EP0946877A1 - P.seketinassays und verfahren zu deren herstellung - Google Patents

P.seketinassays und verfahren zu deren herstellung

Info

Publication number
EP0946877A1
EP0946877A1 EP97950589A EP97950589A EP0946877A1 EP 0946877 A1 EP0946877 A1 EP 0946877A1 EP 97950589 A EP97950589 A EP 97950589A EP 97950589 A EP97950589 A EP 97950589A EP 0946877 A1 EP0946877 A1 EP 0946877A1
Authority
EP
European Patent Office
Prior art keywords
selectin
platelet
sample
soluble
level
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.)
Withdrawn
Application number
EP97950589A
Other languages
English (en)
French (fr)
Inventor
Margaret R. Dalesandro
Bart Frederick
Cindy I. Gumbs
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.)
Janssen Biotech Inc
Original Assignee
Centocor 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 Centocor Inc filed Critical Centocor Inc
Publication of EP0946877A1 publication Critical patent/EP0946877A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • BACKGROUND Platelets are recognized as playing a key role in arterial thrombosis and in acute ischemic coronary syndromes (Gawaz, M. et al . , "Platelet function in acute myocardial infarction tested with direct angioplasty, " Circulation, 93 : 229-237 (1996); Trip, M.D. et al . , "Platelet hyperreactivity and prognosis in survivors of myocardial infarction," N. Engl . J. Med . , 322 : 1549-1554 (1990); Hirsh, J. , “Hyperreactive platelets and complications of coronary artery disease," N. Engl . J. Med .
  • Platelets become activated through numerous stimuli including thrombin, subendothelial interactions, contact with artificial surfaces, and in the presence of some immune complexes (Bellon, J.L. et al . ,
  • platelets Once activated, platelets expose the fibrinogen binding sites on the membrane glycoprotein GPIIb/IIIa complex and platelet aggregation takes place via fibrinogen bridging (McEver, R.P., "The clinical significance of platelet membrane glycoproteins, " Hematol . Oncol . Clin . North Am . , 4 : 87-103 (1990); Du, X. et al . , "Ligands "activate” integrin IIb ⁇ 3 (Platelet GPIIb-IIIa) , " Cell, 65 : 409-416 (1991)).
  • P-selectin also known as CD62, GMP-140, or PADGEM
  • CD62 also known as CD62, GMP-140, or PADGEM
  • P-selectin is a member of the selectin family of adhesion receptors that regulates leukocyte trafficking (Lawrence, M.B. and T.A. Springer, Cell , 65 : 859 (1991); Johnston, G.I. et al . , Cell , 56 : 1033-1044 (1989); McEver, R.P., U.S. Patent No. 5,378,464).
  • P-selectin is an integral membrane glycoprotein found in the granules of unactivated platelets and in the Weibel-Palade bodies of endothelial cells (Peerschke, E.I.B., Am. J. Clin .
  • a platelet alpha granule membrane protein (GMP-140) is expressed on the plasma membrane after activation," J “ . Cell Biol . , 101 : 880-886 (1985)).
  • P-selectin expression on platelets can be increased up to 50-fold, mediating platelet-leukocyte (monocytes, neutrophils) adhesive interactions and endothelial cell-leukocyte adhesive interactions leading to thrombus formation (Wen, D. et al . , J . Lab . Clin . Med . , 124 : 447 (1994); Larsen, E. et al .
  • PADGEM protein a receptor that mediates the interaction of activated platelets with neutrophils and monocytes
  • the ⁇ granule membranes fuse with those of the surface connected canalicular system, and glycoproteins such as P-selectin diffuse out onto the surface where they can be detected with specific antibodies (Nurden, A.T. et al . , Nouv . Rev. Fr . Hematol . , 35 : 67 (1993)).
  • P-selectin is one of three structurally related membrane glycoproteins, including P-, E- and L-selectin, that initiate leukocyte adhesion to vascular endothelium and platelets in response to inflammatory stimuli.
  • Each selectin molecule contains an NH 2 -terminal carbohydrate- recognition domain characteristic of C-type lectins, followed by an EGF-like motif, consensus repeats like those in complement-regulatory proteins, a transmembrane domain, and a cytoplasmic tail (Ushiyama, S. et al . , J. Biol . Chem . , 268 : 15229 (1993)).
  • each of these domains is encoded by separate exons.
  • Cloning data from an endothelial cell library showed evidence of three separate forms of P-selectin, two of which differed in the number of complement regulatory protein repeats, while a third form lacked a transmembrane domain and was predicted to be soluble (Dunlop, L.C. et al . , "Characterization of GMP-140 as a circulating plasma protein", J. Exp . Med . , 175 : 1147- 1150 (1992)).
  • Human platelets have subsequently been found to contain approximately equal amounts of mRNA encoding P-selectin with and without the transmembrane domain (Johnston, G.I. et al . , J. Biol . Chem .
  • the soluble form of P-selectin has been isolated from the plasma of normal donors.
  • the purified protein had a molecular mass (nonreduced) nearly identical to that of platelet membrane P-selectin ( ⁇ 3kD lower, reduced) , and was immunoblotted by polyclonal and monoclonal anti-P-selectin antibodies.
  • Analytical gel filtration studies indicated that the plasma P-selectin eluted as a monomer lacking a transmembrane domain.
  • the present invention relates to a method of assessing the effect of "anti-platelet” agents, which affect the activation state of platelets, by monitoring levels of P-selectin in a sample from an individual.
  • levels of P-selectin are monitored in a sample containing platelets (e.g., a blood sample, platelet rich plasma (PRP) ) using an assay for membrane bound P-selectin.
  • levels of P-selectin are measured in a sample from which platelets have been removed (e.g., platelet poor plasma (PPP) , serum) , using an assay for soluble P-selectin.
  • P-selectin is detected immunologically by means of an anti-P-selectin antibody (i.e., one or more antibodies), such as monoclonal antibodies including S12, W40, GI and VH10, or an antibody having a similar epitopic specificity.
  • an anti-P-selectin antibody i.e., one or more antibodies
  • membrane bound P-selectin is determined using a non- fluorescent immunobinding assay, and even more preferably, soluble P-selectin is determined using an ELISA assay.
  • soluble P-selectin is determined using a sandwich ELISA. It is also particularly preferred to determine both membrane bound and soluble P- selectin, sometimes essentially simultaneously, to establish a P-selectin profile.
  • the method is useful for monitoring the effect of anti-platelet therapy on platelet activation as reflected by membrane bound and/or soluble P-selectin levels.
  • Treatment can be adjusted accordingly to maintain the desired level of platelet activation.
  • the basal activation state of platelets in normal individuals or in a patient can be assessed by measuring membrane bound and/or soluble P-selectin levels, and therapy can be adjusted in order to achieve such a basal activation state, or other desired platelet activation state.
  • percutaneous transluminal coronary angioplasty led to an elevation of soluble P- selectin levels.
  • the invention relates to a method of assessing the effect of a (i.e., one or more) vascular intervention procedure (e.g., cardiovascular intervention, coronary intervention) , and of a coronary artery intervention procedure in particular, on platelet activation state, particularly in patients having coronary disease, comprising determining the level of soluble P- selectin in a sample.
  • a vascular intervention procedure e.g., cardiovascular intervention, coronary intervention
  • a coronary artery intervention procedure in particular, on platelet activation state, particularly in patients having coronary disease
  • angiography angioplasty
  • angioplasty e.g., performed by balloon, coronary atherectomy, laser angioplasty or other suitable methods (with or without rotablation and/or stent placement)
  • coronary artery by-pass surgery e.g., coronary stent
  • stent placement e.g., coronary stent
  • other vascular intervention procedures e.g., vascular surgery, vascular graft, deployment of a peripheral stent, insertion of a prosthetic valve or vessel (e.g., in autologous, non-autologous or synthetic vessel graft)
  • anti-platelet therapy can be maintained or adjusted in order to achieve a basal activation state, or other desired endogenous platelet activation state.
  • platelet activation state as indicated by levels of soluble P-selectin can be assessed, and anti-platelet therapy can be initiated or adjusted to counteract platelet activation.
  • FIG. 1 is a schematic depiction illustrating a radioimmunoassay useful for the determination of membrane bound P-selectin.
  • ACD Acid citrate dextrose
  • PGE prostaglandin E ⁇
  • PRP platelet-rich plasma
  • PPP platelet poor plasma
  • PMA phorbol myristate acetate
  • 15 I-S12 IgG 125 I-labeled anti-P-selectin monoclonal antibody (Mab) S12.
  • FIG 2 is a schematic depiction illustrating the steps of an enzyme-linked immunosorbent assay (EIA or ELISA) useful for the determination of soluble P-selectin.
  • EIA enzyme-linked immunosorbent assay
  • PPP platelet poor plasma
  • HRP horseradish peroxidase
  • OPD HRP substrate O-phenylenediamine dihydrochloride.
  • Other abbreviations are as in Figure 1.
  • Figure 3 is a standard curve generated using the soluble P-selectin ELISA described in Example 2 with increasing concentrations (3.2 ng/ml to 320 ng/ml) of recombinantly produced truncated P-selectin purified from tissue culture supernatant of human 293 transfectants.
  • the assay format used a W40-coated microtiter plate. The standard was added to the plate simultaneously with biotinylated S12 antibody and streptavidin-HRP, and incubated for 2 hours. Color development in the presence of OPD was stopped after 20 minutes with 4N H 2 S0 4 . A correlation coefficient of 0.996 or better was achieved. A log-log fit was chosen as best fit for the data. Inter- and intra-assay precision for the assay is CV ⁇ 10%.
  • Figures 4A-4B are graphs which depict the dose- dependent increase in platelet membrane P-selectin expression determined using a radioimmunoassay (RIA,
  • FIG. 4A shows the dose-dependent increase in the binding of 125 I-S12 to platelets activated by PMA ranging from 5 - 500 nM final concentration.
  • Figure 4B depicts the activation indices for platelet P-selectin expression for a dose titration of PMA. The activation indices are the ratio between the endogenous P-selectin expressed and the P-selectin that could be expressed under conditions designed to stimulate expression of all available P-selectin.
  • Figure 5 is a graph depicting the activation indices for the platelets of two donors which were activated by a titration of PMA in the presence or absence of ReoPro® (final concentration 5 ⁇ g/ml) . For both donors the Activation indices were lower in the presence of ReoPro over the range of the PMA titration, indicating that ReoPro unexpectedly decreased platelet activation as measured by P-selectin expression.
  • Figure 6 is a bar graph illustrating the mean production and detection of soluble P-selectin by ELISA (Example 2) in platelet poor plasma (PPP) prepared from whole blood samples incubated with PMA (0 nM, 20 nM, 100 nM, or 500 nM PMA) in the presence of 5 ⁇ g/ml ReoProTM (filled bars) or in the absence of ReoProTM (clear bars) .
  • the production was dose responsive with respect to PMA and unexpectedly, ReoPro decreased the soluble P-selectin produced by activated platelets over the 24 hour time course of the experiment.
  • This graph depicts whole blood from 3 donors stimulated with various concentrations of PMA.
  • Figure 7 is a schematic depiction illustrating the preparation of a sample for analysis with a flow cytometry assay, an assay useful for the determination of membrane bound P-selectin.
  • ACD Acid citrate dextrose
  • PGE prostaglandin E ⁇
  • PRP platelet-rich plasma
  • A/P apyrase plus prostaglandin E-, ;
  • MTB modified Tyrodes Buffer
  • FITC fluorescein isothiocyanate
  • PMA phorbol 12-myristate 13- acetate
  • S12-FITC FITC-labeled anti-P-selectin monoclonal antibody (Mab) S12.
  • Figure 8 is a schematic depiction illustrating the preparation of a sample for analysis with a volumetric capillary cytometry system useful for the determination of membrane bound P-selectin.
  • ACD Acid citrate dextrose
  • PGEi prostaglandin E
  • WB whole blood
  • A/P apyrase plus prostaglandin E ⁇
  • MTB modified Tyrodes Buffer
  • CD61-Cy5 Cy5-labeled Mab that binds a receptor found on essentially all platelets
  • S12/W40-Cy5 a mixture of equal parts of Cy5-labeled anti-P-selectin Mabs S12 and W40.
  • Figure 9 is a schematic depiction illustrating preparation of a sample for analysis with a volumetric capillary cytometry system useful for the determination of soluble P-selectin in plasma from a patient sample.
  • the following abbreviations are used in the figure which designate the various reagents utilized in the assay: ACD, acid citrate dextrose, A/P, apyrase plus prostaglandin E ⁇ PPP, platelet poor plasma, W40, P-selectin specific Mab used to coat 9.7 ⁇ M polystyrene particles; S12-Cy5, Cy5- labeled P-selectin specific Mab.
  • the present invention relates to a method of assessing the effect of "anti-platelet” agents, which affect the activation state of platelets, by monitoring levels of P-selectin in a sample from an individual.
  • levels of P-selectin are monitored in a sample containing platelets (e.g., a blood sample, platelet rich plasma (PRP) ) using an assay for membrane bound P-selectin.
  • levels of P-selectin are measured in a sample from which platelets have been removed (e.g., platelet poor plasma (PPP) , serum) , using an assay for soluble P-selectin.
  • PPP platelet poor plasma
  • P-selectin is detected immunologically by means of an anti- P-selectin antibody (i.e., one or more antibodies), such as monoclonal antibody S12 or monoclonal antibody W40.
  • an anti- P-selectin antibody i.e., one or more antibodies
  • monoclonal antibody S12 or monoclonal antibody W40 i.e., monoclonal antibody S12 or monoclonal antibody W40.
  • Mixtures of polyclonal and/or monoclonal antibodies can be used (e.g., a cocktail of murine W40, S12 and GI monoclonal antibodies).
  • a sample e.g., tissue and/or fluid
  • a suitable assay can be used to assess the level of P-selectin.
  • Suitable assays include immunological methods, such as FACS analysis, radioimmunoassay, enzyme-linked immunosorbent assays (ELISA) , including che iluminescence assays.
  • P-selectin includes P-selectin molecules such as mature protein (e.g., of platelet origin, of endothelial origin, membrane-bound, soluble) , polymorphic or allelic variants of P-selectin, and other isoforms (e.g., produced by alternative splicing or other cellular processes) , and modified or unmodified forms of the foregoing (e.g., glycosylated, unglycosylated) .
  • Antibodies reactive with P-selectin or portions thereof can be used in the method.
  • the antibodies specifically bind membrane bound and/or soluble P-selectin or a portion thereof (see e.g., Furie et al . , U.S. Patent No. 4,783,330, the teachings of which are incorporated herein by reference in their entirety) .
  • the antibodies can be polyclonal or monoclonal, and the term antibody is intended to encompass both polyclonal and monoclonal antibodies.
  • the terms polyclonal and monoclonal refer to the degree of homogeneity of an antibody preparation, and are not intended to be limited to particular methods of production.
  • Anti-P-selectin antibodies can be raised against an appropriate im unogen, such as isolated and/or recombinant P-selectin or portion thereof (including synthetic molecules, such as synthetic peptides) .
  • antibodies can be raised against an isolated and/or recombinant P-selectin or portion thereof (e.g., a peptide) or against a host cell which expresses recombinant P-selectin (Johnston, G.I. et al . , Cell, 56 : 1033-1044 (1989); and McEver, R.P., U.S. Patent No. 5,378,464, the teachings of which are both incorporated herein by reference in their entirety) .
  • cells expressing recombinant P-selectin can be used as immunogens or in a screen for antibody which binds receptor (See e.g., Chuntharapai et al . , J. Immunol . , 152 : 1783-1789 (1994); Chuntharapai et al . , U.S. Patent No. 5,440,021).
  • Preparation of immunizing antigen, and polyclonal and monoclonal antibody production can be performed using any suitable technique. A variety of methods have been described (see e.g., Kohler et al . , Nature, 256: 495-497 (1975) and Eur. J. Immunol .
  • a hybridoma is produced by fusing a suitable immortal cell line (e.g., a myeloma cell line such as SP2/0) with antibody producing cells.
  • a suitable immortal cell line e.g., a myeloma cell line such as SP2/0
  • the antibody producing cell preferably those of the spleen or lymph nodes, can be obtained from animals immunized with the antigen of interest.
  • the fused cells (hybridomas) can be isolated using selective culture conditions, and cloned by limiting dilution. Cells which produce antibodies with the desired specificity can be selected by a suitable assay (e.g., ELISA).
  • Suitable methods of producing or isolating antibodies of the requisite specificity can be used, including, for example, methods which select recombinant antibody from a library, or which rely upon immunization of transgenic animals (e.g., mice) capable of producing a full repertoire of human antibodies (see e.g., Jakobovits et al . , Proc . Natl . Acad . Sci . USA, 90 : 2551-2555 (1993); Jakobovits et al . , Nature, 362 : 255-258 (1993); Lonberg et al . , U.S. Patent No. 5,545,806; Surani et al . , U.S. Patent No. 5,545,807) .
  • Single chain antibodies and chimeric, humanized or primatized (CDR-grafted) , or veneered antibodies, as well as chimeric, CDR-grafted or veneered single chain antibodies, comprising portions derived from different species, and the like are also encompassed by the present invention and the term "antibody".
  • the various portions of these antibodies can be joined together chemically by conventional techniques, or can be prepared as a contiguous protein using genetic engineering techniques. For example, nucleic acids encoding a chimeric or humanized chain can be expressed to produce a contiguous protein. See, e.g., Cabilly et al . , U.S. Patent No. 4,816,567; Cabilly et al . , European Patent No.
  • functional fragments of antibodies including fragments of chimeric, humanized, primatized, veneered or single chain antibodies
  • functional fragments of foregoing antibodies include those which are reactive with P-selectin.
  • antibody fragments capable of binding to P-selectin or portion thereof, including, but not limited to, Fv, Fab, Fab' and F(ab') 2 fragments are encompassed by the invention.
  • Such fragments can be produced by enzymatic cleavage or by recombinant techniques. For instance, papain or pepsin cleavage can generate Fab or F(ab') 2 fragments, respectively.
  • Antibodies can also be produced in a variety of truncated forms using antibody genes in which one or more stop codons has been introduced upstream of the natural stop site.
  • a chimeric gene encoding a F(ab') 2 heavy chain portion can be designed to include DNA sequences encoding the CH : domain and hinge region of the heavy chain.
  • the antibody can be modified, for example, by incorporation of or attachment (directly or indirectly (e.g., via a linker)) of a detectable label such as a radioisotope, spin label, antigen (e.g., epitope label such as a FLAG tag) or enzyme label, flourescent or chemiluminescent group and the like, and such modified forms are included within the term "antibody” .
  • a detectable label such as a radioisotope, spin label, antigen (e.g., epitope label such as a FLAG tag) or enzyme label, flourescent or chemiluminescent group and the like.
  • dual assay or "P-selectin profile” means an assay capable of determining the levels of soluble and membrane bound P-selectin. In an assay measuring both levels, an elevated level of either can indicate the presence of platelet activation.
  • marker or “marker for platelet activation” refer to either the level of soluble or the level of membrane bound P-selectin or both.
  • a biological sample can be assayed for P-selectin, including membrane bound and/or soluble P-selectin, by combining the sample to be tested with an antibody having specificity for P-selectin, under conditions suitable for formation of a complex between antibody and P-selectin, and detecting or measuring (directly or indirectly) the formation of a complex.
  • the sample can be obtained directly or indirectly (e.g., provided by a healthcare provider) , and can be prepared by a method suitable for the particular sample (e.g., whole blood, platelet rich plasma, platelet poor plasma, serum) and assay format selected.
  • whole blood can be collected by a suitable method, such as by venipuncture into a container containing an anti-coagulant such as ACD-A, heparin, or EDTA, or from an in-dwelling arterial line into such a container.
  • a suitable method such as by venipuncture into a container containing an anti-coagulant such as ACD-A, heparin, or EDTA, or from an in-dwelling arterial line into such a container.
  • an anti-coagulant such as ACD-A, heparin, or EDTA
  • Methods of combining sample and antibody and methods of detecting complex formation are also selected to be compatible with the assay format.
  • Antibodies can be labeled with a suitable label which can be detected directly, such as radioactive, fluorescent or chemiluminescent labels, or indirectly, such as enzyme labels or other antigenic or specific binding partners (e.g., biotin) .
  • suitable labels include, for example, fluorescent labels (e.g., fluorescein, rhodamine) , chemiluminescent labels (e.g., luciferase), radioisotope labels (e.g, 32 P, 125 I, 131 I) , enzyme labels (e.g., horseradish peroxidase, alkaline phosphatase, ⁇ -galactosidase) , biotin, avidin, spin labels and the like.
  • fluorescent labels e.g., fluorescein, rhodamine
  • chemiluminescent labels e.g., luciferase
  • radioisotope labels e.g, 32 P, 125 I, 131 I
  • the detection of antibodies in a complex can also be done immunologically with a second antibody which is then detected (e.g., by means of a label).
  • Conventional methods or other suitable methods can be used to directly or indirectly label an antibody.
  • Assays for Detection of Platelet Membrane Bound P-selectin Methods known now or developed later can be used for measuring membrane bound P-selectin.
  • the present invention provides a method to determine the effects of anti-platelet therapy on the level of platelet activation by measuring the expression of platelet-bound
  • a sample comprising platelets can be contacted with an antibody having specificity for P-selectin under conditions suitable for formation of a complex between antibody and P-selectin expressed on said platelets, and detecting or measuring (directly or indirectly) the formation of a complex.
  • a radioisotope-conjugated immunobinding assay is used. Endogenous platelet activation can be measured as a percent of the total expressible P-selectin for a given sample.
  • Figure 1 illustrates one type of assay which can be used (see also Example 1) .
  • endogenous platelet activation can be assessed by an immunobinding assay comprising: (a) obtaining a first and second sample comprising platelets, wherein each sample contains a preselected number of platelets;
  • a platelet activation agonist such as phorbol myristate acetate (PMA) , ADP (adenosine diphosphate) , thrombin, collagen, and/or TRAP (thrombin receptor activating peptide)
  • PMA phorbol myristate acetate
  • ADP adenosine diphosphate
  • thrombin thrombin receptor activating peptide
  • TRAP thrombin receptor activating peptide
  • P-selectin antibody and activated platelets and activated platelets; and (d) determining (detecting or measuring) the formation of complex in said samples, wherein the amount of complex detected in said second sample as compared to that detected in said first sample is indicative of the extent of platelet activation in said second sample.
  • a ratio reflecting the amount of complex detected in said second sample to that detected in said first sample can provide a measure of the extent of platelet activation in said second sample.
  • formation of complex can be assessed by determining the radioactivity present in the labeled complexes in each sample, wherein a ratio of the radioactivity of said second sample to said first sample provides a measure of the extent of platelet activation in said second sample.
  • the first and second samples are from the same donor.
  • the first and second samples are collected at about the same time (e.g., obtained by dividing a sample from a donor, obtained from two samples collected in series) .
  • the assay can also be performed on whole blood without a pre-isolation step or standardization of platelet number, thus substantially reducing processing time.
  • a sample of whole blood can be obtained from a donor whose level of platelet activation is to be determined and can be divided into two portions.
  • One sample can be treated with a platelet agonist such as PMA to maximally activate platelets, while the other sample is not treated with activation agonists, but is maintained under conditions designed to maintain the endogenous (in vivo) activation level (e.g., by addition of activation inhibitors such as aprotinin, theophylline, apyrase and/or prostaglandin E ⁇ ) .
  • activation inhibitors such as aprotinin, theophylline, apyrase and/or prostaglandin E ⁇
  • Radioactively labeled anti-P-selectin antibody is added to both samples and samples are maintained under conditions suitable for specific binding to P-selectin, and preferably until binding is complete. The extent of binding is the assessed.
  • the samples can be processed to separate complexes from unbound anti-P-selectin antibody.
  • samples can be diluted 1:6 with a buffer that does not alter platelet activation state, such as Tyrode's Modified Buffer, layered over a 30% sucrose barrier (e.g., in preloaded microfuge tubes), and microfuged (e.g., for 4 minutes at 11,000 X g) .
  • the pellet with its bound radiolabeled anti-P-selectin antibody can be clipped and counted in a gamma counter.
  • the percent of radioactivity in the endogenously activated sample compared with the maximally activated sample can be calculated and described as the Activation Index (Al) for the sample. In this manner, endogenous platelet activation can be measured as percent of total expressible P-selectin.
  • the present invention further provides a radioisotope-conjugated immunobinding assay for measuring endogenous platelet activation in a sample of whole blood from a donor whose platelet activation is to be determined comprising:
  • a platelet activation agonist such as phorbol myristate acetate (PMA) under conditions suitable for activation of platelets in said sample, preferably for a period of time effective to maximally activate said platelets, and preferably while maintaining said second sample of whole blood under conditions suitable for maintaining the endogenous platelet activation level;
  • a platelet activation agonist such as phorbol myristate acetate (PMA)
  • composition comprising an anti-P-selectin antibody, such as (i) an anti-P-selectin antibody comprising a radioactive label; or (ii) an anti-P-selectin antibody comprising a binding site for a second antibody which comprises a radioactive label, preferably in excess of that required to bind the anti-P-selectin antibody, such as (i) an anti-P-selectin antibody comprising a radioactive label; or (ii) an anti-P-selectin antibody comprising a binding site for a second antibody which comprises a radioactive label, preferably in excess of that required to bind the
  • P-selectin expressed on the platelets under conditions suitable for the formation of labeled complexes between said anti-P-selectin antibody and activated platelets; and (d) determining (detecting or measuring) the formation of complex in said samples, wherein the amount of complex detected in said second sample as compared to that detected in said first sample is indicative of the extent of platelet activation in said second sample.
  • a ratio reflecting the amount of complex detected in said second sample to that detected in said first sample can provide a measure of the extent of platelet activation in said second sample.
  • Another method for assessing membrane bound P-selectin levels is flow cytometry. Methods of flow cytometry for measuring platelet or membrane bound P-selectin are known in the art. (Shattil, Sanford J, et al. "Detection of Activated Platelets in Whole Blood using Activation- Dependent Monoclonal Antibodies and Flow Cytometry," Blood, Vol. 70, No 1 (July), 1987: pp307-315; Scharf, Rudiger E.
  • a sample comprising platelets can be contacted with an antibody having specificity for P- selectin under conditions suitable for formation of a complex between an antibody and P-selectin expressed on platelets, and detecting or measuring (directly or indirectly) the formation of a complex.
  • the antibody, S-12 is conjugated with FITC.
  • Figure 7 illustrates one type of flow cytometry assay, (see also Example 9) .
  • the level of membrane bound P- selectin can be assessed by flow cytometry comprising: (a) obtaining a first and second sample comprising platelets,
  • a platelet activation agonist such as phorbol myristate acetate (PMA) , ADP (adenosine diphosphate) , thrombin, collagen, and/or TRAP (thrombin receptor activating peptide)
  • PMA phorbol myristate acetate
  • ADP adenosine diphosphate
  • thrombin thrombin
  • collagen thrombin
  • TRAP thrombin receptor activating peptide
  • determining (detecting or measuring) the formation of complex in said samples wherein the amount of complex detected indicates the extent of platelet activation in said second sample.
  • Another method of assaying levels of membrane bound P- selectin involves analysis with a volumetric capillary cytometry system.
  • An example of a volumetric capillary cytometry system is IMAGN2000TM from Biometric Imaging, Mountain View, CA.
  • membrane bound P-selectin is measured using a P- selectin specific antibody or mixture thereof.
  • the antibody is labeled with a fluorophore.
  • the antibodies used are a mixture or cocktail of S-12 and W-40 each of which are labeled with fluorophore, Cy5 (Amersham-Searle) .
  • the volumetric capillary cytometry system detects the number of events and the fluorescent intensity.
  • the level of membrane bound P-selectin can be assessed by volumetric capillary cytometry system comprising:
  • the total platelet count can be measured by contacting the sample with a fluorophore- labeled antibody specific to essentially all platelets, and then detecting the number of events or fluorescence.
  • the antibody is an antibody specific for a receptor existing on essentially all platelets, such as glycoprotein GP Ilb/IIIa, CD61, 10E5, CD41 and CD42. These antibodies can be labeled with a fluorophore, namely Cy5. (Amersham-Searle) .
  • a volumetric capillary cytometry system can assess the total platelet count in a method comprising: (a) obtaining a sample comprising platelets,
  • the present invention provides a method to determine the effects of anti-platelet therapy and/or vascular intervention (e.g., PTCA) on the level of platelet activation by determining the level of soluble P-selectin in the plasma of persons treated with anti-platelet therapeutic agents and/or vascular intervention.
  • Platelet activation state can be assessed before, during and/or after treatment, permitting detection of alterations in the patient's endogenous platelet activation state relative to a basal state at a selected time. Elevated platelet activation can also be assessed by comparing endogenous platelet activation level with that of a suitable control (e.g., normal individuals).
  • soluble P-selectin is determined using an ELISA assay, and in a particularly preferred embodiment a sandwich ELISA is used.
  • Figure 2 illustrates one type of assay which can be performed (see also Example 2) .
  • murine W40 is used as capture antibody and murine S12 is used as detector antibody.
  • a sample e.g., blood
  • platelets are removed (partially or completely) from the sample, for example by preparation of serum or plasma (e.g., isolation of platelet poor plasma).
  • Samples are preferably processed to remove platelets within a time suitable to reduce artifactual increases in soluble P- selectin, such as those due to production of additional P-selectin (e.g., by secretion or proteolysis from platelets) . For example, initiation of such processing within about one hour, and preferably immediately, is desirable.
  • Samples can be further processed as appropriate (e.g., by dilution with assay buffer (e.g., ELISA diluent) ) .
  • the present invention provides a method to determine the effects of anti-platelet therapy and/or vascular intervention (e.g., PTCA) on the level of platelet activation using an assay, such as an enzyme-linked immunosorbent assay, for measuring soluble P-selectin in a suitable sample (e.g., serum, platelet poor plasma (PPP)) comprising: (a) combining a suitable sample, a composition comprising an anti-P-selectin antibody as detector, such as
  • biotinylated anti-P-selectin MAb e.g., S12
  • HRP-streptavidin or
  • HRP-conjugated anti-P-selectin Mab and a solid support, such as a microtiter plate, having an anti-P-selectin capture antibody bound (directly or indirectly) thereto, wherein the detector antibody binds to a different P-selectin epitope from that recognized by the capture antibody, under conditions suitable for the formation of a complex between said anti-P- selectin antibodies and soluble P-selectin; and (b) determining the formation of complex in said samples.
  • the solid support such as a microtiter plate, dipstick, bead, or other suitable support, can be coated directly or indirectly with an anti-P-selectin antibody.
  • a microtiter plate can be coated with an anti-
  • P-selectin antibody or a biotinylated anti-P-selectin Mab can be added to a streptavidin coated support.
  • a variety of solid supports and immobilizing or coating methods can be used, and can be selected according to the desired format.
  • the sample (or soluble P-selectin standard) is combined with the solid support simultaneously with the detector antibody, and optionally with a (i.e., one or more) reagent by which detector can be monitored.
  • the sample e.g., PPP
  • the sample can be combined with the solid support simultaneously with (a) HRP-conjugated anti-P-selectin Mab, or (b) a biotinylated anti-P-selectin Mab and HRP-streptavidin.
  • a known amount of soluble P-selectin standard can be prepared and processed as described above for a suitable sample and used to quantitate the amount of P-selectin detected, permitting measurement of levels relative to a standard.
  • soluble truncated P-selectin is used as a standard.
  • the amount of complex detected can be compared with a suitable control to determine if the levels are elevated.
  • a suitable control For example, the level of soluble P-selectin following a vascular intervention procedure can be compared with a basal level for the individual (e.g., determined prior to or at the time of procedure) , or with levels in normal individuals or suitable controls.
  • the assay can be performed on serum isolated from whole blood of a donor which is allowed to clot in the absence of an anticoagulant with or without a clot-promoting gel.
  • whole blood can be collected (e.g., in a vacutaininer without anticoagulant with or without a clot-promoting gel plug, designed for serum separation) .
  • the blood is allowed to clot and the serum can be harvested from the top of the clotted cell pellet.
  • Serum can be assayed immediately in the ELISA format described above or frozen at -70 C C for later analysis. In the process of clotting, platelet microparticles are released which may be expressing P-selectin on their surface.
  • Table 1 shows that ultracentrifugation at 107,000 x g for 3 hours does not change the detection of P-selectin in the soluble P-selectin ELISA for plasma or serum. These results indicate that microparticles which would be removed by ultracentrifugation are not being detected in the soluble P-selectin ELISA, but that only plasma P-selectin is being detected.
  • ACD-A Acid citrate dextrose, solution A.
  • the assay for measuring soluble P-selectin in a suitable sample comprises the following steps:
  • a sample e.g., plasma
  • a microtiter plate with an anti- P-selectin capture antibody (e.g., W40) or adding a biotinylated anti-P-selectin capture antibody (e.g., W40) to a streptavidin coated solid support such as a microtiter plate; (c) adding, preferably simultaneously, to said microtiter plate the sample to be tested (e.g., final dilution 1:4 with ELISA diluent) and a composition comprising a detector antibody and optionally a reagent for detection, such as (i) HRP-conjugated anti-P-selectin detector antibody (e.g., HRP-S12) , or (ii) a composition comprising biotinylated anti-
  • P-selectin detector antibody e.g., biotinylated Mab S12
  • HRP-streptavidin wherein the anti-P-selectin detector antibody binds to a different P-selectin epitope from that bound by the capture antibody, and incubating same under conditions suitable for the formation of a complex between said anti-P-selectin antibodies and soluble P-selectin, preferably under conditions which maximize binding; (d) separating complexes comprising capture antibody, soluble P-selectin and detector antibody (e.g., by washing) ; and (e) determining the amount of soluble P-selectin in said complexes.
  • biotinylated Mab S12 e.g., biotinylated Mab S12
  • HRP-streptavidin wherein the anti-P-selectin detector antibody binds to a different P-selectin epitope from that bound by the capture antibody, and incubating
  • Typical assays for P-selectin are sequential assays in which a plate is coated with first antibody, plasma is added, the plate is washed, second tagged antibody is added, and the plate is washed and bound second antibody is quantitated.
  • binding kinetics revealed that in a simultaneous format, the off-rate of the second antibody was decreased and the assay was more sensitive (Example 2) .
  • a simultaneous format in which the solid support is coated with a capture antibody (e.g., W40) , and plasma and detector antibody (e.g., S12) are added simultaneously, can achieve enhanced sensitivity and is preferred.
  • the amount of soluble P-selectin in complexes can be determined by a variety of methods.
  • a suitable substrate such as OPD can be added to produce color intensity directly proportional to the bound anti-P-selectin Mab (assessed e.g., by optical density) , and therefore to the soluble P-selectin in the sample.
  • Results can be compared to a suitable control (e.g., a standard, levels of P-selectin in normal individuals, baseline levels of P-selectin in a sample from the same donor) .
  • a suitable control e.g., a standard, levels of P-selectin in normal individuals, baseline levels of P-selectin in a sample from the same donor
  • the assay can be performed using a known amount of soluble P-selectin standard in lieu of sample, and a standard curved established. The amount of complex formed in a sample can then be determined relative to that produced by known amounts of soluble P-selectin standard.
  • FIG. 9 illustrates one type of assay which a volumetric capillary cytometry system can perform (see also Example 11) .
  • murine W40 is used as capture antibody and murine S12 is used as detector antibody.
  • the antibody detection concepts used in the ELISA and sandwich ELISA as described above apply.
  • the above ELISA methods described can be adapted so that the support surface and method of detection utilized is suitable for measurement with a volumetric capillary cytometry system. As described above, a suitable sample is obtained.
  • Samples are processed to remove platelets within a suitable time, preferably within one hour, to reduce artifactual increases in soluble P-selectin, such as those due to production of additional P-selectin.
  • a reagent can be added which stabilizes and prevents in vitro platelet activation.
  • stabilizing reagents are apyrase and PGE ⁇
  • An antibody specific to P-selectin is coated or immobilized on a support surface, such as a bead, solid support strip, or modified capillary surface. The sample is contacted with the coated surface.
  • the coated antibody is W40.
  • another antibody specific to P-selectin or a complex between P-selectin and the coated antibody can contact the sample.
  • This second antibody is detectably labeled with a fluorophore such as Cy5.
  • the volumetric capillary cytometry system can then determine the fluorescent intensity as a measure of soluble P-selectin.
  • the volumetric capillary cytometry system can assess soluble P-selectin in a suitable sample in a method comprising the following steps:
  • the method is useful for monitoring the effect of anti-platelet therapy on platelet activation as reflected by membrane bound and/or soluble P-selectin levels. Treatment can be adjusted accordingly to achieve the desired level of platelet activation. Thus, the method can be used to assess if anti-platelet therapy is indicated or should be altered. Depending on the results obtained, therapy can be maintained or adjusted (increased or decreased, including initiated or discontinued) . For example, the basal activation state of platelets in normal individuals or in a patient can be assessed by measuring membrane bound and/or soluble P-selectin levels, and therapy can be adjusted in order to achieve such a basal activation state, or other desired platelet activation state.
  • the claimed methods can utilize soluble P-selectin and membrane bound P-selectin each independently, or together as in the P-selectin profile.
  • the P-selectin profile provides a particularly sensitive marker for platelet activation and for monitoring and determining the need for anti-platelet therapy.
  • ReoProTM a chimeric Fab antibody fragment directed against glycoprotein Ilb/IIIa
  • GPIIb/IIIa glycoprotein Ilb/IIIa
  • ReoProTM also referred to as abciximab and c7E3 Fab
  • v ⁇ 3 vitronectin receptor Coller, B.S. et al .
  • New Antiplatelet Agents Platelet GPIIb/IIIa Antagonists
  • Thrombosis and Haemosta ⁇ is 74 : 302-308 (1993); Genetta, T.B. and V.F. Mauro, "ABCIXIMAB: A new antiaggregant used in angioplasty," Ann. Phar ocother . , 30 : 251-257 (1996)).
  • anti-platelet agents including other GPIIb/IIIa antagonists, such as other anti-GPIIb/IIIa antibodies (wherein the term "antibody” is as defined herein) , including humanized antibodies, as well as snake venom proteins and their derivatives (e.g., disintegrins, integrelin) , and non-peptide compounds or peptidomimetics, such as Ro 44-9883 (Hoff an-LaRoche) , MK-383 (Merck) , SC54684 (Searle), or other anti-platelet agents (see e.g., Coller, B.S. et al .
  • GPIIb/IIIa antagonists such as other anti-GPIIb/IIIa antibodies (wherein the term "antibody” is as defined herein) , including humanized antibodies, as well as snake venom proteins and their derivatives (e.g., disintegrins, integrelin) , and non-peptide compounds or peptidomimetics, such
  • antibodies having an epitopic specificity similar to that of 7E3 or c7E3 Fab for GPIIb/IIIa and/or the vitronectin receptor including antibodies reactive with the same or a functionally equivalent epitope on GPIIb/IIIa and the vitronectin receptor as bound by c7E3 Fab or 7E3 antibody, can be assessed in this manner.
  • Antibodies with an epitopic specificity similar to that of c7E3 Fab or the 7E3 monoclonal antibody include antibodies which can block the binding of the c7E3 Fab or 7E3 monoclonal antibody to GPIIb/IIIa and/or the vitronectin receptor.
  • the invention relates to a method of assessing the effect of a (i.e., one or more) vascular intervention procedure (e.g., cardiovascular intervention, coronary intervention) , and of a coronary artery intervention procedure in particular, on platelet activation state comprising determining the level of soluble P-selectin in a sample.
  • a vascular intervention procedure e.g., cardiovascular intervention, coronary intervention
  • a coronary artery intervention procedure in particular, on platelet activation state
  • angiography angioplasty
  • angioplasty e.g., performed by balloon, coronary atherectomy, laser angioplasty or other suitable methods (with or without rotablation and/or stent placement)
  • coronary artery by-pass surgery e.g., coronary stent
  • stent placement e.g., coronary stent
  • other vascular intervention procedures e.g., vascular surgery, vascular graft, deployment of a peripheral stent, insertion of a prosthetic valve or vessel (e.g., in autologous, non-autologous or synthetic vessel graft)
  • anti-platelet therapy can be maintained or adjusted in order to achieve a basal activation state, or other desired platelet activation state.
  • the method can be used to assess if antiplatelet therapy is indicated or should be altered. Depending on the results obtained, therapy can be adjusted (increased or decreased, including initiated or discontinued) or maintained. For example, patients whose platelet activation state is elevated relative to a suitable control (e.g. , relative to a pre-procedural control levels, normal control levels) as assessed by monitoring soluble P-selectin levels, can be treated with an anti-platelet agent, such as ReoPro®, in order to reduce soluble P-selectin levels.
  • a suitable control e.g. , relative to a pre-procedural control levels, normal control levels
  • an anti-platelet agent such as ReoPro®
  • patients diagnosed as having deep vein thrombosis display elevated levels of soluble P- selectin preceding vascular intervention (Example 8) .
  • DVT deep vein thrombosis
  • platelet activation state as indicated by levels of soluble P-selectin can be assessed, and anti-platelet therapy can be initiated or adjusted to counteract platelet activation.
  • the P-selectin profile is a particularly sensitive marker for platelet activation and its results can determine the need and effectiveness of anti-platelet therapy.
  • Platelet rich plasma (PRP) and platelet poor plasma (PPP) were prepared from the whole blood of three (3) patients who were undergoing high risk PTCA with stent implantation for coronary artery disease. These patients received the standard dose of an anti-platelet agent for the high risk nature of the PTCA. More specifically, they received a 0.25 mg/kg bolus plus a 10 ⁇ g/min infusion of ReoPro TM during intervention.
  • Flow cytometric analysis as described herein, measured the percent of platelets positive for membrane P- selectin. (See Figure 7 and Example 9).
  • the platelet' s forward and side scatter on a log scale identified the platelets in the patient sample.
  • a collection gate was drawn around the platelets.
  • CellQuest 40TM performed the analysis of the platelet region for the expression of platelet bound P-selectin.
  • the analysis method included overlaying a log FL1 histogram of the negative isotype control mouse IgG-FITC with a log FL1 histogram of P- selectin specific antibody S12-FITC.
  • a technician positioned a statistical marker to obtain a result in 1% of the cells stained with the mouse control antibody being considered positive. Keeping the marker in the same position, the technician determined the percent of the platelets which stain positive for the P-selectin specific antibody, S12-FITC.
  • a sandwich ELISA method as described herein, measured the level of soluble P-selectin in platelet poor plasma
  • PPP produced from the whole blood of patients.
  • this method relies upon the detection of soluble P-selectin in a sample in which two monoclonal antibodies specific for P- selectin will each bind to different epitopes on the P- selectin molecule.
  • Another embodiment of the assay includes a sandwich immunoassay which utilizes a monoclonal and polyclonal P-selectin antibody. In either format, a standard curve of values obtained from samples having known amounts of the soluble P-selectin moiety aid in determining the soluble P-selectin level. The P-selectin profile for sequential samples obtained from various time points before and after the PTCA is evaluated.
  • a positive value for either membrane bound or soluble P-selectin is a value that is greater than or equal to two standard deviations above the mean value of P- selectin in samples from apparently healthy donors.
  • a % positive platelets value greater than 4.4% of platelets and soluble P-selectin value greater than 30 ng/ml are the cut off positive values which indicate a positive result for platelet activation.
  • the table illustrates the ability for the P-selectin profile to determine the need for anti-platelet therapy as well as monitor the effectiveness of anti-platelet therapy.
  • Monitoring the various time points before and after the PTCA allows for determination of the effectiveness of the anti-platelet therapy, and adjustments to the dosage can be made accordingly.
  • patient #1 exhibited an increased level of platelet activation before the PTCA.
  • Patient #l's platelet activation level decreased within 2 hours after the intervention.
  • platelet activation increased 24 hours after the PTCA, indicating that an adjustment might be made to the amount of ReoProTM that patient #1 is receiving.
  • Measuring both soluble and membrane bound P-selectin levels provide a particularly sensitive marker for platelet activation because an elevated level of either indicates a positive diagnosis. For example, measurements at the 24 hour time point for patient #1 indicate that the membrane bound level of P-selectin was within normal limits whereas the soluble P-selectin level was elevated. Measuring only the platelet bound P-selectin level for Patient #1 24 hours after the PTCA could have led to a determination that the anti-platelet therapy was adequate and that the platelet activation state of the patient was within normal limits because the level of membrane bound P-selectin was also within normal limits.
  • Measuring both levels indicate a positive diagnosis for platelet activation because an elevated level of soluble P-selectin exists even though the level of membrane bound P-selectin is within normal limits.
  • the table shows that thirteen of the fifteen or 87% of the time points indicate elevation of only one P-selectin level. Measuring both levels significantly increases the precision for the ability to diagnosis platelet activation for a patient who is receiving anti-platelet therapy. Therefore, measuring both P-selectin levels allows for adjustment of anti-platelet therapy with increased sensitivity.
  • the Table also illustrates that the levels of membrane bound and soluble P-selectin differ among patients receiving a similar procedure and anti-platelet therapy.
  • Patient #1 had an elevated amount of platelet activation at the 24 hour time point and at the 14 day time point.
  • Patient #2 had an elevated level of platelet activation prior to the PTCA, and at the 2 hour, 3 day and 14 day time points.
  • Patient #3 had an elevated level of platelet activation prior to the PTCA, at the 2 hour, 3 day, 7 day and 14 day time points.
  • the radioimmunoassay method used in these in vitro and in vivo studies is described schematically in Figure 1. As shown herein, the method can be used to determine the effects of anti-platelet therapy on the level of platelet activation by measuring the expression of platelet-bound P- selectin. All in vitro and in vivo determinations of platelet bound P-selectin described in the examples were performed according to the following protocol.
  • whole blood (8.5 cc) was collected by venipuncture using a 19-gauge needle in two 10-ml vacutainer tubes containing ACD-A (1.5 cc) as anticoagulant.
  • ACD-A 1.5 cc
  • blood was collected from the in-dwelling arterial line into two plastic syringes containing 1.5 cc ACD-A anticoagulant. In this latter case, each syringe was filled to the 10 cc mark (8.5 cc draw) .
  • the blood with anticoagulant from one vacutainer or syringe was immediately transferred into a polypropylene centrifuge tube (15 ml) containing one premeasured aliquot of apyrase (final concentration 1 U/mL) (Sigma, St. Louis, MO, Catalog No. A 9149) and prostaglandin E x (PGE lr final concentration 1 ⁇ M) (Sigma, St. Louis, MO, Catalog No. P 5515) .
  • Apyrase and PGE ! prevent in vitro platelet activation and stabilize the P-selectin expressed on platelets so that the P-selectin expressed on platelets in this blood sample represents the actual in vivo level of platelet activation.
  • Plasma from the second vacutainer or syringe was immediately transferred into an empty polypropylene centrifuge tube (15 ml) and was subsequently treated with a platelet agonist to establish maximal P- selectin expression for the donor.
  • Platelet rich plasma (PRP) was prepared from whole blood by centrifugation of both polypropylene tubes for 6 minutes at 600 X g. The yellow supernatant PRP was removed from each of the tubes (with or without apyrase and PGE ⁇ ) with plastic pipettes and placed into empty polypropylene tubes.
  • Platelet poor plasma was prepared by centrifuging (10 minutes at 1900 x g) the red cell pellet remaining in the polypropylene centrifuge tube after the preparation of PRP. Platelet counts in PRP were determined using a Coulter counter and the final platelet concentration was adjusted to 1.0 x 10 8 platelets/mL using the appropriate PPP (i.e., with apyrase and PGEj or without apyrase and PGEj) .
  • Platelet bound P-selectin expression was measured in a radioimmunoassay (RIA) using an 125 I-labeled murine anti- human P-selectin monoclonal antibody (MAb) designated S12.
  • MAb murine anti- human P-selectin monoclonal antibody
  • S12 monoclonal antibody which is specific for P- selectin, reacts minimally with unstimulated human platelets, but binds extensively to platelets after activation with thrombin (McEver, R.P and M.N. Martin, "A Monoclonal Antibody to a Membrane Glycoprotein Binds Only to Activated Platelets", J. Biol . Chem . , 259 (15) : 9799-
  • 125 I-labeled anti-P-selectin Mab S12 (final concentration 2 ⁇ g/ml in the PRP) was added to each microfuge tube, and the tubes were incubated for 30 minutes at room temperature. Specific activity was typically in the range of 2 to 4 ⁇ Ci/ ⁇ g. Aliquots (100 ⁇ l) of PRP were removed from each microfuge tube and layered over 30% sucrose (200 ⁇ l) (J.T. Baker, Phillipsburg, NJ, Catalog No. 4097-04) preloaded in slender (400 ⁇ l) polypropylene microfuge tubes.
  • the activation index is the percent of total P-selectin (determined in the PMA activated sample) which is expressed by the platelets in the ex vivo sample (endogenous platelet activation) .
  • Activation Index cpm in pellet of ex vivo PRP X 100 cpm in pellet of PMA activated PRP
  • the activation index (Al) calculated for eight (n 8) normal donors was 2.7 ⁇ 1.5.
  • the ELISA method used in these in vitro and in vivo studies is described schematically in Figure 2. As shown herein, the method can be used to determine the effects of anti-platelet therapy or PTCA on the level of platelet activation by determining the amount of soluble P-selectin in the plasma of persons before, during and/or after treatment with anti-platelet therapeutic agents and/or PTCA. All in vitro and in vivo determinations of soluble P-selectin described in the examples were performed according to the ELISA protocol described below.
  • BIAcore experiments also revealed that the off-rate of soluble P-selectin from S12 was unexpectedly altered when P-selectin was bound with W40 antibody.
  • S12 was coated on a BIAcore chip and soluble P-selectin was bound to saturation, soluble P-selectin was immediately released when antigen flow was discontinued.
  • W40 was used to capture soluble P-selectin on the chip and S12 was allowed to bind to the captured soluble P-selectin, S12 remained attached when buffer was passed over the chip. The off-rate of S12 from soluble P-selectin was decreased when P-selectin was captured by W40.
  • Soluble truncated P-selectin was produced as described below.
  • HRP-streptavidin Horseradish Peroxidase Conjugated Streptavidin (HRP-streptavidin; Jackson Immunoresearch Labs, Catalog No. 016-030-084) ;
  • Biotinylated Murine S12 IgG (bmS12 IgG) was produced as described below.
  • Citric Acid J.T. Baker, Catalog No. 0118-01
  • Sodium Phosphate Dibasic Sigma Chemical Co., Catalog No. S 9763
  • 30 % H 2 0 2 Sigma Chemical Co. , Catalog No. H 1009
  • O-phenylenediamine dihydrochloride O-phenylenediamine dihydrochloride (OPD) (Sigma Chemical Co., Catalog No. P 8287);
  • H 2 S0 4 prepared from concentrated acid (J.T. Baker, Catalog No. 968102) .
  • Citric Acid 20 mM
  • Citric Acid 20 mM
  • (1 liter) 7.1 g
  • Sodium phosphate dibasic (anhydrous) 50 mM
  • Add 900 is water and adjust pH to 5.0;
  • OPD substrate solution Dissolve three 10 mg OPD (25 mis) tablets in 25 mis citrate/phosphate buffer and add 40 ⁇ l 30% H 2 0 2 .
  • Murine W40 IgG Purification
  • Murine W40 IgG ⁇ a murine monoclonal antibody specific for human P-selectin (Johnston, G.I. et al . , J . Biol . Chem . , 264 : 1816-1823 (1989), the teachings of which are incorporated herein by reference)
  • the ascites fluid was then adjusted to 3M NaCl with granular sodium chloride and the pH increased to 8.9 by addition of 1M glycine pH 9.6.
  • Protei_n A Hi-trap columns were equilibrated on a Pharmacia FPLC in MAPS buffer (3M NaCl, 1.5 M glycine, pH 8.9).
  • the ascites fluid, adjusted for salt and pH, was loaded on the Protein A column and flow-through was collected when the OD 280 rose above baseline. Once sample loading was complete the column was washed with additional MAPS buffer until the OD 280 returned to baseline.
  • Bound antibody was first eluted with 0.1M citrate pH 5.5. Collection of eluate was begun and stopped as the OD 280 rose above and returned to baseline. The pool of eluted antibody was immediately neutralized with the addition of 1/3 final volume 1M Tris, pH 8.0. Other non W40 IgG proteins bound to the column were removed by washing with 0.1 M citrate pH 3.5.
  • the pH 5.5 and 3.5 eluates were then concentrated using centriplusTM concentrators and dialyzed into PBS using a Slide-A-lyzerTM apparatus (Pierce) . Finally the sample was 0.2 ⁇ m filtered and the concentration determined by OD 280 .
  • Murine S12 IgGj antibody was purified from hybridoma tissue culture supernatant using Protein A Sepharose column chromatography, and was dialyzed into 200 mM NaHC0 3 , 150 mM KC1, pH 8.5 and concentrated to 3.95 mg/ml for biotinylation. Biotinylation was carried out with a 30:1 molar excess of N. S. -LC-biotin (Pierce) to murine S12 IgG. Briefly, mS12 IgG was transferred to a 5 ml polypropylene tube; N.S. -LC-biotin was weighed out and quickly reconstituted to 4 mg/ml in DI water. The appropriate amount of N.S.
  • Transfectants were cultured in MEM containing 10% FBS and supplemented with L-glutamine, sodium pyruvate, NEAA and geneticin (G-418) in T-150 flasks. When cells reached confluency, supematants were decanted, centrifuged to remove cells and debris and stored at 4°C for purification.
  • Truncated P-selectin Affinity Purification Tissue culture supernatant from 293 cells containing truncated P-selectin (tPS) was collected and pooled for processing. A 25 ml murine GI affinity column was prepared using the anti-P-selectin murine monoclonal antibody GI
  • Bound tPS was eluted with nine column volumes of 100 mM sodium acetate, 100 mM NaCl, pH 4.1. The column was re-equilibrated in 20 mM Tris, 100 mM NaCl, pH 8.3 containing 0.1% NaN 3 and stored for future use.
  • Soluble P-selectin levels were assayed using the following procedure. Table 3. Final concentrations of reagents used in the ELISA.
  • a six point standard curve was prepared by serially diluting tPS from 320 ng/ml to 3.2 ng/ml. Serial dilutions were carried out by transferring 66 ⁇ l standard into wells containing 100 ⁇ l of buffer, mixing and transferring again.
  • Whole blood (8.5 cc) was collected by venipuncture using a 19-gauge needle in two 10-ml vacutainer tubes containing ACD-A (1.5 cc) , heparin or EDTA as anticoagulant. Where the patient had an arterial catheter in place, blood was collected from the in-dwelling arterial line into a plastic syringe containing 1.5 cc ACD-A, heparin or EDTA as anticoagulant. The syringe was filled to the 10 cc mark (8.5 cc draw).
  • the blood with anticoagulant from the vacutainer or syringe was immediately transferred to a polypropylene centrifuge tube.
  • Platelet poor plasma (PPP) was produced by centrifuging the whole blood for 20 minutes at 1900 x g.
  • the PPP was removed from the cell pellet by plastic transfer pipet and was assayed in the ELISA format described below or was aliquoted and frozen at -70°C for later analysis.
  • Soluble P-selectin was measured in an enzyme-linked immunosorbent assay (ELISA) by coating 96-well MaxiSorpTM (Nunc) microtiter plates with murine anti-P-selectin Mab W40 IgG, by adding 100 ⁇ l of antibody (at a concentration of 5 ⁇ g/ml in PBS) to each well. Plates were incubated at 4 C C for approximately 18 hours. The coated microtiter plates were washed three times with 200 ⁇ l/well of PBS, and blocked by the addition of 200 ⁇ l/well of PBS containing 1% bovine serum albumin (BSA, Fraction V, Sigma, St. Louis,
  • BSA bovine serum albumin
  • Plasma samples were evaluated at a final concentration of 1:4 in the assay diluent. After incubation of the samples and standards, plates were washed four times with 200 ⁇ l/well of PBS with 0.05% Tween 20. Color was developed by the addition of 100 ⁇ l/well of the HRP substrate O-phenylenediamine dihydrochloride (OPD) . Color development was stopped after 20 minutes by the addition of 100 ⁇ l/well of 4N H 2 S0 4 .
  • OPD O-phenylenediamine dihydrochloride
  • Figure 3 shows a typical standard curve derived from performance of a soluble P-selectin ELISA for concentrations of soluble P-selectin from 3.2 to 320 ng/ml. As seen in Figure 3 , the mean absorbance for each standard value was plotted on the Y-axis and the concentration of P-selectin on the X-axis. The points were fitted using a log-log curve fitting program. The concentration of soluble P-selectin in samples was determined from the standard curve multiplied by the appropriate dilution factor.
  • the intra-assay variability for the soluble P-selectin ELISA format was determined by adding known amounts of soluble P-selectin to human plasma which had been collected in ACD-A anti- coagulant. In particular, four plasma samples were spiked with high (600 ng/ml) , medium (300 ng/ml) , low (40 ng/ml) or no (0 ng/ml) tPS.
  • the inter-assay variability (precision between assays) of the soluble P-selectin ELISA format was determined in ten (10) different assays in which six replicate determinations of four plasma samples (ACD-A as anticoagulant) were spiked with zero (endogenous soluble P-selectin only) , low (20 ng/ml) , medium (250 ng/ml) , and high (600 ng/ml) amounts of soluble P-selectin (tPS) . As can be observed, all CV's were ⁇ 15% (Table 6). Table 6. Inter-assay variability of the soluble P-selectin ELISA for human plasma
  • Example 3 Use of radioimmunoassay fRIA) to measure induction of expression of platelet P-selectin in response to PMA
  • Platelets in plasma from a healthy donor were isolated from whole blood as described in Example 1 and were activated by the platelet activation agonist PMA at various final concentrations ranging from 5 to 500 nM.
  • P-selectin in the activated platelets was translocated to the membrane in response to PMA in a dose-dependent manner which was measured by the binding of the iodinated anti-P-selectin antibody, 12S I-S12. Greater amounts of antibody represented by higher counts per minute were bound with increasing concentrations of PMA.
  • Figure 4A and Figure 4B show the results of this titration.
  • the data are presented as counts bound and in Figure 4B, the Activation Index (Al) for each titration of PMA is calculated according to the formula set forth in Example 1.
  • Example 4 Expression of platelet bound P-selectin induced by PMA is decreased in the presence of ReoPro® and can be measured by RIA
  • Platelet rich plasma was prepared from the whole blood of two (2) normal donors as described in Example 1, and the PRP from each donor was divided into 2 equal aliquots.
  • ReoPro® also referred to as abciximab or c7E3 Fab; Centocor, Inc., Malvern, PA
  • GPIIb/IIIa glycoprotein Ilb/IIIa receptor on platelets
  • ReoPro® at a final concentration of 5 ⁇ g/ml, was added to one half of the PRP for each donor and incubated for 20 minutes. PRP with and without ReoPro was then divided into 0.5 ml aliquots and stimulated with various concentrations of PMA for 15 minutes. P-selectin expressed on the surface of platelets subjected to all of the various conditions was measured by the binding of 125 I-S12 using the RIA protocol described in Example 1.
  • Example 5 Expression of platelet bound P-selectin in patients undergoing PTCA can be measured in the RIA and is decreased in the presence of ReoPro®
  • Platelet rich plasma was prepared from the whole blood of six (6) patients who were undergoing PTCA for coronary artery disease including unstable angina and myocardial infarction. Two patients received ReoPro® and one patient had rotational atherectomy (rotablation) . The Activation Index calculated for normal donors is 2.7 ⁇ 1.5. Activation Indices of 4.2 are greater than one standard deviation above the mean. Activation Indices of 5.7 or above are two standard deviations above the mean. Table 7 shows that in patients #4 and #6, both of whom had received a bolus plus 12 hour infusion of ReoPro® per the manufacture's directions, the Activation Indices were significantly elevated before the PTCA and immediately after, but had fallen into the normal range by 24 hours after the procedure.
  • anti-platelet therapies including ReoPro® can be determined by using the RIA described in Example 1 to measure platelet activation state through the expression of P-selectin. Table 7. RIA and Soluble P-selectin ELISA Results from Patients Undergoing PTCA
  • Table 7 shows the platelet P-selectin expression in 6 patients before and after PTCA.
  • Platelet P-selectin was measured as percent positive cells by flow cytometric evaluation of PRP using a FITC-labeled S12 Mab (See R.E. Scharf et al., Arteriosclerosis and Thrombosis, 12 : 1475 (1992)) and by RIA using 125 I-labeled S12 as detailed in Example 1.
  • Soluble P-selectin expression pre- and post PTCA was determined using the ELISA protocol in Example 2 Example 6. The expression of soluble P-selectin is induced in vitro by PMA activation in a time- and dose-dependent fashion and is decreased in the presence of ReoPro®
  • GPIIb/IIIa antagonist ReoPro® was observed in an in vitro study in which whole blood was obtained in ACD-A anticoagulant from three (3) normal donors. The whole blood was divided into 2 ml aliquots and incubated at room temperature in separate polypropylene tubes for various time periods with various amounts of the platelet activation agonist PMA. Time points assayed were 1, 5, and 24 hours, and amounts of PMA were 0, 20, 100, and 500 nM. For each donor, duplicate tubes were made for each time point and PMA concentration. One of the duplicate tubes contained ReoPro® (abciximab, c7E3) at a final concentration of 5 ⁇ g/ml and the other had the same amount of ReoPro® diluent added to it.
  • ReoPro® abciximab, c7E3
  • Platelet poor plasma was prepared from the whole blood at the various time points from 1 to 24 hours and the soluble P-selectin was determined using the ELISA protocol described in Example 2.
  • Figure 6 shows that by five hours there is a decrease in the soluble P-selectin observed in blood stimulated by 20, 100, and 500 nM PMA in the presence of ReoPro® (66.1 ⁇ 5 ng/mL vs 96.8 ⁇ 4 ng/mL (500 nM PMA treatment)). In the presence of 500 nM PMA, there was a 31% decrease in soluble P-selectin that can be detected in the presence of ReoPro.
  • Example 7 Soluble P-selectin is increased in patients undergoing PTCA in proportion to the increase in their platelet bound P-selectin and is a useful tool in the monitoring of the activation produced bv PTCA Unexpectedly, the levels of soluble P-selectin increased post-PTCA in those patients where the PTCA procedure induced significant platelet activation.
  • the level of soluble P-selectin in their platelet poor plasma was determined by the ELISA protocol described in Example 2.
  • One example of this observation shown in Table 7 is patient #3 whose unstable plaque architecture resulted in a post-PTCA activation index of 29.95 (2.7 ⁇ 1.5 is normal).
  • the soluble P-selectin level for patient #3 was subsequently elevated at 24 hours after the procedure (from 24.5 ng/ml pre-PTCA to 34.14 ng/ml at 24 hours post-PTCA) .
  • patients #2 and #13 both had diagnoses of DVT and had significantly high levels of soluble P-selectin (patient #2, 65.03 ng/ml; and patient #13, 55.3 ng/ml).
  • soluble and platelet bound P-selectin were not significantly elevated in vascular surgery in the absence of coronary disease, including repair of abdominal aortic aneurysm (AAA) (Patients #4 and #7) .
  • AAAA abdominal aortic aneurysm
  • NCM nonionic contrast media
  • Table 8 shows the platelet P-selectin expression of 13 patients who had vascular surgery or events other than PTCA. Platelet P-selectin was measured as percent positive cells in a flow cytometric protocol (See Table 7) , and by RIA according to the protocol in Example 1. Soluble P-selectin expression pre- and post-surgery was determined using the ELISA protocol in Example 2.
  • AAA repair of abdominal aortic aneurysm
  • NCM nonionic contrast media
  • AMI acute myocardial infarction
  • DVT deep vein thrombosis.
  • Flow cytometry is one method to determine the level of platelet P-selectin and its result contributes to the measurement of the P-selectin profile.
  • Flow cytometry is a method for determining the platelet P-selectin in patient samples, as discussed herein.
  • the soluble P-selectin was determined using an enzyme-linked immunosorbent assay (ELISA) protocol. Normal values for platelet activation as measured by membrane bound P-selectin and normal levels of circulating soluble P-selectin were determined for apparently healthy volunteer donors.
  • ELISA enzyme-linked immunosorbent assay
  • ACD solution A Becton Dickinson, Catalog No 364606
  • ACD solution B Becton-Dickinson, Catalog No 364816
  • VWR polypropylene centrifuge tube
  • PGEj prostaglandin E
  • Platelet rich plasma was prepared by centrifuging the whole blood at 600 x g in a Beckman GS-6KR centrifuge or equivalent, equipped with a rotor with swinging bucket, with no brake for 3 minutes (blood volumes of 3-6 mL) or 6 minutes (blood volumes of lOmL) at room temperature.
  • the supernatant platelet rich plasma was removed from each centrifuge tube using a plastic transfer pipette (Sarstedt, No 86.1174 or equivalent) and transferred to a 5 mL polypropylene snap cap tube (VWR,
  • Subpart b) Processing the Platelet Rich Plasma for flow cytometric analysis Platelets in platelet rich plasma were stained with P-selectin specific monoclonal antibodies for flow cytometric analysis. Normal donors have a low percent of activated platelets or platelets which are expressing P- selectin. These normal donors provided samples which were used to determine the level of significant platelet activation. Patient samples showed significant platelet activation when the percent of total platelets which are positive for P-selectin is greater than or equal to 2 standard deviations above the mean for the percent positive platelets observed in apparently healthy volunteer donors. Platelet rich plasma for flow cytometric analysis was diluted 1:6 in Modified Tyrodes Buffer(MTB) and inverted to mix gently.
  • Three stained samples were prepared by aliquoting 45 ⁇ L of diluted platelet rich plasma into each of two tubes containing 5 ⁇ L of Modified Tyrodes Buffer and one tube containing 5 ⁇ L of phorbol 12-myristate 13-acetate (PMA) (Sigma, P-8139 or equivalent) to produce a final concentration of 20 nM PMA.
  • the 20 nM PMA maximally activated the platelet rich plasma during a 15 minute incubation at room temperature and this sample acted as a control to show that the P-selectin specific antibody bound to its ligand in this system.
  • Subpart c) Flow Cytometric analysis of platelets in PRP The prepared samples were analyzed for platelet P-selectin expression using a FACSan flow cytometer (Becton Dickinson, San Jose, CA) . The instrument was equipped with a 15-mW argon-ion laser at a wavelength of 488nm. The FITC fluorescence was detected using a 530-nm band pass filter. Platelets were identified by their forward and side scatter on a log scale. The characteristic platelet light scatter was confirmed using a 10E5-FITC antibody to stain the GP IIB/IIIa receptor found on all platelets. A collection gate was drawn around the platelet population and used to collect 10,000 platelets at a rate of 400-1000 events per second.
  • a series of color histograms were generated illustrating the diagnostic sensitivity from the flow cytometry assay shown in Figure 1.
  • the histograms were not included in the application because they are in color.
  • the purpose of the histogram was to show the linearity of the addition of an increasing percent of fully activated platelets to whole blood containing non-activated platelets. Blood was drawn from a donor and divided into two parts one of which was not activated and the other was activated with PMA. Activated platelets were added to the non-activated sample to increasing percent of the total. The effect of the addition of activated platelets was determined by flow cytometric measurement of the resulting percent positive platelets. In this particular experiment, the basal activation of the non-activated sample is 5.06 %. The addition to the non-activated blood sample of maximally activated platelets amounting to 1% of the total number resulted in the detection of 6.41 % activated platelets.
  • Example 10 The use of a Volumetric Capillary Cytometry System for measuring membrane bound P-selectin: The volumetric capillary cytometry system utilized to measure membrane bound P-selectin was the IMAGN2000TM from Biometric Imaging, Mountain View, CA. (See Figure 8)
  • Subpart a) Obtaining and preparing a suitable sample for measuring membrane bound P-selectin using a volumetric capillary cytometry system: Whole blood was collected by venipuncture using a 19- gauge needle into a vacutainer tube containing either ACD solution A (Becton Dickinson, Catalog No 364606) or ACD solution B (Becton-Dickinson, Catalog No 364816) as anticoagulant. Within 30 minutes of the draw, the blood with anticoagulant from one vacutainer was transferred into a 15 mL polypropylene centrifuge tube (VWR, Catalog No 21008-102) containing one premeasured aliquot of apyrase (final concentration 1 U/mL (Sigma, St.
  • VWR polypropylene centrifuge tube
  • the stained, fixed, and diluted whole blood sample (40 ⁇ L) was placed in the well of a plastic capillary (Catalog No VC120, Biometric Imaging, Mountain View, CA) and the fluorescence intensity and number of events within the platelet size gate was determined in the IMAGN2000 instrument (Biometric Imaging, Mountain View, CA) .
  • the total platelet count in each sample was determined on the IMAGN2000 Biometric Imaging instrument using a Cy5 labeled CD61 antibody (Becton Dickinson) or a 10E5-Cy5 antibody (Centocor Inc., Malvern PA) both of which bind to essentially all platelets.
  • Cy5-labeled CD61 and 10E5 at 5 ⁇ g/mL (10 X) were stored frozen (-20°C) in 200 ⁇ L aliquots. During use, the reagent is stored at 4°C. Unused refrigerated reagent is discarded monthly.
  • the total platelet count was performed in whole blood by transferring 5 ⁇ L of blood to a 12 x 75 mm polypropylene tube (Falcon 2063 or equivalent) containing 5 mL of Modified Tyrodes Buffer and pipetting up and down twice to complete the 1:1000 dilution. 45 ⁇ L of the diluted blood was then added to an amber tube (Sarstedt Catalog No.72.694.034 or equivalent) containing 5 ⁇ L of CD61-Cy5 or 10E5-Cy5 (5 ⁇ g/mL) and incubated at room temperature for 20 minutes.
  • Example 11 Use of a volumetric capillary cytometry system to measure soluble P-selectin:
  • the IMAGN2000TM volumetric capillary cytometry system was used to measure soluble P-selectin.
  • the preferred embodiment is using a bead-based format. (See Figure 9)
  • the sample for this example was prepared in the same way as the sample that was prepared in Example 2, discussing the ELISA method of determining soluble P-selectin.
  • the technician coated the polystyrene beads with the anti-P-selectin antibody, W40.
  • the beads, also called polystyrene sulfated microparticles, (9,7 ⁇ m) were passively coated at 0.5 x the available particle surface area with the P-selectin specific antibody, W40.
  • the beads were diluted in 30 mM phosphate buffered saline, 1% BSA, 0.01% Tween 20 such that the solids comprised 0.01% of the assay volume.
  • Soluble P- selectin produced by a transfected human kidney cell line (293 cells) was added to the beads in diluent at a range of concentrations.
  • S12-Cy5 a flourescently labeled anti-P- selectin antibody, was added at a final concentration of
  • the technician placed the assay mixture containing the beads in the capillary of the IMAGN2000TM machine. Forty (40) ⁇ L of the assay mixture was placed in the well of a plastic capillary (Catalog No VC120, Biometric Imaging , Mountain View, CA) and the fluorescence intensity and number of events within the platelet size gate was determined on the IMAGN2000 instrument (Biometric Imaging, Mountain View,
  • Example 12 Measurements and sensitivity of P-selectin in the presence of a platelet agonist using a volumetric capillary cytometry system:
  • IMAGN2000TM provides the capability to detect in a sample of whole blood, the number of platelets that are activated and therefore, expressing P-selectin on their surface (membrane) .
  • Activated platelets were detected by the addition of a labeled P-selectin specific Mab (in this embodiment a cocktail of S12-Cy5 and W40-Cy5) .
  • a fixative was finally added to the whole blood to insure that in vitro platelet activation did not occur.
  • Blood which has been incubated with labeled P-selectin Mab(s) and fixed was then placed into a capillary and a predetermined volume of the blood was scanned by the optical (laser) mechanism of the instrument.
  • a size range that allows for the discrimination of different cell types based on size can be pre-programmed into the instrument.
  • every fluorescent event of magnitude sufficiently above background fluorescence was recorded as one event.
  • an increasing number of platelets was activated to the degree that their fluorescence intensity (signal) was sufficiently above background (noise) such that the ratio of signal to noise qualified them to be counted by the instrument as an event.
  • the platelet activation agonist PMA phorbol myristate acetate
  • Each platelet had sufficient CD61-Cy5 on its membrane to be detected as an event (high enough signal to noise ratio) .
  • the number of events positive for CD61-Cy5 when adjusted for the volume scanned and the dilution factor of the blood provided a count of the number of platelets in the whole blood. Dividing the number of events (platelets) positive for P-selectin by the total number of platelets resulted in the percent positive platelets.
  • the percent for normal states and for activated platelet states was established by flow cytometry.
  • the percent positive platelets derived from the current instrument was initially correlated with a similar calculation derived from flow cytometric determinations on the same sample to establish the substantial equivalence of the two methods.
  • Example 13 Measurement of P-selectin using a bead format in a volumetric capillary cytometry system:
  • the article of manufacture, IMAGN2000 provides the capability to detect and quantify the amount of soluble P- selectin present in a sample of platelet poor plasma (PPP) .
  • PPP platelet poor plasma
  • 9.7 ⁇ M latex beads were coated (covalently or passively) with a P-selectin specific Mab. The coating of the beads was carefully controlled so that a uniform amount of P-selectin Mab was present on each bead. A specified number of beads were incubated in PPP containing soluble P-selectin.
  • a Cy5 labeled P-selectin specific Mab binding to a different site on soluble P-selectin from the site bound by the Mab used to coat the bead, or a Cy5 labeled polyclonal anti-P-selectin antibody preparation was added to the PPP.
  • the plasma containing the beads was placed in a capillary and a predetermined volume of plasma was scanned by the optical (laser) mechanism of the instrument.
  • a size determination that includes the 9.7 ⁇ M beads was pre-set within the program of the instrument. Within that size window, the fluorescent intensity of all events which were sufficiently fluorescent (above background) was calculated.
  • the fluorescent intensity of the beads was directly proportional to the amount of soluble P-selectin bound by the surface Mab and detected by the labeled P-selectin antibody(ies) .
  • a series of beads were coated with increasing and carefully determined amounts of isolated recombinant soluble P-selectin and then incubated with the Cy5 labeled anti-P-selectin antibody used in the assay. In this way, a standard curve of fluorescence intensity corresponding to soluble P-selectin concentration was established.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • Analytical Chemistry (AREA)
  • Cell Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
EP97950589A 1996-11-13 1997-09-13 P.seketinassays und verfahren zu deren herstellung Withdrawn EP0946877A1 (de)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US74838796A 1996-11-13 1996-11-13
US748387 1996-11-13
PCT/US1997/020571 WO1998021591A1 (en) 1996-11-13 1997-11-13 P-selectin assays and methods of use thereof

Publications (1)

Publication Number Publication Date
EP0946877A1 true EP0946877A1 (de) 1999-10-06

Family

ID=25009255

Family Applications (1)

Application Number Title Priority Date Filing Date
EP97950589A Withdrawn EP0946877A1 (de) 1996-11-13 1997-09-13 P.seketinassays und verfahren zu deren herstellung

Country Status (5)

Country Link
EP (1) EP0946877A1 (de)
JP (1) JP2001526774A (de)
AU (1) AU5355298A (de)
CA (1) CA2277094A1 (de)
WO (1) WO1998021591A1 (de)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2971600A (en) * 1999-01-26 2000-08-07 Du Pont Pharmaceuticals Company A facs method for detection of gpiib/iiia inhibitor dependent activators in plasma samples
JP2008249552A (ja) * 2007-03-30 2008-10-16 Otsuka Pharmaceut Co Ltd 可溶性血小板膜糖タンパク質viの測定系
CN116840485A (zh) * 2023-05-18 2023-10-03 河络新图生物科技(上海)有限公司 一种检测血小板功能亚群PAC-1标志物、Annexin-V标志物和CD62P标志物的检测系统、检测方法及检测试剂盒

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2868808B2 (ja) * 1989-11-07 1999-03-10 寳酒造株式会社 活性化血小板抗原測定試薬
WO1995012125A1 (en) * 1993-10-26 1995-05-04 Mayo Foundation For Medical Education And Research Direct fluorescence-conjugated immunoassay for platelet activation
BR9507579A (pt) * 1994-10-25 1997-09-09 Watson Clinic Foundation Processo fluorométrico para medir ativaçao de plaqueta endógena em amostra de plaquetas

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9821591A1 *

Also Published As

Publication number Publication date
JP2001526774A (ja) 2001-12-18
CA2277094A1 (en) 1998-05-22
WO1998021591A8 (en) 2000-05-18
AU5355298A (en) 1998-06-03
WO1998021591A1 (en) 1998-05-22

Similar Documents

Publication Publication Date Title
Michelson Flow cytometry: a clinical test of platelet function
US8323905B2 (en) Methods of detection GPVI
Rand et al. Platelet function assays
Michelson et al. Evaluation of platelet function by flow cytometry
Diacovo et al. A functional integrin ligand on the surface of platelets: intercellular adhesion molecule-2.
Gelderman et al. Flow cytometric analysis of cell membrane microparticles
Andrioli et al. Study of platelet adhesion in patients with uncomplicated hypertension
Bednar et al. Fibrinogen receptor antagonist-induced thrombocytopenia in chimpanzee and rhesus monkey associated with preexisting drug-dependent antibodies to platelet glycoprotein IIb/IIIa
Lagadec et al. Involvement of a CD47-dependent pathway in platelet adhesion on inflamed vascular endothelium under flow
US5196309A (en) Characterization of platelet aggregation disorders
US5939276A (en) Flow cytometry assay for heparin-induced thrombocytopenia
WO2007010240A2 (en) Method for high-throughput screening
AU689346B2 (en) Direct fluorescence-conjugated immunoassay for platelet activation
Fanelli et al. Flow cytometric detection of circulating activated platelets in primary antiphospholipid syndrome. Correlation with thrombocytopenia and anticardiolipin antibodies
WO1999018442A1 (en) Diagnosis of thrombotic events by detecting p-selectin
EP0946877A1 (de) P.seketinassays und verfahren zu deren herstellung
WO2000025140A1 (en) Method for determining platelet count
Ozsavci et al. Flow cytometric assay of platelet glycoprotein receptor numbers in hypercholesterolemia
JP3713716B2 (ja) 血小板GPIIb/IIIa受容体の新規分析方法
Cox et al. Activation-specific neo-antigens on platelets detected by monoclonal antibodies
US5457028A (en) Standard platelet samples and method for preparation thereof
DK173362B1 (da) Monoklonalt antistof, hybridoma, som udskiller antistoffet, cellekulturpræparat indeholdende et sådant hybridoma, diagnosti
US20040077028A1 (en) Assay for thrombocytopenia associated with administration of platelet GPIIb-IIIa receptor antagonists
Michelson et al. Flow cytometric analysis of platelet function
JP2925684B2 (ja) 血小板及び血管内皮細胞の疾病の測定方法

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19990602

AK Designated contracting states

Kind code of ref document: A1

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

17Q First examination report despatched

Effective date: 20040318

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20040729