US20050266503A1 - Simultaneous assay of target and target-drug binding - Google Patents

Simultaneous assay of target and target-drug binding Download PDF

Info

Publication number
US20050266503A1
US20050266503A1 US11/136,168 US13616805A US2005266503A1 US 20050266503 A1 US20050266503 A1 US 20050266503A1 US 13616805 A US13616805 A US 13616805A US 2005266503 A1 US2005266503 A1 US 2005266503A1
Authority
US
United States
Prior art keywords
antibody
drug
free
target
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/136,168
Inventor
Norman Purvis
Gregory Stelzer
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Esoterix Inc
Original Assignee
Esoterix Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Esoterix Inc filed Critical Esoterix Inc
Priority to US11/136,168 priority Critical patent/US20050266503A1/en
Assigned to ESOTERIX, INC. reassignment ESOTERIX, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PURVIS, NORMAN B.,, STELZER, GREGORY T.
Publication of US20050266503A1 publication Critical patent/US20050266503A1/en
Priority to US11/927,223 priority patent/US7943327B2/en
Priority to US13/088,841 priority patent/US8389224B2/en
Priority to US13/759,158 priority patent/US8828671B2/en
Priority to US14/452,772 priority patent/US20140357522A1/en
Priority to US15/007,342 priority patent/US20160161514A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • G01N33/94Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving narcotics or drugs or pharmaceuticals, neurotransmitters or associated receptors
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5091Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing the pathological state of an organism
    • 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
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • 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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • 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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • 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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6872Intracellular protein regulatory factors and their receptors, e.g. including ion channels
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/70514CD4
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70546Integrin superfamily, e.g. VLAs, leuCAM, GPIIb/GPIIIa, LPAM
    • G01N2333/70553Integrin beta2-subunit-containing molecules, e.g. CD11, CD18
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70596Molecules with a "CD"-designation not provided for elsewhere in G01N2333/705
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/715Assays involving receptors, cell surface antigens or cell surface determinants for cytokines; for lymphokines; for interferons
    • G01N2333/7158Assays involving receptors, cell surface antigens or cell surface determinants for cytokines; for lymphokines; for interferons for chemokines
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/10Composition for standardization, calibration, simulation, stabilization, preparation or preservation; processes of use in preparation for chemical testing
    • Y10T436/101666Particle count or volume standard or control [e.g., platelet count standards, etc.]

Definitions

  • the invention relates to a new cell based method of detecting or measuring drug activity, by simultaneously measuring target and target-drug combinations by flow cytometry.
  • the method uses a pair of antibodies, one that is drug insensitive and will indicate total target level regardless of whether drug is present or not.
  • the second is drug sensitive and will not bind to target at the same time that drug is bound to target.
  • the drug and second antibody thus compete for the same or overlapping binding sites. Both antibodies can be simultaneously quantified when differentially labeled.
  • a drug target (the protein to which a drug binds in order to achieve its intended effect) is purified and the drug's effect on target activity over time is measured using increasing concentrations of the drug. Drug kinetics are then determined using standard data manipulations, such as the Scatchard plot or Lineweaver Burke.
  • protein purification and multiple assays make such methods laborious and not conducive to the high throughput generation of data.
  • purified proteins by definition are outside their normal body environment, and the changes in environment can complicate or change the way a protein behaves. This can result in misleading or incomplete information about a drug's activity and kinetics.
  • Anti-Total antibody is an antibody that binds to the target at an epitope that is separate or independent from the drug binding site. Thus, such an antibody will detect “total” target level in a given sample.
  • Drug is any pharmaceutical agent. Drugs as used herein also include the use of antibodies and their derivatives as therapeutic agents.
  • Target is defined as the protein that a given drug interacts with.
  • Target-drug is the target as it binds to or otherwise interacts with the drug of interest.
  • MESF and ABC can be calculated directly by the software that controls the cytometer.
  • the invention provides quantitative cell-based measurements of experimental drugs designed to bind to very specific protein “targets.” It allows real time detection of target level and drug-bound target level, thus simplifying and improving on the prior art methods of studying drug kinetics.
  • the method generally is a cell based, two antibody assay that allows detection of total target in the sample and the simultaneous detection of that percentage of targets that are drug-bound and/or drug-free. Simultaneous detection is achieved by using different labels that can each be detected at the same time. In a preferred embodiment, the method uses flow cytometry to detect the different labels simultaneously.
  • the method generally involves the detection of total target using a labeled antibody that binds to the target.
  • target-drug binding pairs are detected with a second labeled antibody that binds to the drug binding site on the target.
  • the principle assumption in the method is that binding of the drug to the target will result in the subsequent inhibition of an antibody directed at drug binding site of the target. Should this assumption hold true, the relationship between the drug blockade of antibody binding and the concentration of drug would form the basis of a very sensitive and specific “inhibition” immunoassay for bound drug.
  • Validation of the assay requires evaluation of several monoclonal antibodies in order to identify suitable antibody pairs, as follows: 1) a first antibody that is not inhibited by drug binding, thus providing measurement of total target level, and 2) a second antibody that is effectively inhibited by drug binding, thus proving the central assumption of competition between antibody and drug binding. If demonstrable antibody inhibition by drug binding is observed, then the assay can be calibrated by performing a drug inhibition standard curve. In this manner, the level of drug binding to target can be calculated based upon the level of antibody staining.
  • the cells are fixed to allow for cytometric analysis. Both cell surface and internal targets can be studied, provided the fixation method is sufficiently gentle to retain cell morphology together with a good level of staining.
  • the two antibodies are then simultaneously detected using flow cytometry, wherein the cytometer is appropriately gated to allow detection of the two labels at the same time.
  • additional labels can be used to type the cells according to surface antigens.
  • the cells need not be separated prior to study and complex samples, such as whole blood, can be studied.
  • the labels should be chosen with the operating characteristics of the cytometer in mind such that there is sufficient separation of signal so as to allow the cytometer to distinguish between the two or more signals.
  • Many such labels are known in the art, including fluorescent isothiocyanate (FITC, aka fluorescein), Phycoerythrin (PE); Cy5PE; Cy7PE; Texas Red (TR); Allophycocyanin (APC); Cy5; Cy7APC; Cascade Blue; and the like.
  • a great many conjugated antibodies are commercially available and these can easily be screened to identify antibody pairs with the requisite binding characteristics (Total and Free-Site binding). Those antibodies that are not conjugated can easily be conjugated with an appropriate dye using one of the many available conjugation kits. Commercial suppliers will also provide custom antibodies on demand.
  • cells taken from subjects exposed to various drug concentrations can be assayed to determine the proportion of total available drug targets per cell that are occupied by drug (% saturation).
  • % saturation the proportion of total available drug targets per cell that are occupied by drug
  • the method allows for simultaneous and independent quantification of target saturation on each different cell population.
  • PK pharmocokinetic
  • the PK application utilizes a cell line that constitutively expresses the receptor protein target to which the drug binds. Incubation of the cell line with a standard range of drug concentrations followed by staining of the cells with fluorochrome-conjugated drug sensitive monoclonal antibodies allows for the construction of a very specific standard curve of antibody fluorescence as it inversely relates to drug concentration. Subsequent to the construction of the standard curve, it is possible to derive the amount of drug in specimens of unknown concentration by extrapolation of the degree of drug-induced inhibition of antibody fluorescence to drug concentration as defined by the standard curve.
  • the invention is a method of measuring drug activity in whole cells by reacting cells with labeled anti-Total antibody (detects total target in the sample) and with a labeled anti-Free-Site antibody (detects only drug free target).
  • the cells are fixed and both antibodies are quantified by flow cytometry.
  • the level of drug bound target is proportional to the level of anti-total antibody staining minus the anti-Free-Site antibody staining.
  • the method can also be applied to mixed populations of cells if the cells are pre-typed by staining with a third antibody.
  • the method is also applicable to both cell surface and internal targets.
  • the method can be used to determine a variety of drug kinetic parameters, including pharmacokinetics and pharmacodynamics. It can also be used to determine circulating free drug levels in serum or plasma if used with a standard curve of anti-Free-Site antibody binding versus drug concentration.
  • FIG. 1 Flow Cytometry: FIG. 1A shows side-scatter (SSC-H) on the x-axis plotted against forward-scatter (FSC-H) on the y-axis. All nucleated cells remaining in the lysed blood sample are indicated. The cytometer is then gated to separate the APC-labeled and non-labeled cells in FIG. 1B , which shows side-scatter (SSC-H) on the x-axis plotted against the APC labeled anti-CD14 antibody scatter (“CD14-APC”) on the y-axis. The separated monocytes (moderate side-scatter, CD14+) and neutrophils (high side-scatter, CD14 ⁇ ) are indicated.
  • SSC-H side-scatter
  • FSC-H forward-scatter
  • CD14-APC APC labeled anti-CD14 antibody scatter
  • FIG. 2 Antibody Binding Characteristics: Fluorescence intensity (MESF) on the x-axis is plotted against the total number of cells (Number) on the y-axis.
  • the left panel indicates ⁇ AgX-FITC, which is the drug insensitive antibody (anti-Total).
  • the right panel is ⁇ AgX-PE, which is the drug sensitive antibody (anti-Free-Site). Varying amounts of drug were added to each sample (0-Top, 50 ng/ml-middle, 100 ng/ml-bottom). Inhibition of the drug sensitive antibody by drug is indicated by a loss of fluorescence intensity in the staining by the anti-Free-Site antibody, shown by the peak shifting to the left (right panel).
  • FIG. 3 Total Antibody Binding to Neutrophils: Drug concentration in ng/ml on the x-axis is plotted against ⁇ AgX-FITC ABC. The antibody staining is constant, thus this antibody detects total AgX levels and is not drug sensitive.
  • FIG. 4 Drug Sensitive Antibody Binding to Neutrophils: Drug concentration in ng/ml on the x-axis is plotted against ⁇ AgX-PE ABC. The antibody staining decreases with increasing drug concentration, thus this antibody binds to the drug binding site and the drug competes with antibody binding.
  • FIG. 6 Standard Curve: Standard curve of free drug concentration verses fluorescence (MESF) of the drug sensitive anti-AgX antibody on a target cell line.
  • the present invention is exemplified with respect to CD11b and a proprietary drug known by the code name Neutrophil Inhibitory Factor.
  • the invention has general applicability to the study of any drug-target combinations for which suitable antibody pairs can be identified.
  • the following examples describe the invention as practiced in additional detail, but should not be construed as limiting.
  • conjugated antibodies against various antigens were identified and ordered for evaluation in the method of the invention.
  • Non-conjugated, purified antibodies as well as proprietary pharmaceutical grade antibodies can also be obtained for evaluation and potential custom conjugation.
  • All anti-AgX antibodies against were evaluated in triplicate in the presence and absence of saturating concentrations of the appropriate drug (in each instance, the drug is specific for a single epitope on the protein AgX) by performing individual multi-point two-fold serial dilutions of each antibody. Additional cell subtype specific antibodies labeled with different fluorochromes may be added to each tube to allow the study of specific populations of cells as necessary.
  • Antibody titration data was plotted to show 1) staining intensity versus antibody staining concentration, 2) percent positive staining versus concentration and 3) the signal to noise ratio versus concentration. These plots aided in the identification of the optimal staining concentration of each antibody, indicated whether saturation staining was possible for each antibody and indicated whether the particular antibody measured total AgX expression or was inhibited by the presence of drug.
  • Anti-Total and anti-Free-Site antibodies were used to allow monitoring of drug occupancy on the cells of interest during PK and PD experiments. Ex-vivo spiking experiments using appropriate biological samples spiked at varied concentrations of the drug were used to verify operational characteristics of the antibody cocktails and proper drug saturation calculations based on anti-Total and anti-Free-Site antibody measurements.
  • the antibodies, target and drugs validated to date for use in the methods of the inventions include: TABLE 2 Exemplary Target-Drug Combinations and Antibody Pairs Anti-Total Anti-Free Site Target Drug Antibody Vendor (Cat. #) Antibody Vendor (Cat.
  • CD11b Neutrophil anti-human Beckman Coulter anti-human BD Biosciences inhibitory factor CD11b-FITC (IM0530) CD11b-PE (30455S) CD11a chimeric anti-human BD Biosciences anti-human Dako-Cytomation humanized CD11a-PE (555384) CD11a-FITC (F0712) anti-CD11a CD18 Proprietary anti-human BD Biosciences anti-human Dako-Cytomation CD18-PE (555924) CD18-FITC (F0839) CD22 Proprietary anti-human Serotec anti-human Immunotech CD22-FITC (MCA553F) CD22-PE (IM1835) CD25 Proprietary anti-human BD Biosciences anti-human BD Biosciences CD25-PE (555432) CD25-PE (341009)
  • Additional antibody pairs that can be used in the general method of the invention for particular target and drug combinations can be identified using the method described generally in Example 1. Some possible combinations are listed in Table 3. However, the combinations are unlimited and additional combinations can identified by searching MEDLINE, ATCC or the web. TABLE 3 Additional Target-Drug Combinations and Antibody Pairs Target Drug Antibodies CD20 Rituximab, PRO70769, Rituximab, Bexxar Bexxar (Tositumomab), B1, Immu-106, Ibritumomab, HI47, L27 CD52 Campath-1H Campath-1H (Alemtuzumab) CD33 In Development HB-10306, Mab 251, M195, huM195, PC251, L4F3 CD4 In Development OKT4, TNX-355, HuMax-CD4, GK1.5, W3/25, YTS177.9, ORTHOCLONE OKTcdr4a CD3 In Development HB231, HB10166, UCHT1, PS1,
  • the cells were stained with the appropriate antibody or antibodies.
  • a saturating amount (as determined in the titration evaluation of each antibody against the target) of desired antibody was added to each tube, and the tubes were incubated at 37° C. for 1 hr. Immediately after incubation, the cells were collected by centrifugation, and erythrocytes were lysed using a standard ammonium chloride lysing procedure.
  • the cells were collected by centrifugation, and 2 ml of cold PBS+2% FCS was added to each tube.
  • the tubes were placed in the dark at room temperature for at least 10 minutes and up to 1 hour to allow RBC lysis upon resuspension of the fixed whole blood samples in this isotonic solution.
  • the cells were collected again and washed once or twice with PBS+2% FCS with a final resuspension in 1% paraformaldehyde in PBS. The samples were thus ready for data collection.
  • FIG. 1A shows light scatter patterns of the nucleated cells (leukocytes) stained with anti-CD14-APC.
  • FIG. 1B an electronic gating window was constructed around the nucleated cells to separate the cells based on the expression of the CD14 marker. The APC signal was used to separate the leukocytes into CD 14 positive cells (monocytes) and CD14 negative (neutrophils).
  • Additional electronic gating windows can be applied to these two cell populations for simultaneous and independent analysis of the binding of the drug sensitive antibody (anti-Free-Site antibody) and the drug insensitive antibody (anti-Total) on each cell type respectively. This type of analysis is shown in FIG. 2 .
  • Antibody Pair Binding As above, whole blood was incubated with or without drug, stained with the appropriate antibodies, fixed, lysed and analyzed. This particular experiment was designed so that drug insensitive antibody (anti-Total) was labeled with FITC ( ⁇ AgX-FITC) and drug sensitive antibody (anti-Free-Site) was labeled with PE ( ⁇ AgX-PE). However, other labels could be used. Blood was treated with three different concentrations of drug and only the neutrophil results are shown in FIG. 2 .
  • the histograms in FIG. 2 show fluorescence intensity at ⁇ 525 nm (left panel, FITC signal) and ⁇ 575 nm (right panel, PE signal) on the x-axis plotted against the number of cells counted (Number) on the y-axis.
  • FITC signal left panel
  • PE signal right panel
  • the ⁇ AgX-PE peak shifts to the left indicating a decrease in fluorescence intensity.
  • anti-Free-Site antibody binding decreases due to the competition by the drug for the drug sensitive epitope.
  • the fluorescence intensity of the anti-AgX-FITC stain was invariant at all drug concentrations, indicating constant binding of this antibody to the drug insensitive epitope.
  • Target-Drug Binding In the next two experiments, broader ranges of drug concentrations were investigated. Whole blood was incubated with various concentrations of drug ranging from 0-200 ng/ml, followed by antibody staining, fixing, lysis and analysis as described above. The data in FIG. 3 demonstrates constant binding of drug insensitive (anti-Total) antibody over a broad range of drug concentration. In contrast, the data in FIG. 4 demonstrates a dose dependent inhibition of the binding of the drug sensitive anti-AgX-PE (anti-Free-Site) to neutrophils.
  • Standard Curve A target cell line (HL-60) was exposed to different drug concentrations ranging from 0 to 150 ng/ml. Subsequent to drug incubation, the cells were stained with drug sensitive anti-AgX and binding of the fluorescent antibody was quantified on the cytometer as described above and used to prepare this standard curve. Note: This curve was performed using monoclonal antibody binding intensity (MESF). However, the same curve may be constructed using monoclonal ABC.
  • EMF monoclonal antibody binding intensity
  • Unknown concentrations of drug in patient specimens can be derived by extrapolation of quantitative fluorescence of the anti-AgX antibody to the respective drug concentration on this standard curve.
  • Acceptable fixatives are defined herein as fixatives that allow both cell fixation and permeation, while retaining cell surface morphology and DNA and RNA content, sufficient to allow separation of cells based on light scatter, surface epitopes and/or nucleic acid content.
  • An acceptable fixative contains 0.75-0.85% formaldehyde, 25-30 mM DNBS, 6.8-7% DMSO and 0.08-0.1% Tween 20 detergent.
  • a preferred fixative is PermiFlow.TM Mild denaturing temperatures are those temperature that improve access to internal antigens, without compromising cell morphology, surface antigens or nucleic acid content. Preferred temperatures range from 39 to 43° C., and most preferred 43° C.

Abstract

Whole cell, simultaneous target and drug-target assay using differentially labeled antibodies and flow cytometry. First antibody binds to total target and second antibody binds to the drug binding site of the target, thus drug binding will competitively inhibit the second antibody allowing for a competitive inhibition assay of drug-target binding. The assay allows for whole cell analysis and even analysis of mixed populations of cells, yet provides detailed kinetic assessment of drug activity.

Description

    PRIOR RELATED APPLICATIONS
  • This application claims the benefit of U.S. Provisional Application No. 60/573,783 filed May 24, 2004.
  • FEDERALLY SPONSORED RESEARCH STATEMENT
  • Not applicable.
  • FIELD OF THE INVENTION
  • The invention relates to a new cell based method of detecting or measuring drug activity, by simultaneously measuring target and target-drug combinations by flow cytometry. The method uses a pair of antibodies, one that is drug insensitive and will indicate total target level regardless of whether drug is present or not. The second is drug sensitive and will not bind to target at the same time that drug is bound to target. The drug and second antibody thus compete for the same or overlapping binding sites. Both antibodies can be simultaneously quantified when differentially labeled.
  • BACKGROUND OF THE INVENTION
  • Previous methods of measuring drug activity have been laborious and complex. Typically, a drug target (the protein to which a drug binds in order to achieve its intended effect) is purified and the drug's effect on target activity over time is measured using increasing concentrations of the drug. Drug kinetics are then determined using standard data manipulations, such as the Scatchard plot or Lineweaver Burke.
  • However, protein purification and multiple assays make such methods laborious and not conducive to the high throughput generation of data. Further, purified proteins by definition are outside their normal body environment, and the changes in environment can complicate or change the way a protein behaves. This can result in misleading or incomplete information about a drug's activity and kinetics.
  • What is needed in the art is a method that allows the simultaneous detection of both total target and target-drug binding that would simplify and improve the accuracy of the determination of drug kinetics. It would be preferred if the method allowed such measurements without the prior purification of targets, for example in whole cells. It would be especially preferred if mixed populations of cells, such as are found in whole blood, could be studied without the need for prior separation of cell populations.
  • SUMMARY OF THE INVENTION
  • The following definitions are used herein:
  • “Anti-Total” antibody is an antibody that binds to the target at an epitope that is separate or independent from the drug binding site. Thus, such an antibody will detect “total” target level in a given sample.
  • “Anti-Free-Site” antibody is an antibody that binds to the drug binding or interaction site on the target, such that binding of the drug to the target and binding of the anti-free site antibody to the target are exclusive. Thus, such an antibody will detect drug-free target and be inhibited by drug binding. Bound target-drug concentration can thus be determined, as follows:
    [Total target]=[Free-Site]+[Target-drug]
    [Target-drug]=[Total target]−[Free-Site]
  • “Drug” is any pharmaceutical agent. Drugs as used herein also include the use of antibodies and their derivatives as therapeutic agents.
  • By “simultaneous” what is meant is that the measurements are taken at the same time from the same sample, whether or not the cytometer performs the measurements at the same actual instant or performs the measurements sequentially.
  • “Target” is defined as the protein that a given drug interacts with.
  • “Target-drug” is the target as it binds to or otherwise interacts with the drug of interest.
  • The following abbreviations are used in herein:
    TABLE 1
    Abbreviations
    Abbre-
    viation Expansion
    ABC Antibody Binding Capacity - The ABC is the number of
    monoclonal antibodies a sample will bind, and correlates
    to the number of antigens expressed on the cell surface.
    Eff. F/P The effective number of fluorochrome molecules conjugated
    per each antibody molecule determined by measured
    fluorescence intensity of antibody capture microspheres
    measured on a flow cytometer calibrated in units of
    fluorchrome specific MESF
    F/P Number of fluorochrome molecules conjugated per each
    antibody molecule determined by absorbance on a
    spectrophotometer.
    FCS Fetal Calf Serum
    FSC-H Forward angle light scatter
    MESF Molecules of Equivalent Soluble Fluorochrome. Corrects
    for changes in extinction coefficient, quenching, and
    small spectra shifts. Using the appropriate calibration
    controls, MESF and ABC can be calculated directly by the
    software that controls the cytometer.
    MFI Mean Fluorescence Intensity
    PBS phosphate buffered saline (200 mg/l KCl; 200 mg/l KH2PO4;
    8 g/l NaCl; 2.16 g/l Na2HPO4.7H2O, pH 7.4)
    PD Pharmacodynamics
    PK Pharmacokinetics
    SSC-H Right angle light scatter
    αAgX anti-antigen X - an antibody directed against a generic
    antigen called X
  • The invention provides quantitative cell-based measurements of experimental drugs designed to bind to very specific protein “targets.” It allows real time detection of target level and drug-bound target level, thus simplifying and improving on the prior art methods of studying drug kinetics.
  • The method generally is a cell based, two antibody assay that allows detection of total target in the sample and the simultaneous detection of that percentage of targets that are drug-bound and/or drug-free. Simultaneous detection is achieved by using different labels that can each be detected at the same time. In a preferred embodiment, the method uses flow cytometry to detect the different labels simultaneously.
  • The method generally involves the detection of total target using a labeled antibody that binds to the target. At the same time, target-drug binding pairs are detected with a second labeled antibody that binds to the drug binding site on the target. The principle assumption in the method is that binding of the drug to the target will result in the subsequent inhibition of an antibody directed at drug binding site of the target. Should this assumption hold true, the relationship between the drug blockade of antibody binding and the concentration of drug would form the basis of a very sensitive and specific “inhibition” immunoassay for bound drug.
  • Validation of the assay requires evaluation of several monoclonal antibodies in order to identify suitable antibody pairs, as follows: 1) a first antibody that is not inhibited by drug binding, thus providing measurement of total target level, and 2) a second antibody that is effectively inhibited by drug binding, thus proving the central assumption of competition between antibody and drug binding. If demonstrable antibody inhibition by drug binding is observed, then the assay can be calibrated by performing a drug inhibition standard curve. In this manner, the level of drug binding to target can be calculated based upon the level of antibody staining.
  • After antibody staining, the cells are fixed to allow for cytometric analysis. Both cell surface and internal targets can be studied, provided the fixation method is sufficiently gentle to retain cell morphology together with a good level of staining. The two antibodies are then simultaneously detected using flow cytometry, wherein the cytometer is appropriately gated to allow detection of the two labels at the same time. In a preferred embodiment, additional labels can be used to type the cells according to surface antigens. Thus, the cells need not be separated prior to study and complex samples, such as whole blood, can be studied.
  • The labels should be chosen with the operating characteristics of the cytometer in mind such that there is sufficient separation of signal so as to allow the cytometer to distinguish between the two or more signals. Many such labels are known in the art, including fluorescent isothiocyanate (FITC, aka fluorescein), Phycoerythrin (PE); Cy5PE; Cy7PE; Texas Red (TR); Allophycocyanin (APC); Cy5; Cy7APC; Cascade Blue; and the like.
  • A great many conjugated antibodies are commercially available and these can easily be screened to identify antibody pairs with the requisite binding characteristics (Total and Free-Site binding). Those antibodies that are not conjugated can easily be conjugated with an appropriate dye using one of the many available conjugation kits. Commercial suppliers will also provide custom antibodies on demand.
  • We have exemplified the method with whole blood by first lysing the RBCs in isotonic solution, and then by separating the cells of interest based on gating according to surface antigen staining. For example, using CD14 as a surface antigen, it was possible to separately analyze both monocytes and neutrophils in the same blood sample. The method can be generally applied and other cell populations, such as bone marrow, can also be studied.
  • In this novel approach to cell-based analysis of drug pharmacodynamics (PD), cells taken from subjects exposed to various drug concentrations can be assayed to determine the proportion of total available drug targets per cell that are occupied by drug (% saturation). In addition, should the target be expressed at various levels in different populations of cells within the sample (or a different levels between patients), the method allows for simultaneous and independent quantification of target saturation on each different cell population.
  • An alternative application of this method is for the analysis of free drug in plasma or serum of subjects exposed to various drug dosage regimens. Data from this assay is useful in pharmocokinetic (PK) modeling of functional drug activity. The PK application utilizes a cell line that constitutively expresses the receptor protein target to which the drug binds. Incubation of the cell line with a standard range of drug concentrations followed by staining of the cells with fluorochrome-conjugated drug sensitive monoclonal antibodies allows for the construction of a very specific standard curve of antibody fluorescence as it inversely relates to drug concentration. Subsequent to the construction of the standard curve, it is possible to derive the amount of drug in specimens of unknown concentration by extrapolation of the degree of drug-induced inhibition of antibody fluorescence to drug concentration as defined by the standard curve.
  • In one embodiment, the invention is a method of measuring drug activity in whole cells by reacting cells with labeled anti-Total antibody (detects total target in the sample) and with a labeled anti-Free-Site antibody (detects only drug free target). The cells are fixed and both antibodies are quantified by flow cytometry. The level of drug bound target is proportional to the level of anti-total antibody staining minus the anti-Free-Site antibody staining. The method can also be applied to mixed populations of cells if the cells are pre-typed by staining with a third antibody. The method is also applicable to both cell surface and internal targets.
  • The method can be used to determine a variety of drug kinetic parameters, including pharmacokinetics and pharmacodynamics. It can also be used to determine circulating free drug levels in serum or plasma if used with a standard curve of anti-Free-Site antibody binding versus drug concentration.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1. Flow Cytometry: FIG. 1A shows side-scatter (SSC-H) on the x-axis plotted against forward-scatter (FSC-H) on the y-axis. All nucleated cells remaining in the lysed blood sample are indicated. The cytometer is then gated to separate the APC-labeled and non-labeled cells in FIG. 1B, which shows side-scatter (SSC-H) on the x-axis plotted against the APC labeled anti-CD14 antibody scatter (“CD14-APC”) on the y-axis. The separated monocytes (moderate side-scatter, CD14+) and neutrophils (high side-scatter, CD14−) are indicated.
  • FIG. 2. Antibody Binding Characteristics: Fluorescence intensity (MESF) on the x-axis is plotted against the total number of cells (Number) on the y-axis. The left panel indicates αAgX-FITC, which is the drug insensitive antibody (anti-Total). The right panel is αAgX-PE, which is the drug sensitive antibody (anti-Free-Site). Varying amounts of drug were added to each sample (0-Top, 50 ng/ml-middle, 100 ng/ml-bottom). Inhibition of the drug sensitive antibody by drug is indicated by a loss of fluorescence intensity in the staining by the anti-Free-Site antibody, shown by the peak shifting to the left (right panel).
  • FIG. 3. Total Antibody Binding to Neutrophils: Drug concentration in ng/ml on the x-axis is plotted against αAgX-FITC ABC. The antibody staining is constant, thus this antibody detects total AgX levels and is not drug sensitive.
  • FIG. 4. Drug Sensitive Antibody Binding to Neutrophils: Drug concentration in ng/ml on the x-axis is plotted against αAgX-PE ABC. The antibody staining decreases with increasing drug concentration, thus this antibody binds to the drug binding site and the drug competes with antibody binding.
  • FIG. 5. Percent Saturation on Neutrophils: Drug concentration in ng/ml on the x-axis is plotted against percent saturation (αAg Drug saturation) on the y-axis. Percent saturation is calculated by: % Saturation=[(αAgX-FITC ABC−αAg X-PE ABC)/(αAg X-FITC ABC)]X100.
  • FIG. 6. Standard Curve: Standard curve of free drug concentration verses fluorescence (MESF) of the drug sensitive anti-AgX antibody on a target cell line.
  • DESCRIPTION OF EMBODIMENTS OF THE INVENTION
  • The present invention is exemplified with respect to CD11b and a proprietary drug known by the code name Neutrophil Inhibitory Factor. However, the invention has general applicability to the study of any drug-target combinations for which suitable antibody pairs can be identified. The following examples describe the invention as practiced in additional detail, but should not be construed as limiting.
  • EXAMPLE 1 Antibody Optimization
  • The following method was employed to validate the method of the invention using the targets and antibodies described in Table 2, below. However, it can be applied to any novel combinations of antibodies to find suitable antibody pairs useful for any target-drug combinations of interest.
  • Commercially available conjugated antibodies against various antigens (generically called AgX) were identified and ordered for evaluation in the method of the invention. Non-conjugated, purified antibodies as well as proprietary pharmaceutical grade antibodies can also be obtained for evaluation and potential custom conjugation.
  • All anti-AgX antibodies against were evaluated in triplicate in the presence and absence of saturating concentrations of the appropriate drug (in each instance, the drug is specific for a single epitope on the protein AgX) by performing individual multi-point two-fold serial dilutions of each antibody. Additional cell subtype specific antibodies labeled with different fluorochromes may be added to each tube to allow the study of specific populations of cells as necessary.
  • Antibody titration data was plotted to show 1) staining intensity versus antibody staining concentration, 2) percent positive staining versus concentration and 3) the signal to noise ratio versus concentration. These plots aided in the identification of the optimal staining concentration of each antibody, indicated whether saturation staining was possible for each antibody and indicated whether the particular antibody measured total AgX expression or was inhibited by the presence of drug.
  • If multiple anti-Total AgX antibodies and anti-Free-Site AgX antibodies were identified, the antibodies that provided the best signal to noise ratio were chosen. In cases were the commercially available antibodies are sub-saturating or not available in the ideal fluorochrome conjugate, custom conjugates can be ordered or prepared in-house. The effective F/P (effective fluorescence to probe ratio) was determined for each of the anti-Total and anti-Free-Site antibodies identified for use in the drug saturation assay.
  • Drug specific antibody panels were and will continue to be developed. Anti-Total and anti-Free-Site antibodies were used to allow monitoring of drug occupancy on the cells of interest during PK and PD experiments. Ex-vivo spiking experiments using appropriate biological samples spiked at varied concentrations of the drug were used to verify operational characteristics of the antibody cocktails and proper drug saturation calculations based on anti-Total and anti-Free-Site antibody measurements.
  • The antibodies, target and drugs validated to date for use in the methods of the inventions include:
    TABLE 2
    Exemplary Target-Drug Combinations and Antibody Pairs
    Anti-Total Anti-Free Site
    Target Drug Antibody Vendor (Cat. #) Antibody Vendor (Cat. #)
    CD11b Neutrophil anti-human Beckman Coulter anti-human BD Biosciences
    inhibitory factor CD11b-FITC (IM0530) CD11b-PE (30455S)
    CD11a chimeric anti-human BD Biosciences anti-human Dako-Cytomation
    humanized CD11a-PE (555384) CD11a-FITC (F0712)
    anti-CD11a
    CD18 Proprietary anti-human BD Biosciences anti-human Dako-Cytomation
    CD18-PE (555924) CD18-FITC (F0839)
    CD22 Proprietary anti-human Serotec anti-human Immunotech
    CD22-FITC (MCA553F) CD22-PE (IM1835)
    CD25 Proprietary anti-human BD Biosciences anti-human BD Biosciences
    CD25-PE (555432) CD25-PE (341009)
  • EXAMPLE 2 Additional Antibody Pairs
  • Additional antibody pairs that can be used in the general method of the invention for particular target and drug combinations can be identified using the method described generally in Example 1. Some possible combinations are listed in Table 3. However, the combinations are unlimited and additional combinations can identified by searching MEDLINE, ATCC or the web.
    TABLE 3
    Additional Target-Drug Combinations and Antibody Pairs
    Target Drug Antibodies
    CD20 Rituximab, PRO70769, Rituximab, Bexxar
    Bexxar (Tositumomab), B1, Immu-106,
    Ibritumomab, HI47, L27
    CD52 Campath-1H Campath-1H
    (Alemtuzumab)
    CD33 In Development HB-10306, Mab 251, M195,
    huM195, PC251, L4F3
    CD4 In Development OKT4, TNX-355, HuMax-CD4,
    GK1.5, W3/25, YTS177.9,
    ORTHOCLONE OKTcdr4a
    CD3 In Development HB231, HB10166, UCHT1, PS1,
    OKT3,
    CD14 In Development HB-247, HB-246, UCHM-1, IC14,
    M5E2, MφP9
    CD30 In Development cAC10-vcMMAE, SGN-30, Ki-1,
    K0145-3, HSR-4, BER-H2
    HLA-Dr Hu1D10 Hu1D10, L243, BRA30, 1DO9C3,
    TNB-211, 1D10, LN3,
    MAP Kinase In Development SC-154
    (e.g.,
    MAPK1-18)
    RAF-1 In Development Ab-2
    AKT In Development SKB-1
    BCL-2 In Development YTH-10C4, 124, 10C4
    Chemokine Sch-417690/Sch- S3504 (CCR1), 48607 (CCR2),
    Receptors D, GSK-873, 140 83103 (CCR3), Ig1 (CCR4),
    (e.g. CCR1 UK-427, 857 140706 (CCR6), 11A9 (CCR6),
    to CCR7) (CCR5) 3D12 (CCR7), 112509 (CCR9)
    ESRA fulvestrant 6F11, ER1D5, AER311, ER88
  • EXAMPLE 3 Flow Cytometry
  • Although the invention has been exemplified with respect to several antibody pair-drug combinations, as indicated in Table 2, only the data from a single study (CD11b) are presented herein for simplicity. These results are representative, although the details for each study will vary. The protocols described are also exemplary, but cell harvesting, antibody staining, fixation, and gating parameters should be (and were) optimized for each experiment.
  • General Protocol: For analysis of whole blood by flow cytometry, the following protocol was employed: 100 μl of whole blood was sterilely collected for each data point. For each sample, 2 ml of standard culture media plus or minus drug was added to the blood and the tubes vortexed briefly to mix. The samples were then incubated to allow drug binding for 1 hr at 37° C.
  • Next the cells were stained with the appropriate antibody or antibodies. A saturating amount (as determined in the titration evaluation of each antibody against the target) of desired antibody was added to each tube, and the tubes were incubated at 37° C. for 1 hr. Immediately after incubation, the cells were collected by centrifugation, and erythrocytes were lysed using a standard ammonium chloride lysing procedure.
  • The cells were collected by centrifugation, and 2 ml of cold PBS+2% FCS was added to each tube. The tubes were placed in the dark at room temperature for at least 10 minutes and up to 1 hour to allow RBC lysis upon resuspension of the fixed whole blood samples in this isotonic solution. The cells were collected again and washed once or twice with PBS+2% FCS with a final resuspension in 1% paraformaldehyde in PBS. The samples were thus ready for data collection.
  • We have used the BECKMAN COULTER™ Epics XL or BECTON DICKINSON™ FACS Calibur Flow cytometer herein, but any properly quality controlled flow cytometer that satisfies established windows of analysis can be used, provided the laser excitation lines and filter configurations are correct for the excitation and detection of all fluorescence labels used in the assay. The analysis that is actually performed will vary depending on the experiment being performed, which of the cellular parameters are of interest, and the actual labels employed. In general, we collect 30,000 ungated events. Data was analyzed in WinList (VERITY SOFTWARE™) and Excel (MICROSOFT™) or equivalent. Exemplary results for the CD11b/Neutrophil Inhibitory Factor experiments are shown in FIG. 1 through 6.
  • Separation of Monocytes and Neutrophils: In the first experiment, whole blood was stained with anti-CD14-APC, the cells were fixed, the RBCs lysed, and the remaining nucleated cells analyzed by cytometer. FIG. 1A shows light scatter patterns of the nucleated cells (leukocytes) stained with anti-CD14-APC. In FIG. 1B, an electronic gating window was constructed around the nucleated cells to separate the cells based on the expression of the CD14 marker. The APC signal was used to separate the leukocytes into CD 14 positive cells (monocytes) and CD14 negative (neutrophils).
  • Additional electronic gating windows can be applied to these two cell populations for simultaneous and independent analysis of the binding of the drug sensitive antibody (anti-Free-Site antibody) and the drug insensitive antibody (anti-Total) on each cell type respectively. This type of analysis is shown in FIG. 2.
  • Antibody Pair Binding: As above, whole blood was incubated with or without drug, stained with the appropriate antibodies, fixed, lysed and analyzed. This particular experiment was designed so that drug insensitive antibody (anti-Total) was labeled with FITC (αAgX-FITC) and drug sensitive antibody (anti-Free-Site) was labeled with PE (αAgX-PE). However, other labels could be used. Blood was treated with three different concentrations of drug and only the neutrophil results are shown in FIG. 2.
  • The histograms in FIG. 2 show fluorescence intensity at ˜525 nm (left panel, FITC signal) and ˜575 nm (right panel, PE signal) on the x-axis plotted against the number of cells counted (Number) on the y-axis. As the drug level increases, the αAgX-PE peak shifts to the left indicating a decrease in fluorescence intensity. Thus, anti-Free-Site antibody binding decreases due to the competition by the drug for the drug sensitive epitope. In contrast, the fluorescence intensity of the anti-AgX-FITC stain was invariant at all drug concentrations, indicating constant binding of this antibody to the drug insensitive epitope.
  • Target-Drug Binding: In the next two experiments, broader ranges of drug concentrations were investigated. Whole blood was incubated with various concentrations of drug ranging from 0-200 ng/ml, followed by antibody staining, fixing, lysis and analysis as described above. The data in FIG. 3 demonstrates constant binding of drug insensitive (anti-Total) antibody over a broad range of drug concentration. In contrast, the data in FIG. 4 demonstrates a dose dependent inhibition of the binding of the drug sensitive anti-AgX-PE (anti-Free-Site) to neutrophils.
  • The data from FIGS. 3 and 4 were used to calculate percent saturation as shown in FIG. 5. Percent Saturation was calculated as the anti-AgX-FITC signal minus the anti-Ag X-PE signal divided by the anti-AgX-FITC signal times 100. The data demonstrates that the drug causes a dose dependent inhibition of drug sensitive antibody binding to neutrophils.
  • Standard Curve: A target cell line (HL-60) was exposed to different drug concentrations ranging from 0 to 150 ng/ml. Subsequent to drug incubation, the cells were stained with drug sensitive anti-AgX and binding of the fluorescent antibody was quantified on the cytometer as described above and used to prepare this standard curve. Note: This curve was performed using monoclonal antibody binding intensity (MESF). However, the same curve may be constructed using monoclonal ABC.
  • Unknown concentrations of drug in patient specimens can be derived by extrapolation of quantitative fluorescence of the anti-AgX antibody to the respective drug concentration on this standard curve.
  • EXAMPLE 4 Intracellular Targets
  • Although the invention has been thus far exemplified with surface targets, such as CD11 b, it is also possible to study internal targets using the methods of the invention because fixatives are now available that allow the detection of internal epitopes. Cells are permeabilized with agents such as 0.05% Triton X-100 in PBS, stained with antibody and then fixed. For example, PermeaFix™ or PermiFlow™ at mild denaturation temperatures preserves cell morphology and thus allows subsequent analysis by flow cytometry. Such methods are described in more detail in co-pending application 60/512,834 filed Oct. 20, 2003.
  • Acceptable fixatives are defined herein as fixatives that allow both cell fixation and permeation, while retaining cell surface morphology and DNA and RNA content, sufficient to allow separation of cells based on light scatter, surface epitopes and/or nucleic acid content. An acceptable fixative contains 0.75-0.85% formaldehyde, 25-30 mM DNBS, 6.8-7% DMSO and 0.08-0.1% Tween 20 detergent. A preferred fixative is PermiFlow.™ Mild denaturing temperatures are those temperature that improve access to internal antigens, without compromising cell morphology, surface antigens or nucleic acid content. Preferred temperatures range from 39 to 43° C., and most preferred 43° C.

Claims (13)

1) a method of measuring drug activity in whole cells, comprising:
a) reacting whole cells with a labeled anti-Total antibody and with a labeled anti-Free-Site antibody;
b) fixing said cells;
c) simultaneously quantifying both anti-Total antibody and anti-Free-Site antibody by flow cytometry,
i) wherein the anti-Total antibody detects total target level and the anti-Free-Site antibody detects drug-free target level; and
ii) wherein the level of drug-free target is proportional to the level of anti-total antibody staining minus the anti-Free-Site antibody staining.
2) The method of claim (1), wherein the whole cells are a mixed population of cells and the cells are further typed by staining with a third antibody specific for a particular cell type.
3) The method of claim (1), wherein the target is on the cell surface.
4) The method of claim (1), where the target is inside the cell and the fixation is sufficient to preserve cell morphology.
5) The method of claim (4), wherein the fixative comprises the ingredients 0.75-0.85% formaldehyde, 25-30 mM DNBS, 6.8-7% DMSO and 0.08-0.1% Tween 20 detergent and fixation is performed at about 43° C.
6) The method of claim (1), wherein the anti-Total antibody and anti-Free-Site antibody are selected from the group consisting of those antibody pairs shown in Table 2.
7) A method of assessing drug kinetics in a subject, comprising:
a) administering a drug to a subject, wherein the drug binds to a target;
b) collecting whole cells from the subject;
c) incubating the whole cells with a labeled anti-Total antibody and with a labeled anti-Free-Site antibody;
d) fixing said cells;
e) simultaneously quantifying both anti-Total antibody and anti-Free-Site antibody by cytometry,
i) wherein the anti-Total antibody detects total target and the anti-Free-Site antibody detects drug-free target; and
ii) wherein drug-free target is proportional to anti-total antibody minus anti-Free-Site antibody.
8) The method of claim (7), wherein the whole cells are a mixed population of cells and the cells are further typed by staining with a third antibody specific for a particular cell type and sorted according to cell type before quantifying anti-Total antibody and anti-Free-Site antibody.
9) The method of claim (7), wherein the target is on the cell surface.
10) The method of claim (7), where the target is inside the cell and the fixation is sufficient to preserve cell morphology.
11) The method of claim (10), wherein the fixative comprises the ingredients 0.75-0.85% formaldehyde, 25-30 mM DNBS, 6.8-7% DMSO and 0.08-0.1% Tween 20 detergent and fixation is performed at about 43° C.
12) The method of claim (7), wherein the anti-Total antibody and anti-Free-Site antibody are selected from the group consisting of those antibody pairs shown in Table 2.
13) A method of measuring circulating free drug in a patient, comprising:
a) collecting blood from a patient, taking a drug, and purifying serum or plasma from the blood;
b) reacting the serum or plasma with a homogeneous population of cells which express the drug target antigen;
c) reacting the target cell with a labeled anti-Free-Site antibody,
wherein the anti-Free-Site antibody detects drug-free target, but is competitively inhibited by drug-target binding;
d) quantifying anti-Free-Site antibody by flow cytometry;
e) comparing the result in step d) against a standard curve wherein the level of free drug in serum is inversely proportional to the level of anti-free-site antibody staining,
wherein the standard curve is prepared by reacting a cell with known amounts of said drug, reacting said cells with labeled anti-Free-Site antibody; fixing said cells and quantifying anti-Free-Site antibody by flow cytometry, and preparing said standard curve from the results.
US11/136,168 2004-05-24 2005-05-24 Simultaneous assay of target and target-drug binding Abandoned US20050266503A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US11/136,168 US20050266503A1 (en) 2004-05-24 2005-05-24 Simultaneous assay of target and target-drug binding
US11/927,223 US7943327B2 (en) 2004-05-24 2007-10-29 Simultaneous assay of target and target-drug binding
US13/088,841 US8389224B2 (en) 2004-05-24 2011-04-18 Assay for analysis of free drug in samples
US13/759,158 US8828671B2 (en) 2004-05-24 2013-02-05 Simultaneous assay of target and target-drug binding
US14/452,772 US20140357522A1 (en) 2004-05-24 2014-08-06 Simultaneous Assay of Target and Target-Drug Binding
US15/007,342 US20160161514A1 (en) 2004-05-24 2016-01-27 Simultaneous Assay of Target and Target-Drug Binding

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US57378304P 2004-05-24 2004-05-24
US11/136,168 US20050266503A1 (en) 2004-05-24 2005-05-24 Simultaneous assay of target and target-drug binding

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US11/927,223 Division US7943327B2 (en) 2004-05-24 2007-10-29 Simultaneous assay of target and target-drug binding
US92723307A Division 2004-05-24 2007-10-29

Publications (1)

Publication Number Publication Date
US20050266503A1 true US20050266503A1 (en) 2005-12-01

Family

ID=35425827

Family Applications (6)

Application Number Title Priority Date Filing Date
US11/136,168 Abandoned US20050266503A1 (en) 2004-05-24 2005-05-24 Simultaneous assay of target and target-drug binding
US11/927,223 Expired - Fee Related US7943327B2 (en) 2004-05-24 2007-10-29 Simultaneous assay of target and target-drug binding
US13/088,841 Expired - Fee Related US8389224B2 (en) 2004-05-24 2011-04-18 Assay for analysis of free drug in samples
US13/759,158 Active US8828671B2 (en) 2004-05-24 2013-02-05 Simultaneous assay of target and target-drug binding
US14/452,772 Abandoned US20140357522A1 (en) 2004-05-24 2014-08-06 Simultaneous Assay of Target and Target-Drug Binding
US15/007,342 Abandoned US20160161514A1 (en) 2004-05-24 2016-01-27 Simultaneous Assay of Target and Target-Drug Binding

Family Applications After (5)

Application Number Title Priority Date Filing Date
US11/927,223 Expired - Fee Related US7943327B2 (en) 2004-05-24 2007-10-29 Simultaneous assay of target and target-drug binding
US13/088,841 Expired - Fee Related US8389224B2 (en) 2004-05-24 2011-04-18 Assay for analysis of free drug in samples
US13/759,158 Active US8828671B2 (en) 2004-05-24 2013-02-05 Simultaneous assay of target and target-drug binding
US14/452,772 Abandoned US20140357522A1 (en) 2004-05-24 2014-08-06 Simultaneous Assay of Target and Target-Drug Binding
US15/007,342 Abandoned US20160161514A1 (en) 2004-05-24 2016-01-27 Simultaneous Assay of Target and Target-Drug Binding

Country Status (1)

Country Link
US (6) US20050266503A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100304998A1 (en) * 2009-06-02 2010-12-02 Marquette University Chemical Proteomic Assay for Optimizing Drug Binding to Target Proteins
EP2607899A1 (en) * 2011-12-21 2013-06-26 Beckman Coulter, Inc. Method for labeling intracellular and extracellular targets of leukocytes
CN116297120A (en) * 2023-03-30 2023-06-23 深圳市血液中心(深圳市输血医学研究所) Method for detecting drug antibody in sample

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050266503A1 (en) * 2004-05-24 2005-12-01 Esoterix, Inc. Simultaneous assay of target and target-drug binding
US9535065B2 (en) * 2011-09-06 2017-01-03 Becton, Dickinson And Company Methods and compositions for cytometric detection of rare target cells in a sample
RU2015120052A (en) * 2015-05-28 2016-12-20 Аджиномото Ко., Инк. A method for producing an L-amino acid using a bacterium of the Enterobacteriaceae family in which gshA gene expression is weakened

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5597688A (en) * 1992-03-27 1997-01-28 Ortho Diagnostic Systems Inc. Cell fixative and method of analyzing virally infected cells
US5599677A (en) * 1993-12-29 1997-02-04 Abbott Laboratories Immunoassays for prostate specific antigen
US6579687B1 (en) * 1995-04-18 2003-06-17 Biosite Incorporated Methods for the assay of troponin I and T complexes of troponin I and T and selection of antibodies for use in immunoassays
US20030133936A1 (en) * 2001-07-12 2003-07-17 Byrne Michael Chapman CD25markers and uses thereof

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US103075A (en) * 1870-05-17 Improvement in saw-filing instrument
US28221A (en) * 1860-05-08 Improvement in hose-couplings
US133075A (en) * 1872-11-19 Improvement in sewing-machine tables
US47441A (en) * 1865-04-25 Improvement in oil-presses
WO1996041476A1 (en) 1995-06-07 1996-12-19 Imagetel International, Inc. Videoconferencing and multimedia system
US20010047441A1 (en) 2000-02-23 2001-11-29 Mark Robertson Communications system conduit for transferring data
US20030103075A1 (en) 2001-12-03 2003-06-05 Rosselot Robert Charles System and method for control of conference facilities and equipment
US7995090B2 (en) 2003-07-28 2011-08-09 Fuji Xerox Co., Ltd. Video enabled tele-presence control host
US7326577B2 (en) * 2003-10-20 2008-02-05 Esoterix, Inc. Cell fixation and use in phospho-proteome screening
US20050266503A1 (en) 2004-05-24 2005-12-01 Esoterix, Inc. Simultaneous assay of target and target-drug binding

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5597688A (en) * 1992-03-27 1997-01-28 Ortho Diagnostic Systems Inc. Cell fixative and method of analyzing virally infected cells
US5599677A (en) * 1993-12-29 1997-02-04 Abbott Laboratories Immunoassays for prostate specific antigen
US6579687B1 (en) * 1995-04-18 2003-06-17 Biosite Incorporated Methods for the assay of troponin I and T complexes of troponin I and T and selection of antibodies for use in immunoassays
US20030133936A1 (en) * 2001-07-12 2003-07-17 Byrne Michael Chapman CD25markers and uses thereof

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100304998A1 (en) * 2009-06-02 2010-12-02 Marquette University Chemical Proteomic Assay for Optimizing Drug Binding to Target Proteins
WO2010141596A2 (en) * 2009-06-02 2010-12-09 Marquette University Chemical proteomic assay for optimizing drug binding to target proteins
WO2010141596A3 (en) * 2009-06-02 2011-02-24 Marquette University Chemical proteomic assay for optimizing drug binding to target proteins
EP2607899A1 (en) * 2011-12-21 2013-06-26 Beckman Coulter, Inc. Method for labeling intracellular and extracellular targets of leukocytes
WO2013093642A1 (en) * 2011-12-21 2013-06-27 Beckman Coulter, Inc. Method for labeling intracellular and extracellular targets of leukocytes
CN104011542A (en) * 2011-12-21 2014-08-27 贝克曼考尔特公司 Method for labeling intracellular and extracellular targets of leukocytes
US20150010923A1 (en) * 2011-12-21 2015-01-08 Beckman Coulter, Inc. Method for labeling intracellular and extracellular targets of leukocytes
EP3104177A1 (en) * 2011-12-21 2016-12-14 Beckman Coulter, Inc. Method for labeling intracellular and extracellular targets of leukocytes
US9678073B2 (en) * 2011-12-21 2017-06-13 Beckman Coulter, Inc. Method for labeling intracellular and extracellular targets of leukocytes
US11635433B2 (en) 2011-12-21 2023-04-25 Beckman Coulter, Inc. Method for labeling intracellular and extracellular targets of leukocytes
CN116297120A (en) * 2023-03-30 2023-06-23 深圳市血液中心(深圳市输血医学研究所) Method for detecting drug antibody in sample

Also Published As

Publication number Publication date
US20130177926A1 (en) 2013-07-11
US20140357522A1 (en) 2014-12-04
US8828671B2 (en) 2014-09-09
US20110212469A1 (en) 2011-09-01
US20080096231A1 (en) 2008-04-24
US8389224B2 (en) 2013-03-05
US7943327B2 (en) 2011-05-17
US20160161514A1 (en) 2016-06-09

Similar Documents

Publication Publication Date Title
US20160161514A1 (en) Simultaneous Assay of Target and Target-Drug Binding
US8137975B2 (en) Method for a rapid antibody-based analysis of platelet populations
CN101027557B (en) Whole blood preparation for cytometric analysis of cell signaling pathways
Illingworth et al. ICCS/ESCCA consensus guidelines to detect GPI‐deficient cells in paroxysmal nocturnal hemoglobinuria (PNH) and related disorders part 3–data analysis, reporting and case studies
US7625712B2 (en) Method for a fully automated monoclonal antibody-based extended differential
US6977156B2 (en) Flow cytometry reagent and system
CN101023353A (en) Methods and compositions for risk stratification
EP1664719A2 (en) Methods for substantially simultaneous evaluation of a sample containing a cellular target and a soluble analyte
Cheng et al. Monitoring anti‐CD19 chimeric antigen receptor T cell population by flow cytometry and its consistency with digital droplet polymerase chain reaction
Spurgeon et al. Immunophenotypic analysis of platelets by flow cytometry
Salvagno et al. Evaluation of platelet turnover by flow cytometry
US20060024744A1 (en) Methods for substantially simultaneous evaluation of a sample containing a cellular target and a soluble analyte
US20020004212A1 (en) Detection of surface-associated human leukocyte elastase
US6835551B2 (en) Basophil degranulation test
US6696243B2 (en) Method for the analysis of soluble analytes
EP0170345B1 (en) Flow cytometry
Szerémy et al. Determination of reference values of MDR‐ABC transporter activities in CD3+ lymphocytes of healthy volunteers using a flow cytometry based method
WO2020014175A1 (en) Methods and compositions for analyzing immortalized megakaryocyte progenitor cell lines and platelet-like particles derived therefrom
Smith et al. Cytometric analysis of BAL T cells labeled with a standardized antibody cocktail correlates with immunohistochemical staining
US20070048802A1 (en) Method and kit for detection of autoimmune chronic urticaria
Park et al. MRP1 and P-glycoprotein expression assays would be useful in the additional detection of treatment non-responders in CML patients without ABL1 mutation
Wong et al. Monochromatic gating method by flow cytometry for high purity monocyte analysis
WO2000052472A1 (en) Rapid assay for infection in small children
US6667149B1 (en) Procedure for the analysis of the functional activation of leukocytes, platelets and other cells, induced in vivo or in vitro, based on the stabilization of cytoplasmic membrane proteins and its detection using quantitative cytometric methods in the absence of additional sample manipulation
EP2506011A2 (en) Method and kit for detecting and quantifying intracellular histamine and histamine release

Legal Events

Date Code Title Description
AS Assignment

Owner name: ESOTERIX, INC., TEXAS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PURVIS, NORMAN B.,;STELZER, GREGORY T.;REEL/FRAME:016578/0786

Effective date: 20050726

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION