Method For Virus Detection

Borg; Peter ;   et al.

Patent Application Summary

U.S. patent application number 12/994189 was filed with the patent office on 2011-03-24 for method for virus detection. This patent application is currently assigned to GE HEALTHCARE BIO-SCIENCES AB. Invention is credited to Peter Borg, Asa Frostell-Karlsson, Markku Hamalainen, Anita Larsson.

Application Number20110070574 12/994189
Document ID /
Family ID41398332
Filed Date2011-03-24

United States Patent Application 20110070574
Kind Code A1
Borg; Peter ;   et al. March 24, 2011

METHOD FOR VIRUS DETECTION

Abstract

A method of determining the concentration of a virus or antigen thereof in a sample comprises the steps of: providing a sensor surface having immobilized thereto a virus antigen or a virus antigen analogue, mixing the sample with a known amount of antibody to the virus antigen to obtain a predetermined concentration of antibody to the antigen in the sample mixture, contacting the sample mixture with the sensor surface to bind free antibody in the mixture to the sensor surface, measuring the response of the sensor surface to the binding of free antibody, and determining the concentration of the virus or antigen in the sample from a calibration curve prepared by measuring the responses obtained for mixtures containing the predetermined concentration of antibody and different concentrations of virus.


Inventors: Borg; Peter; (Uppsala, SE) ; Frostell-Karlsson; Asa; (Uppsala, SE) ; Hamalainen; Markku; (Uppsala, SE) ; Larsson; Anita; (Uppsala, SE)
Assignee: ; GE HEALTHCARE BIO-SCIENCES AB
Uppsala
SE

Family ID: 41398332
Appl. No.: 12/994189
Filed: June 1, 2009
PCT Filed: June 1, 2009
PCT NO: PCT/SE09/50637
371 Date: November 23, 2010

Current U.S. Class: 435/5 ; 436/501
Current CPC Class: G01N 21/658 20130101; G01N 21/553 20130101; G01N 33/54373 20130101; G01N 33/54306 20130101
Class at Publication: 435/5 ; 436/501
International Class: C12Q 1/70 20060101 C12Q001/70; G01N 33/53 20060101 G01N033/53

Foreign Application Data

Date Code Application Number
Jun 2, 2008 SE 0801304-7
Apr 24, 2009 SE 0950272-5

Claims



1: A method of determining the concentration of a virus or antigen thereof in a sample comprising the steps of: providing a sensor surface having immobilized thereto a virus antigen or a virus antigen analogue; mixing the sample with a known amount of antibody to the virus antigen to obtain a predetermined concentration of antibody to the antigen in the sample mixture; contacting the sample mixture with the sensor surface to bind free antibody in the mixture to the sensor surface; measuring the response of the sensor surface to the binding of free antibody; and determining the concentration of the virus or antigen in the sample from a calibration curve prepared by measuring the responses obtained for mixtures containing the predetermined concentration of antibody and different concentrations of virus or virus antigen, wherein multiple analysis cycles are performed on the sensor surface with intermediate regenerations and a virtual calibration curve is calculated for each analysis cycle.

2. (canceled)

3: The method of claim 1, wherein calculating a calibration curve comprises: fitting each of the known concentrations in the curves to a double exponential equation using cycle number as x and response as y; using these equations for calculation of a virtual calibration curve for each cycle; and determining the concentration of the virus or antigen in the sample from a virtual calibration curve for that particular cycle.

4: The method of claim 1, wherein the concentration of at least two, preferably at least three different viruses or virus antigens in a sample are determined, and wherein mixtures of sample and antibodies selective to the respective virus antigens are successively, in separate analysis cycles, contacted with a sensor surface having a different one of the antigens or analogues immobilized thereto.

5: The method of claim 1, wherein the concentration of at least two, preferably at least three different viruses or antigens in a sample are determined, and wherein mixtures of sample and antibodies selective to the respective virus antigens are contacted with a sensor surface having discrete sensing areas with a respective antigen or analogue immobilized thereto.

6: The method of claim 1 wherein the concentration of at least two, preferably at least three different viruses or antigens in a sample are determined, and wherein mixtures of sample and antibodies selective to the respective virus antigens are contacted with a sensor surface comprising a mix of respective antigen immobilized to the same sensing area.

7: The method of claim 1, wherein the concentration of at least two, preferably at least three different viruses or virus antigens in a sample are determined, and wherein a mixture of sample and antibodies selective to the respective virus antigens, in a single analysis cycle, is contacted with a sensor surface having discrete sensing areas with a respective antigen or analogue immobilized thereto.

8. (canceled)

9: The method of claim 1, wherein the virus is an influenza virus.

10: The method of claim 1, wherein the different viruses are selected from influenza virus types and subtypes.

11: The method of claim 1, wherein the virus antigen comprises hemagglutinin.

12: The method of claim 1, wherein the sample is derived from influenza vaccine production.

13: The method of claim 12, wherein the sample is derived from a multivalent influenza vaccine.

14: The method of claim 12, wherein the virus antigen comprises hemagglutinin, and the immobilized hemagglutinin is generic for a plurality of different strains of an influenza virus type or subtype.

15: The method of claim 12, wherein the virus antigen comprises hemagglutinin, and the immobilized hemagglutinin is derived from at least 2 different strains of an influenza virus type or subtype.

16: The method of claim 13, wherein the multivalent influenza vaccine is trivalent and derived from influenza A/H1N1, A/H3N2 and B strains.

17: The method of claim 1, wherein binding interactions on the sensor surface are detected by mass-sensing, preferably evanescent wave sensing, particularly surface plasmon resonance (SPR).

18-19. (canceled)
Description



CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application is a filing under 35 U.S.C. .sctn.371 and claims priority to international patent application number PCT/SE2009/050637 filed Jun. 1, 2009, published on Dec. 10, 2009 as WO 2009/148395, which claims priority to application number 0801304-7 filed in Sweden on Jun. 2, 2008 and application number 0950272-5 filed in Sweden on Apr. 24, 2009.

FIELD OF THE INVENTION

[0002] The present invention relates to the detection and quantification of virus or virus antigen in a virus-containing or virus-derived medium, particularly determination of the virus antigen concentration in vaccine manufacturing and process development.

BACKGROUND OF THE INVENTION

[0003] Influenza viruses are generally divided into three types, A, B and C, based on the antigenic differences between their nucleoprotein and matrix protein antigens. Influenza A viruses are further divided into subtypes on the basis of the two main surface glycoproteins hemagglutinin (HA) and neuraminidase (NA) which appear as spikes on the surface of the viral body.

[0004] Infection of a host cell starts with the binding of virus HA to sialic structures on the cell causing the virus particles to stick to the cell surface and induce uptake of the virus. RNA and viral proteins are duplicated and assemble into new viral particles, which bud from the cell. The progeny virus particles are released from the cell surface by the enzyme NA on the virus cleaving terminal sialic residues.

[0005] Currently, there are 15 different HA subtypes (H1-H15) and 9 different NA subtypes (N1-N9). Subtypes of influenza A virus are named according to their HA and NA surface proteins, e.g. H1N1, H1N2, H3N2, H5N1. Influenza B viruses and substypes of influenza A are further characterized into strains, e.g. B Malaysia, H3N2 Beijing, H1N1 Taiwan.

[0006] Both HA and NA carry antigenic epitopes. Antibodies raised against HA and NA reduce the risk of infection or illness in humans and animals. While the first influenza vaccines contained inactivated or killed whole virus particles, commercially available influenza vaccines are usually of two types, "split vaccines" and "subunit vaccines".

[0007] Split vaccines are prepared by disintegration of purified virus particles with ether or detergents, and then removal of the detergent with the bulk of the viral lipid material. Split vaccines thereby contain essentially the same elements as whole virus vaccines and in the same proportions. In subunit vaccines, on the other hand, the surface glycoproteins HA and NA are purified separately and then combined into a vaccine.

[0008] Influenza viruses can change in two different ways, by "antigenic drift" and by "antigenic shift". Antigenic drift produces new virus strains that may not be recognized by the body's immune system, whereas antigenic shift is an abrupt, major change in the influenza A viruses, resulting in new hemagglutinin and neuraminidase proteins and producing a new influenza A subtype. In most years, one or two of the three virus strains in the influenza vaccine are updated to keep up with the changes in the circulating flu viruses.

[0009] An influenza vaccine is usually multivalent (polyvalent), i.e. the vaccine is prepared from cultures of two or more strains of the same species of virus. Currently, influenza A/H1N1, A/H3N2 and influenza B strains are typically included in each year's influenza vaccine.

[0010] The efficacy of a vaccination against influenza is largely determined by the amount of immunogenic HA in a vaccine. The HA concentration in vaccines has typically been determined by single radial immuno-diffusion (SRID) assay. In SRID, influenza virions are disrupted by detergent, and submitted to diffusion of whole virus or purified viral antigens into agarose gel containing specific anti-hemagglutinin (anti-HA) antibodies. The resulting antigen/antibody reaction or zone (visible by staining) is directly proportional to the amount of HA antigen in the preparation. However, the SRID assay has a number of disadvantages. In addition to having a high detection limit (about 20 .mu.g/ml) with high variation (about 10%), it is laborious and has a low throughput. Despite its shortcomings, SRID is still, however, the method recommended by the European Pharmacopeia and WHO and approved by regulatory authorities for the evaluation of influenza vaccines. Due to the poor precision of the method, vaccine doses are usually "overfilled" by the manufacturers.

[0011] Other methods for quantification of influenza virus include reversed-phase high performance liquid chromatography (RP-HPLC).

[0012] Also biosensor-based methods have been developed for the detection of virus, including influenza virus.

[0013] EP 0276142 A1 discloses a method for detecting influenza A virus using surface plasmon resonance (SPR) on a gold coated diffraction grating. Monoclonal antibodies to discrete determinants of influenza A virus were immobilized on the gold surface, and virus was applied and incubated. Monoclonal antibodies to the influenza A virus determinants were then incubated with the surface, and the enhanced response obtained when the second antibody was bound to the influenza virus particle was detected.

[0014] JP 3054467A discloses measurement of the concentration of a virus by a piezoelectric vibrator having its electrode coated with an antibody to a surface antigen of a virus. When antigen binds to the electrode, the vibration frequency of the vibrator becomes lower.

[0015] Shofield, D. J., and Dimmock, N. J., J. Virol. Methods 62 (1996) 33-42 discloses use of an SPR biosensor instrument for detection of influenza virus. A monoclonal antibody for capture of influenza virus was coupled to a sensor chip coated with carboxylated dextran. Influenza virus was then injected into the instrument flow system to contact the sensor chip, and the binding affinity with the immobilized antibody was monitored.

[0016] Boltovets, P. M., et al., J. Virol. Methods 121 (2004) 101-106 discloses detection of plant virus using surface plasmon resonance (SPR) by detecting the binding of complexes between viral antigen and antibody formed during a pre-incubation step to an SPR sensor surface with immobilized protein A.

[0017] Jie Xu, et al., Analytical and Bioanalytical Chemistry 389, 4 (2007) 1193-1199 discloses an interferometric biosensor immunoassay for detection of avian influenza. Whole virus particles were captured by antigen-specific (hemagglutinin) antibodies (both polyclonal and monoclonal) on a waveguide surface, and the refraction changes resulting from the binding were measured. Three influenza virus subtypes (two H7 and one H8) were tested.

[0018] All these other methods, however, also suffer from substantial disadvantages. There is therefore a need for improved methods and means for accurate determination of the concentration of influenza virus, specifically HA concentration, in crude as well as in purified samples.

SUMMARY OF THE INVENTION

[0019] One object of the present invention is to provide a method for detection and quantification of virus, especially influenza virus, which is devoid of the disadvantages of the prior art methods, and which in particular has a high precision, a high detection range, and is less laborious than the standard SRID assay.

[0020] Another object of the present invention is to provide a method which is suitable for quantifying virus or virus antigen in vaccine production, both in process samples and final vaccine samples.

[0021] These objects as well as other objects and advantages are obtained with a method according to claim 1.

[0022] The method according to the present invention is based on the use of biosensor-technology and an inhibition type assay format.

[0023] Broadly, the method of determining the concentration of a virus or virus antigen in a sample comprises the steps of:

[0024] providing a sensor surface having immobilized thereto a virus antigen or a virus antigen analogue,

[0025] mixing the sample with a known amount of antibody to the virus antigen to obtain a predetermined (total) concentration of antibody to the antigen in the sample mixture,

[0026] contacting the sample mixture with the sensor surface to bind free antibody in the mixture to the sensor surface,

[0027] measuring the response of the sensor surface to the binding of free antibody, and

[0028] determining the concentration of the virus or antigen in the sample from a calibration curve prepared by measuring the responses obtained for mixtures containing the predetermined concentration of antibody and different concentrations of virus or virus antigen.

[0029] Preferably, multiple analysis cycles are performed on the sensor surface with intermediate regenerations and a virtual calibration curve is calculated for each analysis cycle. This may be done by:

[0030] fitting each of the known concentrations in the curves to a double exponential equation using cycle number as x and response as y,

[0031] using these equations for calculation of a virtual calibration curve for each cycle and,

[0032] determining the concentration of the virus or antigen in the sample from a virtual calibration curve for that particular cycle.

[0033] The virus antigen may be an internal antigen or, preferably, a surface antigen of the virus. Optionally, the virus antigen is the whole virus particle. A virus antigen to analogue may, for example, be a synthetic peptide.

[0034] Preferably, the sample contains a plurality of different virus or virus types which are determined simultaneously by the method.

[0035] The term "antibody" as used herein refers to an immunoglobulin which may be natural or partly or wholly synthetically produced and also includes active fragments, including Fab antigen-binding fragments, univalent fragments and bivalent fragments. The term also covers any protein having a binding domain which is homologous to an immunoglobulin binding domain. Such proteins can be derived from natural sources, or partly or wholly synthetically produced. Exemplary antibodies are the immunoglobulin isotypes and the Fab, Fab', F(ab').sub.2, scFv, Fv, dAb, and Fd fragments.

[0036] Typically, the antibody is a serum to the virus or virus antigen.

[0037] In one embodiment, the sensor chip has multiple sensing areas and virus antigens or analogues specific to a respective virus or virus type are immobilized on different discrete sensing areas. The sample is mixed with a fixed amount of the antibody to one virus, and the mixture is then contacted with either only the sensing area with the antigen specific to the antibody, or--provided that the antibody does not cross-react with the other antigens or analogues--all sensing areas, and the response is detected. This is successively repeated for the other antibodies and sensing areas.

[0038] In another embodiment, the sensor chip has a single sensing area, or only a single sensing area of a sensor chip with multiple sensing surfaces is used. In this case, the virus antigens or analogues are co-immobilized on a single sensing area, the sample is mixed with a fixed amount of a respective antibody, one at a time, and the respective mixtures are successively contacted with the sensing area.

[0039] In a preferred embodiment, however, provided that the antibodies are selective lacking cross-reactivity to the other viruses or antigens, all the different antibodies are to mixed with the sample which is then contacted with a sensor chip with multiple sensing areas, each with a respective immobilized virus antigen or analogue, and the responses of the different sensing areas are detected. The sensor surface may alternatively comprise a mix of respective antigen immobilized to the same sensing area. In this way, all viruses or virus antigens in a sample may be detected in a single analytical cycle.

[0040] The viruses or virus antigens in a sample to be quantified are preferably different influenza virus types or antigens thereof, preferably hemagglutinins.

[0041] The immobilized hemagglutinin may be generic for several strains of influenza virus types or subtypes or be derived from at least 2 different strains of an influenza virus type or subtype.

[0042] Generally, in a biosensor assay, when analytes (here free antibodies in the sample), have bound to immobilized ligands (here virus antigen or analogue) on a sensor surface, the bound antibodies are released by treatment with a suitable fluid to prepare the surface for contact with a new sample, a process referred to as regeneration. Usually, a sensor surface can be subjected to fairly large number of analysis cycles. Many ligands (such as e.g. virus antigens), however, often have poor stability making the analyte binding capacity of the surface decrease with the number of cycles and may hamper the use of the ligand for quantitative purposes. While minor decreases in binding capacity can often be compensated by frequent calibrations, this significantly decreases the throughput.

[0043] The method of the invention therefore includes a normalization step wherein each analysis cycle is evaluated using a virtual calibration curve (i.e. each cycle obtains a unique calibration curve), thus minimizing the need of frequent calibrations during drift and significantly improving the quality of the quantitative measurements.

[0044] A more complete understanding of the present invention, as well as further features and advantages thereof, will be obtained by reference to the following detailed description and drawings.

BRIEF DESCRIPTION OF THE DRAWINGS

[0045] FIG. 1 is a schematic illustration of an inhibition type virus assay on a sensor surface for three cases (a-c) with different virus concentrations in the sample.

[0046] FIG. 2 is a schematic illustration similar to FIG. 1 where three different virus antigens are immobilized to respective separate spots on a sensor surface and three different antibodies specific for each antigen are used for quantification.

[0047] FIG. 3 is a diagram showing measured relative response/stability versus concentration of different influenza virus/anti-serum mixtures in an inhibition type assay with immobilized influenza virus antigen on a sensor surface.

[0048] FIG. 4 is a diagram showing measured relative response versus analysis cycle number for the binding of a plurality of different influenza virus anti-sera to a sensor surface with immobilized influenza virus antigen.

[0049] FIG. 5 is a diagram showing fitted normalisation curves based on cycle number as x and the measured response as y for seven concentrations of virus control samples at four ordinary calibrations in an inhibition type assay run on a sensor surface with immobilized virus antigen. These normalisation curves are used for prediction of virtual concentrations for each cycle. These virtual concentrations are then used for the construction of a cycle specific calibration curve using the virtual concentration as x and the known concentration as y.

[0050] FIG. 6 is a diagram showing calculated concentration versus analysis cycle number for two different control sample concentrations with four ordinary calibrations at different cycle numbers in an inhibition type assay run on a sensor surface with immobilized virus antigen.

[0051] FIG. 7 is a diagram showing application of a virtual calibration curve (according to FIG. 5) for each cycle to the same raw data as in FIG. 6 for two control sample concentrations.

[0052] FIG. 8 is a similar diagram as FIG. 7 showing measured binding data for a control sample and corresponding data normalized by a virtual calibration curve for each cycle.

[0053] FIGS. 9A-C show calibration curves prepared in an assay for simultaneous detection of three different virus types.

DETAILED DESCRIPTION OF THE INVENTION

[0054] As mentioned above, the invention relates to a method for the detection and quantification of at least one virus or virus antigen in a sample medium, using biosensor technology and an inhibition type assay format.

[0055] First, with regard to biosensor technology, a biosensor is broadly defined as a device that uses a component for molecular recognition (for example a layer with immobilised antibodies) in either direct conjunction with a solid state physicochemical transducer, or with a mobile carrier bead/particle being in conjunction with the transducer. While such sensors are typically based on label-free techniques detecting a change in mass, refractive index or thickness for the immobilized layer, there are also biosensors relying on some kind of labelling. Typical sensors for the purposes of the present invention include, but are not limited to, mass detection methods, such as optical methods and piezoelectric or acoustic wave methods, including e.g. surface acoustic wave (SAW) and quartz crystal microbalance (QCM) methods. Representative optical detection methods include those that detect mass surface concentration, such as reflection-optical methods, including both external and internal reflection methods, which may be angle, wavelength, polarization, or phase resolved, for example evanescent wave ellipsometry to and evanescent wave spectroscopy (EWS, or Internal Reflection Spectroscopy), both of which may include evanescent field enhancement via surface plasmon resonance (SPR), Brewster angle refractometry, critical angle refractometry, frustrated total reflection (FTR), scattered total internal reflection (STIR) (which may include scatter enhancing labels), optical wave guide sensors, external reflection imaging, evanescent wave-based imaging such as critical angle resolved imaging, Brewster angle resolved imaging, SPR-angle resolved imaging, and the like. Further, photometric and imaging/microscopy methods, "per se" or combined with reflection methods, based on for example surface enhanced Raman spectroscopy (SERS), surface enhanced resonance Raman spectroscopy (SERRS), evanescent wave fluorescence (TIRF) and phosphorescence may be mentioned, as well as waveguide interferometers, waveguide leaking mode spectroscopy, reflective interference spectroscopy (RIfS), transmission interferometry, holographic spectroscopy, and atomic force microscopy (AFR).

[0056] Biosensor systems based on SPR as well as on other detection techniques including QCM, for example, are commercially available, both as flow-through systems having one or more flow cells and as cuvette-based systems. Exemplary SPR-biosensors with multiple sensing surfaces and a flow system include the BIACORE.TM. systems (GE Healthcare, Uppsala, Sweden) and the PROTEON.TM. XPR36 system (Bio-Rad Laboratories). These systems permit monitoring of surface binding interactions in real time between a bound ligand and an analyte of interest. In this context, "ligand" is a molecule that has a known or unknown affinity for a given analyte and includes any capturing or catching agent immobilized on the surface, whereas "analyte" includes any specific binding partner thereto.

[0057] With regard to SPR biosensors, the phenomenon of SPR is well known. Suffice it to say that SPR arises when light is reflected under certain conditions at the interface to between two media of different refractive indices, and the interface is coated by a metal film, typically silver or gold. In the BIACORE.TM. system, the media are the sample and the glass of a sensor chip which is contacted with the sample by a microfluidic flow system. The metal film is a thin layer of gold on the chip surface. SPR causes a reduction in the intensity of the reflected light at a specific angle of reflection. This angle of minimum reflected light intensity varies with the refractive index close to the surface on the side opposite from the reflected light, in the BIACORE.TM. system the sample side.

[0058] A detailed discussion of the technical aspects of the BIACORE.TM. instruments and the phenomenon of SPR may be found in U.S. Pat. No. 5,313,264. More detailed information on matrix coatings for biosensor sensing surfaces is given in, for example, U.S. Pat. Nos. 5,242,828 and 5,436,161. In addition, a detailed discussion of the technical aspects of the biosensor chips used in connection with the BIACORE.TM. instrument may be found in U.S. Pat. No. 5,492,840. The full disclosures of the above-mentioned U.S. patents are incorporated by reference herein.

[0059] While in the Examples that follow, the present invention is illustrated in the context of SPR spectroscopy, and more particularly a BIACORE.TM. system, it is to be understood that the present invention is not limited to this detection method. Rather, any affinity-based detection method where an analyte binds to a ligand immobilised on a sensing surface may be employed, provided that a change at the sensing surface can be measured which is quantitatively indicative of binding of the analyte to the immobilised ligand thereon.

[0060] Now to the detection and quantification assay. Generally, in an inhibition type assay (also called solution competition), a known amount of a detecting molecule (here an antibody) is mixed with the sample (here a virus), and the amount of free detecting molecule in the mixture is measured. More specifically, an inhibition type assay for to concentration measurements in the present biosensor context may typically comprise the following steps:

1. The analyte or a derivative thereof is attached to the sensor surface as ligand. 2. A constant (known or unknown) concentration of detecting molecule is added to different concentrations of the calibrant solutions (analyte). 3. The mixtures are contacted with the sensor surface (injected over the surface in a flow system) and the response is measured. 4. Calibration curves are calculated. 5. The measurements are then performed by mixing the samples (analyte) with the constant concentration of detecting molecule, the samples are contacted with the sensor surface (injected over the surface in a flow system) and the response is measured. 6. The calibration curve is used for calculation of the analyte concentration in the sample. The amount of free detecting molecule is inversely related to the concentration of analyte in the sample. 7. The surface is regenerated and a new sample can be injected.

[0061] In the method of the present invention, the ligand is a virus antigen, preferably a surface antigen (or optionally the whole virus), whereas the analyte is an antibody to the antigen. The antibody may be polyclonal, e.g. serum, or monoclonal. Due to the inhibition type assay format, diffusion effects of the large virus particles to the surface are avoided.

[0062] For purposes of illustration, without any limitation thereto, the invention will in the following be described with regard to the determination of the concentration of at least one influenza virus in a sample, and more particularly of the concentrations of hemagglutinin (HA) of three different virus types in a trivalent flu vaccine. Reference is made to FIGS. 1 to 9 in the accompanying drawings.

[0063] As schematically depicted in FIG. 1a, purified virus HA designated by reference numeral 1 is immobilized on a biosensor sensor surface 2. A mixture of virus particles 3 and anti-serum containing antibodies 4 is made to pass as a liquid flow over the sensor surface 2. As illustrated in FIG. 1a, the antibodies 4 can either be bound to the virus particle or to the immobilized HA antigen or be free in solution. Binding to the sensor surface increases the response signal from the sensor surface.

[0064] FIG. 1b illustrates the case when no virus is present in the sample. A maximum amount of antibodies 4 then bind to the HA antigen 1 on the sensor surface, resulting in a high response signal.

[0065] In FIG. 1c, on the other hand, a high concentration of virus particles 3 results in a low amount of free antibodies 4, and a low response signal is therefore measured. Thus, the higher the concentration of virus in the sample, the lower is the amount of binding antibodies to the surface HA, resulting in a lower response level.

[0066] If the sensor surface has, or is capable of providing multiple discrete sensing areas or "spots", such as three or more, e.g. three different HA's may be immobilized as is schematically illustrated in FIG. 2, where HA specific to virus types/subtypes A/H1N1, A/H3N2 and B (which are typically used in current flu vaccines) are immobilized to the respective spots on the sensor surface.

[0067] As will be demonstrated below, the binding of different virus anti-sera to HA is selective, i.e. there is no cross-reactivity between different virus types or subtypes. Due to this selectivity, two or more different virus components in a sample, such as a multivalent vaccine, may be determined simultaneously.

[0068] An exemplary method embodiment of the invention applied to a sample containing the three above-mentioned virus types/subtypes A/H1N1, A/H3N2 and B will now be described.

[0069] HA from the three different virus types is immobilized on three different spots on the sensor surface.

[0070] A calibration procedure is then performed. Calibrants consisting of a fixed concentration of a standard anti-serum for each virus type are mixed with different known concentrations of virus (or virus antigen) covering the concentration range to be measured. The calibrants are then injected, either separately or together for all three types, over the sensor surface spots and the response is measured. From the results of the measurements, calibration curves are then calculated.

[0071] Measurement of the sample content of virus HA is then performed by mixing each virus with the fixed concentration of the anti-serum, either one at a time, or, preferably, with all three anti-sera. The sample is injected over the sensor surface and the free anti-sera concentration is measured. The calibration curve is used for the calculation of virus antigen concentration in the sample.

[0072] The surface is then regenerated (i.e. bound antibodies are dissociated from the immobilized HA by contacting the surface with a suitable regeneration fluid), and a new sample can be passed over the surface.

[0073] By mixing the sample with all three anti-sera and injecting the sample over all three spots as preferred above, the concentration of the HA from the three different virus types/subtypes can be analyzed in a single analysis cycle. An assay may therefore be developed which can simultaneously measure all virus components of a multivalent, e.g. trivalent, flu vaccine.

[0074] As will be shown below, it has been found that there is substantial cross-reactivity between different strains of influenza virus subtypes to the hemagglutinin of a strain of the subtype. It is therefore likely that a generic assay for each of the common virus strains could be developed. Such an assay could thus have general use for measuring the virus antigen content in a flu vaccine irrespective of the yearly changing combination of virus strains thereof.

[0075] In the above described assay, a prerequisite for high quality analytical results is a constant binding capacity of the ligand (HA) immobilized on the sensor surface and a high stability of the calibrants injected over the surface. Generally, a minor decrease in binding capacity can often be compensated by frequent calibrations. However, frequent calibrations decrease the throughput and increase the cost due to reagent consumption.

[0076] According to the present invention a method has been devised where each analysis cycle is evaluated using a "virtual" calibration curve, thus minimizing the need of frequent calibration during drift and significantly improving the quality of quantitative measurements using biosensor systems, such as e.g. the above-mentioned BIACORE.TM. systems. While the method basically is generally applicable to any ligand, and may be used in various different assay formats including inter alia direct binding assays, inhibition assays and sandwich assays, the method has particular relevancy in the present virus detection context, since virus antigens like HA usually exhibit significant instability causing drift on sensor surfaces.

[0077] While the binding capacity of the sensor surface decreases, the measured/calculated concentration of the controls increases as a function of the number of analytical cycles performed (or cycle number) until a new calibration run is performed, since in the inhibition type assay format the calibration curve interprets the binding capacity decrease as an increased HA concentration in the sample. This drift increases with an increased number of analysis cycles, and is often exponential. In the present method, an analysis cycle includes the steps of passing the mixture of virus and detecting antibodies over the sensor surface with immobilized HA, and then regenerating the surface to prepare it for the next analysis cycle.

[0078] In accordance with the invention, the new calibration routine can be designed in different ways.

[0079] In one variant, raw data from calibration runs is used for prediction of virtual concentrations for each analysis cycle followed by calculation of a cycle specific calibration curve and prediction of the concentration for the sample/control.

[0080] In another variant, calibration equations are calculated for each of the real calibrations followed by prediction of calibration coefficients for each cycle which are then used for prediction of samples/controls.

[0081] The first-mentioned method above will be described in more detail in Examples 4 and 5, and the second variant in Example 6 below.

[0082] An assay kit for carrying out the method of the present invention for analysis of, for example, a multivalent flu vaccine may comprise hemagglutinin (or a hemagglutinin analogue) for the target influenza virus types/subtypes, standard sera and virus standards for the virus types/subtype, and optionally also a sensor chip.

[0083] In the following Examples, various aspects of the present invention are disclosed more specifically for purposes of illustration and not limitation.

EXAMPLES

[0084] The present examples are provided for illustrative purposes only, and should not be construed as limiting the invention as defined in the appended claims.

Instrumentation

[0085] A BIACORE.TM. T100 (GE Healthcare, Uppsala, Sweden) was used. This instrument, which is based on surface plasmon resonance (SPR) detection at a gold surface on a sensor chip, uses a micro-fluidic system (integrated micro-fluidic cartridge--IFC) for passing samples and running buffer through four individually detected flow cells, designated Fc 1 to Fc 4, one by one or in series. The IFC is pressed into contact with the sensor chip by a docking mechanism within the BIACORE.TM. T100 instrument.

[0086] As sensor chip was used Sensor Chip CM5 (GE Healthcare, Uppsala, Sweden) which has a gold-coated (about 50 nm) surface with a covalently linked hydrogel matrix (about 100 nm) of carboxymethyl-modified dextran polymer.

[0087] The output from the instrument is a "sensorgram" which is a plot of detector response (measured in "resonance units", RU) as a function of time. An increase of 1000 RU corresponds to an increase of mass on the sensor surface of approximately 1 ng/mm.sup.2

Example 1

Assay for Influenza Virus A/H3N2/Wyoming, A/H3N2/New York and B/Jilin

Materials

[0088] Hemagglutinin (HA) A/H3N2, Wyoming/3/2003, Wisconsin and New York was from Protein Sciences Corp., Meriden, USA. HA A/H1N1, New Caledonia/20/99 was from ProsPec, Rehovot, Israel. HB/Jilin was from GenWay Biotech Inc., San Diego, USA. Sera as well as virus strains were from NIBSC--National Institute for Biological Standards and Control, Potters Bar, Hertfordshire, U.K. Assay and sample buffer: HBS-EP+, GE Healthcare.

Surfactant P20, GE Healthcare.

Method

[0089] HA (H3N2, H1N1 and B) are immobilized to a Sensor Chip CM5 in three respective flow cells of the BIACORE.TM. T100 using amine coupling as follows: H3N2/Wyoming and Wisconsin: 10 .mu.g/ml in 10 mM phosphate buffer, pH 7.0, 0.05% Surfactant P20, 7 min. H3N2/New York: 10 .mu.g/ml in 10 mM maleate buffer, pH 6.5, 0.05% Surfactant P20, 7 min. B/Jilin: 5 .mu.g/ml in 10 mM maleate buffer, pH 6.5, 0.05% Surfactant P20, 20-30 min. Immobilisation levels are 5000-10000 RU. Sera to the respective virus strains are diluted using a dilution factor based on SRID titre, as recommended by the supplier. E.g. 7 .mu.l serum diluted to 1 ml for SRID corresponds to .times.200 dilution in the BIACORE.TM. T100. (Dilutions are made to obtain approximately 500-1500 RU.) The injection time is 5 min. Three to ten start-up cycles with serum are performed. Calibration curves are prepared with virus antigen (HA), first diluted in MQ as recommended by the supplier (HA is then kept frozen in aliquots) and then further diluted in sera to typically 0.1-15 .mu.g/ml. Standards and samples have 400 s injection time. Regeneration is performed with 20-50 mM HCl, 0.05% Surfactant P20, 30 s followed by 30 s stabilization.

Example 2

Generality of Detection of Different Strains of the Same Virus Subtype

[0090] H3N2 strain Wyoming HA was immobilized to a Sensor Chip CM5 and the surface was contacted with different virus/antiserum combinations: virus/anti-serum from Wyoming (W/W); virus/anti-serum from New York (N.Y./N.Y.), Wyoming virus and serum from New York (W/N.Y.); New York virus and serum from Wyoming (N.Y./W). Calibration curves with the respective combinations were run. The results are shown in FIG. 3. From the figure, it is clear that there is cross-reactivity between the different virus strains. The Wyoming HA and virus/anti-serum can therefore be used for quantification of the New York strain and vice versa.

Example 3

Selectivity in Binding of Anti-Sera to Different Influenza Virus Types/Subtypes HA

[0091] 27 different anti-sera to different strains of influenza virus A/H3N2, A H1N1 and B were injected over immobilized H3N2 Wyoming HA and the binding thereof was detected. The results as well as a listing of the strains used are indicated in FIG. 4. As apparent from the figure, all H3N2 anti-sera bind with signals higher than 100 RU while all H1N1 and B anti-sera have signals below 50 RU. This indicates that several virus strains may be quantified simultaneously and that one or only a few HA's are required for measurement of H3N2.

Example 4

Virtual Calibration Procedure

[0092] A number of assay cycles (about 100) were run on the BIACORE.TM. T100 and a Sensor Chip CM5, during which four ordinary calibrations were performed with seven different concentrations of control samples (0.156, 0.31, 0.625, 1.25, 2.5, 5 and 10 .mu.g/ml). The function y(x)=a*exp(-b*x)+c*exp(-d*x)+e, using cycle number as x, response as y, and a, b, c, d and e as fitted parameters, was fitted for each of the seven different concentrations. The results are shown in FIG. 5, the top curve represents the lowest concentration of the control (i.e. the highest response--inhibition assay) and the bottom curve the highest (i.e. the lowest response). The equations were then used for calculation of a virtual response for each cycle. These responses were then used for the calculation of a calibration curve for each cycle which were used for the prediction of samples and controls run at exactly that cycle, as described below with reference to FIGS. 6 and 7.

[0093] FIG. 6 illustrates the drift on the calculated concentration of 2 controls, 1.0 .mu.g/ml and 0.5 .mu.g/ml. A large number of assay cycles were run and four intermediate calibrations were performed at cycle numbers indicated by the double dotted arrows. The concentrations of the 3 (2) controls following each calibration were calculated against the closest preceding calibration curve. As indicated by the dotted arrows, there is a systematic increase in calculated concentrations with increased distance to the calibration. This increase in calculated concentration is due to a decreased signal from the control sample. This is in turn due to a decrease in binding capacity of the surface as a function of cycle numbers, which the calibration curve interprets as an increased concentration. This decrease in binding capacity is also visible in FIG. 3 and FIG. 4.

[0094] Application of the virtual calibration method described above to the raw data in FIG. 6 gives the concentration estimates for shown in FIG. 7 for the 0.5 and 1.0 .mu.g/ml controls, which is a considerable improvement of the repeatability in the prediction of the concentration of control samples.

Example 5

Normalization of Binding Data by a Virtual Calibration Procedure

[0095] HA recombinant proteins HB/Jilin, H1N1/New Caledonia and H3N2/Wyoming were immobilized. Calibration curves were obtained. Samples were diluted and concentrations between 0.5-15 .mu.g/ml were measured and recalculated. To avoid drift of the response, the results were normalized using the normalization procedure outlined in Example 4 above, each cycle obtaining a unique calibration curve. FIG. 8 shows the results before and after normalization for control samples, 5 .mu.g/ml of Baiangsu/10/2003, giving a response of 250 RU, CV=1.2%.

Example 6

Simultaneous Detection of Three Different Virus Types

[0096] Three flow cells were immobilized with three different recombinant influenza virus HA proteins: H1N1/New Caledonia, H3N2/Wisconsin and B/Jilin.

Virus standards from the three influenza strains, H1N1/New Caledonia, H3N2/Wisconsin and B/Malaysia, were diluted and mixed together so that the final concentration of each standard was 16 .mu.g/ml. Calibration curves were then made as 2-fold serial dilutions from 16 .mu.g/ml to 0.5 .mu.g/ml.

[0097] The three vaccines, H1N1, H3N2 and B, to be analysed, were diluted 8, 16, 32 and 64 times.

[0098] Three serums (H1N1/New Caledonia, H3N2/Wisconsin and B/Malaysia from NIBSC) were diluted to concentrations giving responses of 500-700 RU and mixed together.

TABLE-US-00001 Prior mix with ag End dilution H1N1/New Caledonia 60x dilution 180x H3N2/Wisconsin 70x dilution 210x B/Malaysia 20x dilution 60x

[0099] To analyze the vaccines, duplicates of the standards and vaccines were first mixed with the serum solution and then allowed to flow through all flow cells using a method created in "Method Builder".

[0100] The general method from "Method Builder":

Start-up (7 cycles, buffer instead of sample followed by regeneration. 2 hours) Calibration curve 1 (14 cycles) Samples (12 cycles) Calibration curve 2 (14 cycles) Samples (12 cycles) Calibration curve 3 (14 cycles).

[0101] The results were then normalized in respect to the three calibration curves. This was done by performing a four parameter fit of the calibration curves to the four-parameter regression curve (Equation 1) conventionally used for concentration determinations with BIACORE.TM. systems to determine the four coefficients:

Response = R high - ( R high - R low ) 1 + ( X A 1 ) A 2 ( 1 ) ##EQU00001##

[0102] where R.sub.high is the response at low virus concentration, R.sub.low is the response at low virus concentration, A.sub.1 (EC50) and A.sub.2 (Hill slope) are fitting parameters and X is the concentration of virus.

[0103] The values obtained for each one of the four coefficients at the different concentrations were then plotted against analysis cycle number, whereby an equation for each coefficient was obtained. Using the coefficients obtained with Equation 1 above, the normalized concentrations were calculated.

Results

TABLE-US-00002 [0104] Concentration Surface HA Sample (ug/ml) Std dev CV % B vaccine 1 102.9 1.07 1.0 vaccine 2 30.9 0.30 1.0 vaccine 3 27.8 0.44 1.6 H1N1 vaccine 1 37.4 0.05 0.1 vaccine 2 34.9 0.04 0.1 vaccine 3 30.5 0.10 0.3 H3N2 vaccine 1 36.2 0.02 0.1 vaccine 2 27.8 0.06 0.2 vaccine 3 37.7 0.12 0.3

[0105] According to the manufacturers, the HA concentration of each strain in the vaccine should be 30 .mu.g/ml, analyzed with SRID.

[0106] The three calibration curves are shown in FIG. 9 (the above-mentioned calibration curves 1 to 3 in each figure).

[0107] From the foregoing, it will be appreciated that, although specific embodiments of this invention have been described herein for purposes of illustration, various modifications may be made without departing from the spirit and scope of invention. Accordingly, the invention is not limited except by the appended claims.

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed