M. 2. Protein preparation
  • Three anti-angiotensin I antibody samples (approximately 2 mg/ml) were prepared and placed in dialysis membranes (molecular weight cut-off 3500 g/mol) as follows: Samples A and B, l ml protein aliquot added to 6 ml borate buffer A; and Sample C, 2 ml protein aliquot added to 6 ml sodium bicarbonate buffer B.
  • the protein samples were placed in the label solutions and stirred overnight in the dark at 4°C as follows: The dialysis membranes were placed near the top of the 200 ml label solutions where they were not disturbed by the stir bar. The volume of the protein sample was carefully maintained below the top of the label solution. Sample A was placed in label solution A. Sample B was placed in label Solution B. Sample C was placed in label solution C.
  • Dialysis was chosen as the most effective and simplest purification method based on the results of Example 1.
  • Each dialysis membrane was dialyzed against 1000 ml phosphate buffer C in the dark at 4°C.
  • the buffer was changed four times during the dialysis, resulting in dialysis against the first 1000 ml of phosphate buffer for 2 hours, the second volume for 12 hours, the third volume for 30 hours, and the fourth volume for 24 hours.
  • the dialysis buffer was changed more frequently at the beginning when the largest amount of unbound label was removed from the sample.
  • the yield after dialysis for sample A was 5.2 ml, sample B 6.25 ml, and sample C 4.8 ml. These samples were separated into 0.5 ml aliquots and stored in the freezer after snap freezing on dry ice. 5. Determination of label/protein ratio
  • the label to protein ratios were determined using the absorption properties of the conjugates as described in Example l.
  • the samples were diluted five times with phosphate buffer to bring the protein absorbance (280 nm) to about 0.5 absorbance units. Some of the samples contained particulate matter, which was removed by centrifugation. The results are shown in Table 3.
  • Samples A and C had identical label/protein ratios (3.6) , indicating that conjugation can be carried out with DTAF and anti-angiotensin I antibody at pH 8.2 rather than pH 9.3. This was important for the scale-up, since less of the protein and DTAF were degraded at the lower pH.
  • Sample B had a higher label/protein ratio (5.01), which was expected because of the larger concentration of DTAF (twice that used in samples A and C) in the conjugation step. In comparison to the results of Example 1, these ratios were slightly higher (3.6 in contrast to approximately 3.3). This was probably due to the larger volume of label solution used in Example 2 than in Example 1 (200 ml versus 100 ml) .
  • NBD chloride 7-chloro-4-nitrobenzo-2-oxo-l,3-diazole
  • This example demonstrates the attachment of the fluorescent label to the antigen or analyte rather than to the antibody.
  • a one:two molar ratio of NBD chloride to angiotensin I was used to ensure that only the most reactive amino group was conjugated with NBD chloride.
  • Angiotensin I (0.0078 g, Aldrich Chemical Co., Milwaukee, WI) and NBD chloride (0.0006 g, 0.5 equivalents, Aldrich) were added to a 10 ml round-bottomed flask (14/20, single neck) with a stir bar. Distilled water (3 ml) and 10% sodium bicarbonate solution (85 microliters) were added to the flask. The flask was capped and stirred at 23°C for 24 hours. At the start of the reaction, the angiotensin I was soluble, but the yellow NBD chloride solids floated on top of the aqueous solution. As the reaction proceeded, the solids dissolved and the aqueous solution turned orange.
  • the reaction mixture was lyophilized to remove the water.
  • the results of a repeated TLC after lyophilization were identical to those of the untreated reaction mixture.
  • aqueous acetic acid extracts were centrifuged to remove suspended material, then evaporated to a film under a jet of nitrogen gas.
  • the yellow residue was redissolved in 0.2 ml 20% acetic acid and applied in a strip covering approximately the central 4 inches of a silica gel G plate (plastic backing, about 1.5 inches from the bottom of the plate) .
  • the strip was applied in this position on the plate to allow slow movement of the solvent when it reached the sample, thus ensuring clean development of the bands and efficient separation.
  • the sample was concentrated into a tight band by chromatographing 20% aqueous acetic acid into the sample band from both sides.
  • the plate was thoroughly air dried, then developed in 4:1:1 n-butanol/acetic acid/water. The development was allowed to proceed overnight and went all the way to the top of the plate. Two fluorescent bands appeared on the plate, one at about a quarter of the way up the plate, and the other almost at the solvent front.
  • the plate was air-dried.
  • the chromatographic layer was scored all the way across on either side of the lower fluorescent band (cut with a scalpel to give a clean-cut edge) , and then a strip was scraped away on the distal side of the clean cut.
  • the chromatogram was turned at right angles to the orientation in which it was originally developed and processed again in 20% aqueous acetic acid. This step was repeated several times, until the yellow color between the scored lines was concentrated into about 1-2 square centimeters of the silica layer. After complete air drying of the layer, the yellow zone was scraped into a small test tube and stirred vigorously with 0.5 ml water. Only a small amount of the yellow color was extracted into the water, and the suspension was filtered through a glass wool plug in a Pasteur pipet. The solid was caught on the filter and washed with another 0.2 ml water.
  • the filter cake was then washed into a fresh tube with 1 ml of 20% acetic acid in water, added in small portions and allowed to pass through the filter-cake slowly.
  • the yellow color eluted from the filter-cake by the time about half of the acetic acid solution was through the filter.
  • Both the water wash and the 20% acetic acid wash were separately evaporated to films under a jet of nitrogen.
  • the molecular weight is 1456.79 g/mol.
  • Example 4 Preparation of solid phase (immobilized) antigen using activated CH-SepharoseTM 4B.
  • Activated CH-SepharoseTM 4B (Pharmacia Fine Chemicals, Piscataway, N.J.) was washed and reswelled with 1 mM HC1 (1 g lyophilized SepharoseTM in 200 ml of 1 mM HC1, several changes) and then combined with 5 mg/ml angiotensin I in 0.1 M NaHC0 3 , 0.5 M NaCl, pH 8.0, by mixing on a rotating table for 8 hours at 4°C.
  • the affinity gel was washed with 0.1 M Tris - HC1, pH 8, to block excess active groups and then with 0.1 M NaHC03, 0.5 M NaCl, pH 8.0, followed by 0.05 M Tris, 0.5 M NaCl, pH 8.0, and 0.05 M sodium acetate, 0.5 M NaCl, pH 4.
  • Solid phase antigen in suitable storage buffer was stored at 4°C.
  • Example 5 Fluorescent immunoassay for determination of plasma angiotensin levels.
  • the antibody capture assay included the following steps. All incubation mixture aliquots were placed in 1.5 ml microcentrifuge tubes.
  • the control incubation mixture 1 contained 1.0 ml fluorescent antibody diluted with 0.05 M phosphate/0.9% NaCl; 100 ⁇ l control plasma consisted of Sigma coagulation control level 1 (catalog # 7916) which was diluted 1:2 with phosphate buffer; and made 3 mM EDTA, 1.2 mM PMSF, 0.0276 M maleic acid and 0.1% Neomycin pH 7.4, or egg albumin, and 100 ⁇ l buffer, and was measured spectrofluorometrically against "blank" tubes of incubation mixture 1 (defined as containing control plasma and buffer but no fluorescent antibody) .
  • incubation mixture 2 contained 1.0 ml diluted fluorescent antibody, 100 ⁇ l control plasma or egg albumin, and 20 ⁇ l of 0.1% immobilized RAA component, such as angiotensin I bound to CH-SepharoseTM 4B, in each of a series of tubes, and increasing amounts of free angiotensin I (Bachem/Cal, Torrance, CA) (0.01 to 10 ng) in the series of tubes.
  • Incubation mixture 3 contained the same amounts of fluorescent antibody and immobilized RAA component as in the second mixture, but not control plasma, and 100 ⁇ l plasma test sample. The incubation mixtures were incubated at room temperature for 90 min on a shaker table. After incubation, the assay tubes were centrifuged at 18,000 x g for 30 min. A 1 ml aliquot of test sample was removed and transferred to a cuvette.
  • Figure 1 is a graph showing the measurement of plasma angiotensin I by the spectrofluorometric assay method versus the radioimmunoassay measurement of plasma angiotensin I, illustrates that there is approximately a 94% correlation between the values achieved by the two methods.
  • log (Y) spectrofluorometric assay-PDC.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Endocrinology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

L'invention concerne un procédé quantitatif de dosage servant à mesurer avec rapidité, sensibilité et précision un constituant de l'axe hormonal rénine-angiotensine-aldostérone ('RAA') dans un spécimen biologique au moyen d'une mesure spectrofluorométrique. Dans le mode de réalisation préféré, on fait incuber une quantité connue d'anticorps à marquage fluorescent et susceptibles d'immunoréaction avec un constituant spécifique de RAA, avec des volumes connus du constituant de RAA conjugué avec un substrat inerte et un spécimen biologique. On détermine les quantités d'anticorps fluorescent fixé et non fixé par spectrofluorométrie, soit après, soit pendant l'incubation. On quantifie ensuite la quantité du constituant de RAA dans le spécimen après élaboration d'une courbe standart basée sur des mesures spectrofluorométriques d'anticorps fluorescent fixé à des quantités croissantes de constituant de RAA dans un deuxième mélange d'incubation. Comme le démontrent des exemples de quantification de l'angiotensine, ce procédé permet de mesurer des constituants de RAA en quantités de picogrammes en minutes et présente un efficacité dans le diagnostic de l'hypertension, de maladies rénales, de maladies des glandes surrénales ou de maladies cardiovasculaires.
PCT/US1994/003595 1993-04-02 1994-04-01 Methode diagnostique de dosage de renine/angiotensine i WO1994023301A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU65289/94A AU6528994A (en) 1993-04-02 1994-04-01 Renin/angiotensin i diagnostic assay

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US042,149 1987-04-24
US4214993A 1993-04-02 1993-04-02

Publications (1)

Publication Number Publication Date
WO1994023301A1 true WO1994023301A1 (fr) 1994-10-13

Family

ID=21920288

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1994/003595 WO1994023301A1 (fr) 1993-04-02 1994-04-01 Methode diagnostique de dosage de renine/angiotensine i

Country Status (2)

Country Link
AU (1) AU6528994A (fr)
WO (1) WO1994023301A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7749725B2 (en) * 1998-09-04 2010-07-06 University Of Utah Research Foundation Drug screening and diagnosis based on paracrine tubular renin-angiotensin system
CN102749455A (zh) * 2012-06-26 2012-10-24 博奥赛斯(天津)生物科技有限公司 血管紧张素ⅱ化学发光免疫定量检测试剂盒及其制备方法

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS63217272A (ja) * 1987-03-06 1988-09-09 Daiichi Rajio Isotope Kenkyusho:Kk 活性型レニン測定法
JPH022935A (ja) * 1988-06-20 1990-01-08 Tosoh Corp アンジオテンシンの測定法
JPH0257976A (ja) * 1989-07-20 1990-02-27 Shionogi & Co Ltd アルドステロンのイムノアツセイおよびそのためのキット
EP0466565A1 (fr) * 1990-07-05 1992-01-15 Sanofi Trousse pour le dosage spécifique de l'angiotensine II

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS63217272A (ja) * 1987-03-06 1988-09-09 Daiichi Rajio Isotope Kenkyusho:Kk 活性型レニン測定法
JPH022935A (ja) * 1988-06-20 1990-01-08 Tosoh Corp アンジオテンシンの測定法
JPH0257976A (ja) * 1989-07-20 1990-02-27 Shionogi & Co Ltd アルドステロンのイムノアツセイおよびそのためのキット
EP0466565A1 (fr) * 1990-07-05 1992-01-15 Sanofi Trousse pour le dosage spécifique de l'angiotensine II

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
DATABASE WPI Section Ch Week 8842, Derwent World Patents Index; Class A12, AN 88-296601 *
DATABASE WPI Section Ch Week 9007, Derwent World Patents Index; Class B04, AN 90-048974 *
DATABASE WPI Section Ch Week 9014, Derwent World Patents Index; Class B01, AN 90-105129 *
J. E. SEALEY: "Plasma renin activity and plasma prorenin assays.", CLINICAL CHEMISTRY., vol. 37, no. 10(B, October 1991 (1991-10-01), WINSTON US, pages 1811 - 1819 *
R. HAMPL ET AL.: "Recent advances in immunoassay of steroid hormones.", TRAC, TRENDS IN ANALYTICAL CHEMISTRY, vol. 8, no. 2, February 1989 (1989-02-01), AMSTERDAM, NL., pages 72 - 75 *
T. P. LEE ET AL.: "A direct radioimmunoassay for aldosterone in unextracted serum and plasma.", CLINICAL CHEMISTRY., vol. 27, no. 12, December 1981 (1981-12-01), WINSTON US, pages 2019 - 2021 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7749725B2 (en) * 1998-09-04 2010-07-06 University Of Utah Research Foundation Drug screening and diagnosis based on paracrine tubular renin-angiotensin system
CN102749455A (zh) * 2012-06-26 2012-10-24 博奥赛斯(天津)生物科技有限公司 血管紧张素ⅱ化学发光免疫定量检测试剂盒及其制备方法
CN102749455B (zh) * 2012-06-26 2014-12-10 博奥赛斯(天津)生物科技有限公司 血管紧张素ⅱ化学发光免疫定量检测试剂盒及其制备方法

Also Published As

Publication number Publication date
AU6528994A (en) 1994-10-24

Similar Documents

Publication Publication Date Title
JP2651060B2 (ja) 同時較正不均質イムノアツセイ用装置
US5914241A (en) Assays and kits for detecting analytes in the presence of cross-reacting substances
US4900662A (en) CK-MM myocardial infarction immunoassay
US5188939A (en) Displacement immunoassay utilizing an oligavalent labelled antibody
EP0461828B1 (fr) Détection de analytes possédant les sites de fixation pour au moins deux entités de fixation
JPH0145026B2 (fr)
US6362008B1 (en) Generic signalling mechanism for detection of analytes
US20060127938A1 (en) Immunoassay for HIV protease inhibitors
JPH0812196B2 (ja) グリコシル化ヘモグロビンのアッセイ
EP0770876A1 (fr) Méthode de screening pour l'identification de ligands des protéines cibles
AU615823B2 (en) Enzyme-linked immunoassay for measurement of cyclosporin a levels in whole blood samples
AU654497B2 (en) Analyte variant analysis
US4837170A (en) Monoclonal antibody, measuring method of glucosylated protein by utilizing the monoclonal antibody, and kit therefor
EP0123265A1 (fr) Activation de la zymogène, immuno-essai avec amplification en cascade
WO1994023301A1 (fr) Methode diagnostique de dosage de renine/angiotensine i
Pulido-Tofiño et al. Flow-through fluoroimmunosensor for isoproturon determination in agricultural foodstuff: Evaluation of antibody immobilization on solid support
US5958785A (en) Detection of a carbohydrate biomarker directly associated with chronic alcoholism
JPH0421819B2 (fr)
US4847194A (en) Colorimetric detection of delta-5-3-ketosteroid isomerase and immunoassay based thereon
Al-Bassam et al. Double-antibody enzyme immunoassay for nortriptyline.
EP0488755B1 (fr) Procédé de mesure rapide de composants de trace
JPS61501047A (ja) 固定化したリポ−タ−グル−プ類の測定方法
CA1195612A (fr) Reactif pour le dosage de la kallikreine dans l'urine des humains
Haasnoot et al. 5. Immunochemical and receptor technologies
AU601674B2 (en) A method for determining a ligand

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP

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

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

Ref country code: CA