CA2263063C - Method for diagnosing and distinguishing stroke and diagnostic devices for use therein - Google Patents

Method for diagnosing and distinguishing stroke and diagnostic devices for use therein Download PDF

Info

Publication number
CA2263063C
CA2263063C CA002263063A CA2263063A CA2263063C CA 2263063 C CA2263063 C CA 2263063C CA 002263063 A CA002263063 A CA 002263063A CA 2263063 A CA2263063 A CA 2263063A CA 2263063 C CA2263063 C CA 2263063C
Authority
CA
Canada
Prior art keywords
protein
marker
body fluid
specific
ischemic
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.)
Expired - Fee Related
Application number
CA002263063A
Other languages
French (fr)
Other versions
CA2263063A1 (en
Inventor
George Jackowski
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.)
Nanogen Inc
Original Assignee
Syn X Pharma 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
Priority to CA002263063A priority Critical patent/CA2263063C/en
Application filed by Syn X Pharma Inc filed Critical Syn X Pharma Inc
Priority to JP2000602637A priority patent/JP4560212B2/en
Priority to AU27884/00A priority patent/AU765923B2/en
Priority to CN00803524.5A priority patent/CN1339108A/en
Priority to PCT/CA2000/000182 priority patent/WO2000052476A1/en
Priority to PT00906097T priority patent/PT1155325E/en
Priority to EP00906097A priority patent/EP1155325B1/en
Priority to BR0008317-8A priority patent/BR0008317A/en
Priority to EP04027808A priority patent/EP1521083A3/en
Priority to AT00906097T priority patent/ATE283485T1/en
Priority to NZ513005A priority patent/NZ513005A/en
Priority to DE60016178T priority patent/DE60016178T2/en
Priority to DK00906097T priority patent/DK1155325T3/en
Priority to US09/510,700 priority patent/US6235489B1/en
Priority to ES00906097T priority patent/ES2232424T3/en
Priority to US09/621,592 priority patent/US6780606B1/en
Publication of CA2263063A1 publication Critical patent/CA2263063A1/en
Publication of CA2263063C publication Critical patent/CA2263063C/en
Application granted granted Critical
Priority to US10/924,283 priority patent/US7655424B2/en
Priority to US12/698,032 priority patent/US7955811B2/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/001Enzyme electrodes
    • 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
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • 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
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/2871Cerebrovascular disorders, e.g. stroke, cerebral infarct, cerebral haemorrhage, transient ischemic event
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/969Multiple layering of reactants
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/97Test strip or test slide
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/973Simultaneous determination of more than one analyte
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/975Kit
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/807Apparatus included in process claim, e.g. physical support structures
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/807Apparatus included in process claim, e.g. physical support structures
    • Y10S436/808Automated or kit
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/807Apparatus included in process claim, e.g. physical support structures
    • Y10S436/81Tube, bottle, or dipstick
    • 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
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/811Test for named disease, body condition or organ function

Abstract

A method for determining whether a subject has had a stroke and, if so, the type of stroke which includes analyzing the subject's body fluid for at least four selected markers of stroke, namely, myelin basic protein, S100 protein, neuronal specific enolase and a brain endothelial membrane protein such as thrombomodulin or a similar molecule. The data obtained from the analyses provide information as to the type of stroke, the onset of occurrence and the extent of brain damage and allow a physician to determine quickly the type of treatment required by the subject.

Description

METHOD FOR DIAGNOSING AND DISTINGUISHING STROKE
AND DIAGNOSTIC DEVICES FOR USE THEREIN
BACKGROUND OF THE INVENTION
This application is directed to a method for diagnosing whether a subject has had a stroke and, if so, differentiating between the different types of stroke. More specifically, the method includes analyzing the subject's body fluid for at least four selected markers of stroke. There are also described diagnostic devices and kits for use in the method.
to The impact of stroke on the health of human beings is very great when considered in terms of mortality and even more devastating when disability is considered. For example, stroke is the third leading cause of death in adults in the United States, after ischemic heart disease and all forms of cancer. For people who survive, stroke is the leading cause of disability. The direct medical costs due to stroke and the cost of lost employment amount to billions of dollars annually.
Approximately 85% of all strokes are ischemic (thrombotic and embolic) with the remainder being hemorrhagic.
Stroke is an underserved market for both therapeutics and diagnostic techniques. In the United States alone over 700,000 people have strokes each year. A
multiple of that number would be suspected of having strokes with diagnostics only confirmed by expensive technology including computer-assisted tomography (CAT) . scans and magnetic resonance imaging (MRI). However, these sophisticated technologies are not available in all hospitals and they are also not sensitive enough to diagnose ischemic stroke at an early stage.
Stroke is a clinical diagnosis made by a neurologist, usually as a consultation.
Current methods for diagnosing stroke include symptom evaluation, medical history, chest X-ray, ECG (electrical heart activity), EEG (brain nerve cell activity), CAT scan to assess brain damage and MRI to obtain internal body visuals. A number of blood tests may be performed to search for internal bleeding. These include complete blood count, prothrombin time, partial thromboplastin time, serum electrolytes and blood glucose.
Determining the immediate cause of a stroke can be di~cult especially upon presentation where the diagnosis relies mainly on imaging techniques.
Approximately 50% of cerebral infarctions are not visible on a CAT scan.
Further, even though a CAT scan can be very sensitive for the identification of hemorrhagic stroke, it is not very sensitive for cerebral ischemia during evaluation of stroke and is usually positive at from 24 to 36 hours after onset of stroke. As a result a window of opportunity for rapid treatment would usually have expired once the current diagnostic techniques positively identify a stroke.
The treatment of stroke includes preventive therapies such as antihypertensive and antiplatelet drugs which control and reduce blood pressure and thus reduce the likelihood of stroke. Also, the development of thrombolytic drugs such as t-PA
(tissue plasminogen activator) has provided a significant advance in the treatment of t 5 ischemic stroke victims but to be effective and minimize damage from acute stroke it is necessary to begin treatment very early, for example, within about three hours a$er the onset of symptoms. These drugs dissolve blood vessel clots which block blood flow to the brain and which are the cause of approximately 80% of strokes.
However, these drugs can also present the side effect of increased risk of bleeding.
Various neuroprotectors such as calcium channel antagonist can stop damage to the brain as a result of ischemic insult. The window of treatment for these drugs is typically broader than that for the clot dissolvers and they do not increase the risk of bleeding.
Diagnostic techniques for the early diagnosis of stroke and identification of the type of stroke are needed to allow the physician to prescribe the appropriate therapeutic drugs at an early stage in the cerebral event. Various markers 'for stroke are known and analytical techniques for the determination of such markers have been described in the art. As used herein the term "marker" refers to a protein or other molecule that is released from the brain during a cerebral ischemic or hemorrhagic event. Such markers include isoforms of proteins that are unique to the brain.
3o It has been reported in the literature that myelin basic protein (MBP) concentration, in cerebrospinal fluid (CSF) increases after sufficient damage to neuronal tissue, head trauma and AIDS dementia. Further, it has been reported that ultrastructural immunocytochemistry studies using anti-MBP antibodies have shown that MBP is localized exclusively in the myelin sheath. Thus, ~t has been suggested the MBP levels in CSF or serum be used as a marker of cerebral damage in acute cerebrovascular disease. See Strand, T., et al., Brain and plasma proteins in spinal fluid as markers for brain damage and severity of stroke, Stroke (1984) 15;
138-144.
The increase in MBP concentration in CSF is most evident in about four to five days after the onset of thrombotic stroke while in cerebral hemorrhage the increase was highest almost immediately after onset. See Garcia-Alex, A., et al., Neuron-specific to enolase and myelin basic protein: Relationship of cerebrospinal fluid concentration to the neurologic condition of asphyxiated fiall~term infants, Pediatrics (1994) 93;
234-240. It has also been found that patients with transitory ischemic attack (TIA) had normal CSF values for MBP while those with cerebral infarction and hemorrhage had elevated values. In cerebral infarction there was a significant increase in MBP
t 5 concentration in CSF from the first to second lumbar puncture while patients with intracerebral hemorrhage had reached already markedly elevated levels at the first lumbar puncture. It was reported that the kinetic difference in MBP release may be useful in the differential diagnosis of hemorrhagic and ischemic stroke. MBP
levels in CSF also correlated to the visibility of the cerebral lesion at CT scan and to the 20 short-term outcome of the patients. Further, the concentration of MBP
increased with the extent of brain lesion and high values indicated a poor short-term prognosis for the patient. See Strand, T. et al, previously cited.
S I 00 protein is another marker which may be taken as a useful marker for assessing neurologic damage and for determining the extent of brain damage and for 25 determining the extent of brain lesions. Thus, it has been suggested for use' as an aid in the diagnosis and assessment of brain lesions and neurological damage due to stroke. See Missler, U., Weismann, M., Friedrich, C. and Kaps, M., S 100 protein and neuron-specific enolase concentrations in blood as indicators of infarction volume and prognosis in acute ischemic stroke, Stroke (1997) 28; 1956-60.
3o Neuron-specific enolase (NSE) also has been suggested as a useful marker of neurologic damage in the study of stroke with particular application in the assessment of treatment. See Teasdale, G. and Jennett, B., Assessment of coma and impaired consciousness, Lancet (1974) 2; 81-84.
There continues to be a need for diagnostic techniques which can provide timely information concerning the type of stroke suffered by a patient, the onset of occurrence, the location of the event, the identification of appropriate patients who will benefit from treatment with the appropriate drug and the identification of patients who are at risk of bleeding as a result of treatment. Such techniques can provide data which will allow a physician to determine quickly the appropriate treatment required by the patient and permit early intervention.
to It is therefore an object of this invention to provide a method for rapidly diagnosing and distinguishing stroke.
It is a further object of the invention to provide a method for distinguishing between thrombotic strokes and hemorrhagic strokes.
It is another object of the invention to provide such a method which includes ~ 5 analyzing the body fluid of a patient for at least four markers of stroke.
It is yet another object to provide a method which can provide information relating to the time of onset of the stroke.
It is still another object to provide diagnostic assay devices for use in the method.
2o SUMMARY OF THE INVENTION
These and other objects and advantages are accomplished in accordance with the invention by providing a method that is capable of determining whether a patient has suffered a stroke and, if so, whether the event is thrombotic or hemorrhagic.
According to the method, a body fluid of the patient is analyzed for four molecules 25 which are cell type specific, three of which are specific ischemic markers, namely S100 protein, myelin basic protein (MBP) and specific neuronal enolase (NSE) and one brain endothelial membrane protein, for example, thrombomodulin (Tm). The method analyzes the isoforms of the marker proteins which are specific to the brain.
The analyses of these markers may be carried out on the same sample of body fluid or on multiple samples of body fluid. In the latter embodiment the different body fluid samples may be taken at the same time or at different time periods.
The information which is obtained according to the method of the invention can be pro~~ided at the critically important early stages of a stroke, e.g., within the first three to six hours after onset of symptoms since the analysis of the patient's body fluid <;an be c;~rried out in about 45 to 50 minutes after the body fluid is collected. 'Che data can be vital to the physician by assisting in the determination of how to treat a patient presenting with symptoms of stroke or suspected of having a stroke. 'the data can rule stroke in or out, and differentiate between ischemic and hemorrhagic stroke and therefore exclude hemorrhagic stroke patients from being given clot dissolving therapeutics because of the risk of increased bleeding. rChe data can also identify patients who are at risk of bleeding as a result of treatment, i.~e., patients with compromised brain vasculature.
Further, the method c:an provide at an early stage prognostic information relating to the outcome of intervention which can improve patient selection for appropriate therapeutics and intervention. The method of the invention is diagnostic well before the imaging te:chnologiies. In addition, these data can indicate the location of the stroke within the brain and the extent of damage to the brain as well as determine whether the extent of the stroke is increasing. The cerebral infarct associated with stro~;e, made up of dead and dying brain tissue, which forms because of inadequate oxygenation typically increases in size during the acute period after ischemia begins. By measuring the markers in samples of body fluid taken at different points in time the progress of the stroke can be ascertained.
According to a further broad aspect of the present invention there is provided a method for diagnosing and distinguishing stroke. The method comprises analyzing the body fluid of a patient to detect the presence and concentration of four markers of stroke and wherein a first marker is myelin basic protein, a second marker is the beta isoform of S 100 protein, a third marker is neuronal specific enolase, amd a fourth marker is a brain endothelial cell membrane protein. From the information obtained from the analyses one verifies whether an ischemic or hemorrahagic cerebral event has occurred and differentiates a particular type of cerebral event.
According to a further broad aspect of the present invention there is provided a diagnostic kit for diagnosing and distinguishing stroke and which comprises at least four antibodies which are specific for each of four different marker proteins, and wherein the antibodies immobilized on a solid support. A
first marker protein is myelin basic protein and a first antibody is specific therefor.
A second marker protein is the beta isoform of S 100 protein and a second antibody is specific therefor. A third marker protein is neuronal specific enolase and a third antibody is specific therefor. A fourth marker protein is a brain endothelial cell membrane protein and a fourth antibody is specific therefor and at least four labeled antibodies. Each of the labeled antibodies binds to one of the maxker protein.
According to a still further broad aspect of the present invention there is provided a method for the differential diagnosis of ischemic and hemorrhagic cerebral events. The method comprises analyzing the body fluid of a patient to detect the presence and concentration level of one or more ischemic marker proteins selected from the group consisting of myelin basic protein, the beta isoform of S 100 protein, neuronal specific enolase and combinations thereof.
The body fluid of the patient is analyzed to detect the presence and concentration level of a brain endothelial cell membrane protein. From the information obtained from the analyses, the occurrence of an ischemic or hemorrhagic cerebral event is verified, and differentiating a particular type of cerebral event.
According to a still further broad aspect of the present invention there is provided a method for the differential diagnosis of ischemic and hemorrhagic cerebral events comprising a. analyzing a body fluid of a patient to detect the presence and concentration level of one or more ischemic marker proteins selected from the group consisting of myelin basic protein, the beta isoform of 5100 protein, neuronal specific enolase and combinations thereof, -Sa-b. analyzing a body fluid of said patient to detect the presence and concentration level of a brain endothelial cell membrane protein, and c. comparing said concentration of each protein detected in steps a and b to specific threshold values to determine the presence of statistically significant concentrations thereof, wherein the presence of said proteins in statistically significant concentrations is indicative that an ischemic cerebral event or a hemorrhagic cerebral event has occurred and differentiates which type of cerebral event has occurred.
BRIEF DESCRIPTION OF THE DRAWINGS
The invention will now be described further in detail with respect to various preferred embodiments thereof in conjunction with the accompanying drawings wherein:
Fig. 1 is a graphical illustration of the concentration over time (in minutes) of two marker proteins which are indicative of cerebral condition or status;
-Sb-Fig. 2 is a flow chart illustrating how data obtained according to an embodiment of the invention can be used for the diagnosis of cerebral condition or status; and Figs. 3-10 are graphical illustrations of the concentration over time (in days) of four marker proteins analyzed according to an embodiment of the invention.
DESCRIPTION OF THE PREFERRED EMBODIMENTS
The markers which are analyzed according to the method of the invention are released into the circulation and are present in the blood and other body fluids.
Preferably blood, or any blood product that contains them such as, for example, plasma, serum, cytolyzed blood (e.g., by treatment with hypotonic buffer or detergents), and dilutions and preparations thereof is analyzed according to the invention. In another preferred embodiment the concentration of the markers in CSF
is measured.
The terms "above normal" and "above threshold" are used herein to refer to a level of a marker that is greater than the level of the marker observed in normal individuals, that is, individuals who are not undergoing a cerebral event, i.e. an injury to the brain which may be ischemic, mechanical or infectious. For some markers, no or infinitesimally low levels of the marker may be present normally in an individual's blood. For others of the markers analyzed for according to the invention, detectable levels may be present normally in blood Thus, these terms contemplate a level that is significantly above the normal level found in individuals. The term "significantly"
refers to statistical significance and generally means a two standard .deviation (SD) above normal, or higher, concentration of the marker is present. The assay method by which the analysis for any particular marker protein is carried out must be sufficiently sensitive to be able to detect the level of the marker which is present over the concentration range of interest and also must be highly specific.
The four primary markers which are measured according to the present method are proteins which are released by the specific brain cells as the cells become damaged during a cerebral event. These proteins can be either in their native form or 3o immunologically detectable fragments of the proteins resulting, for example, by enzyme activity from proteolytic breakdown. The specific four primary markers when mentioned in the present application, including the claims hereof, are intended to include fragments of the proteins which can be immunolo~ically detected. By "immunologically detectable" is meant that the protein fragments contain an epitope which is specifically recognized by a cognate antibody.
As mentioned previously, the markers analyzed according to the method of the invention are cell type specific. Myelin basic protein (1V)BP) is a highly basic protein, localized in the myelin sheath, and accounts for about 30% of the total protein of the myelin in the human brain. The protein exists as a single polypeptide chain of to amino acid residues which has a rod-like structure with dimensions of I.5 x 150 nm and a molecular weight of about 18,500 Dalton. It is a flexible protein which exists in a random coil devoid of a helices (3 conformations.
The increase of MBP concentration in blood and CSF is most evident about four to five days after the onset of ischemic stroke while in cerebral hemorrhage the increase is highest almost immediately after the onset. Further, patients with TIA
have normal values for MBP while those with cerebral infarction and intercerebral hemorrhage have elevated values. A normal value for a person who has not had a cerebral event is from 0.00 to about 0.016 ng/mL. MBP has a half life in serum of about one hour and is a sensitive marker for cerebral hemorrhage.
2o The S 100 protein is a cytoplasmic acidic calcium binding protein found predominantly in the gray matter of the brain, primarily in glia and Schwann cells.
The protein exists in several homo- or heterodimeric isoforms consisting of two ~, immunologically distinct subunits, alpha (MW = 10,400 Dalton) and beta (MW
=
10,500 Dalton) while the S100aa is the homodimer as which is found mainly in striated muscle, heart and kidney. The SIOOb isoform is the 21,000 Dalton homodimer X3(3. It is present in high concentration in glial cells and Schwann cells and is thus tissue specific. It is released during acute damage to the central nervous system and is a sensitive marker for cerebral infarction. According to the method of the invention, the assay is specific for the ~i-subunit of the S 100 protein.
_7_ The S 1 OOb isoform is a specific brain marker released during acute damage to the central nervous system. It is eliminated by the kidney and has a half life of about two hours in human serum. Repeated measurements of S 100 sebum levels are useful to follow the course of neurologic damage. Additionally, the presence of elevated S 100 levels in CSF or serum, in association with stroke symptoms, can be useful in the differential diagnosis of stroke and may be a valuable indicator of cerebral infarction.
The enzyme enolase (EC 4.2.1.11 ) catalyzes the interconversion of 2-phosphoglycerate and phosphoenolpyruvate in the glycolytic pathway. The enzyme to exists in three isoproteins each the product of a separate gene. The gene loci have been designated ENO1, EN02 and EN03. The gene product of ENO1 is the nonneuronal enolase (NNE or a), which is widely distributed in various mammalian tissues. The gene product of EN02 is the muscle specific enolase (MSE or ~3) which is localized mainly in the cardiac and striated muscle, while the product of the EN03 gene is the neuronal specific enolase (NSE or y) which is largely found in the neurons and neuroendocrine cells. The native enzymes are found as homo- or heterodimeric isoforms composed of three immunologically distinct subunits, a, (3 and y.
Each subunit has a molecular weight of approximately 39,000 Dalton.
The ay and yy enolase isoforms, which have been designated neuronal specific 2o enolase (NSE) each have a molecular weight of approximately 80,000 Dalton.
It has been shown that NSE concentration in CSF increases a$er experimental focal ischemia and the release of NSE from damaged cerebral tissue into the CSF
reflects the development and size of the infarcts. NSE has a serum half life of about 48 hours and its peak concentration has been shown to occur later after cerebral artery (MCA) occlusion. NSE levels in CSF have been found to be elevated in acute and/or extensive disorders including subarachnoid hemorrhage and acute cerebral infarction.
The fourth marker protein measured according to the invention is a brain endothelial membrane protein. Endothelial cells which line the small blood vessels of the brain possess a unique expression of cell surface, receptors, transporters and 3o intracellular enzymes that serve to tightly regulate exchange of solutes between blood _g_ and brain parenchyma. Brain endothelial membrane proteins include:
Thrombomodulin (Tm), a 105,000 Dalton surface glycoprotein involved in the regulation of intravascular coagulation; Glucose Transporter (Glue 1), a 55,000 Dalton cell surface transmembrane protein which may exist in dimeric or tetrameric form; Neurothelin/HT7, a 43,000 Dalton protein integrated into the cytoplasmic membrane transport protein; Gamma Glutamyl Transpeptidase, a protein which is found as a heterodimeric isoform composed of 22,000 and 25,000 Dalton subunits and is involved in the transfer of gamma glutamyl residue from glutathione to amino acids; and P-glycoprotein, a multidrug resistant membrane spanning protein. In a preferred embodiment of the method Tm is the brain endothelial membrane protein which is measured. Tm is a sensitive marker for lacunar infarcts.
The data obtained according to the method indicate whether a stroke has occurred and, if so, the type of stroke, the localization of the damage and the spread of the damage. Where the levels of all four markers are negative, i.e., within the normal range, there is no cerebral injury. When only the brain endothelial membrane protein, e.g., Tm, is elevated, or positive, i.e., the level is at least 25D above normal, the stroke is a lacunar infarct present in the basal ganglia and deep white matter of the brain.
When the NSE level is positive and the S 100 and/or MBP levels are negative (the brain endothelial membrane protein marker is positive or negative) the patient has 2o suffered a TIA.
According to another preferred embodiment, a fifth marker, which is from the specific cell type of one of the three ischemic markers analyzed according to the method of the invention, is measured to provide information related to the time of onset of the stroke. It should be recognized that the onset of stroke symptoms is not always known, particularly if the patient is unconscious or elderly and 'a reliable clinical history is not always available. An indication of the time of onset of the stroke can be obtained by relying on the differing release kinetics of brain markers having different molecular weights. The time release of brain markers into the circulation following brain injury is dependent on the size of the marker, with smaller 3o markers tending to be released earlier in the event while larger markers tend to be released later. Fig. 1 illustrates the release kinetics of two marker proteins which are analyzed according to the method of the invention, namely MBP and S 100. These data were obtained from fluid collected from the brain tissue of a pig after coronary bypass surgery was performed. The samples were collected at 0, 30, 120, 180 and 240 minutes after the subject had been removed from the bypass machine. The concentration values are expressed in multiples of a baseline value which was the concentration at time zero. These data indicate that the release of MBP (MW =
18,500) appears to reach a maximum about 120 minutes after the ischemic event whereas the release of 5100 (MW = 21,000) does so at after about 180 minutes.
Thus, by measuring an additional protein marker from the specific cell type of one of to the three ischemic markers utilized in the method of the invention, data relating to the time of onset can be obtained. The time of onset is defined as the moment of onset of clinical symptoms of stroke. In this preferred embodiment the second marker protein - is a larger, i.e., a higher molecular weight marker, than the primary marker of the same cell type.
The three ischemic markers utilized according to the invention and various other high molecular weight markers from the same specific cell type are shown in Table I.

TABLE I

SMALLEST
MARKER SIZE (D) FRAGMENT (D) SPECIFIC GLIAL MARKERS: , S 100 21,000 10,500 Growth Associated Protein 43 (GAP-43)43,000 43,000 Glutamine Synthetase (GS) ~ 400,000 44,000 Glial Fibrillary Acid Protein (GFAP)51,000 51,000 GlycineTransporter(GLYT1) 50-70,000 50-70,000 Glycine Transporter (GLYT2) 90-110,000 90-110,000 SPECIFIC NEURONAL MARKERS:

Neuron Specific Enolase (NSE) 78,000 39,000 Neruon Specific Glycoprotein (GP50)42,000 42,000 Calpain 80,000 55,000 Neurofibrillary Protein (NF) 68,000 68,000 Heat Shock Protein 72 (HSP-72 72,000 72,000 Beta Amyloid Precursor Protein 250,000 125,000 (beta APP) SPECIFIC AXONAL MARKERS:

Myelin Basic Protein (MBP) 18,500 18,500 Calbindin D-28K 28,000 28,000 Proteolipid Protein (PLP) 23-30,000 23-30,000 Myelin Associated Glycoprotein 90-100,000 58,000 (MAG) Neurofilament H (HFN) 200,000 200,000 In a preferred embodiment of the invention body fluid samples taken from a patient at different points in time are analyzed. Typically a first body fluid sample is taken from a patient upon presentation with symptoms of stroke and analyzed according to the invention. Subsequently, some period of time after presentation, for example, about two hours after presentation, a second body fluid sample is taken and analyzed according to the invention. Referring now to Fig. 2 there is seen a flow chart illustrating how the data obtained from four marker proteins analyzed according to the invention, in the embodiment illustrated NSE, S 100, MBP and Tm, can be used to triage the patient. The data can be used to diagnose stroke, rule out stroke, distinguish between thrombotic and hemorrhagic stroke, identify appropriate patients for thrombolytic treatment and determine how the stroke is evolving.
As stated previously, the level of each of the four specific markers in the patient's body fluid can be measured from one single sample or one or more individual markers can be measured in one sample and at least one marker measured in one or more additional samples. _ By "sample" is meant a volume of body fluid such as blood or CSF which is obtained at one point in time. Further, as will be discussed in detail below, all the markers can be measured with one assay device or by using a separate assay device for each marker in which case aliquots of the same fluid sample t o can be used or different fluid samples can be used. 1t is apparent that the analyses should be carried out within some short time frame after the sample is taken, e.g., within about one-half hour, so the data can be used to prescribe treatment as quickly as possible. It is preferred to measure each of the four markers in the same single sample, irrespective of whether the analyses are carried out in a single analytical device or in separate such devices so the level of each marker simultaneously present in a single sample can be used to provide meaningful data.
Generally speaking, the presence of each marker is determined using antibodies specific for each of the markers and detecting immunospecific binding of each antibody to its respective cognate marker. Any suitable immunoassay method 2o may be utilized, including those which are commercially available, to determine the level of each of the specific markers measured according to the invention.
Extensive discussion of the known immunoassay techniques is not required here since these are known to those of skill in the art. Typical suitable immunoassay techniques include sandwich enzyme-linked immunoassays (ELISA), radioimmunoassays (RIA), competitive binding assays, homogeneous assays, heterogeneous assays, etc'.
Various of the known immunoassay methods are reviewed in Methods in Enzymology, 70, pp.
30-70 and 166-198 (1980). Direct and indirect labels can be used in immunoassays.
A direct label can be defined as an entity, which in its natural state, is visible either to the naked eye or with the aid of an optical filter and/or applied stimulation, e.g., 3o ultraviolet light, to promote fluorescence. Examples of colored labels which can be used include metallic sol particles, gold sol particles, dye sol particles, dyed latex particles or dyes encapsulated in liposomes. Other direct labels include radionuclides and fluorescent or luminescent moieties. Indirect labels such as enzymes can also be used according to the invention. Various enzymes are known ~or use as labels such as, for example, alkaline phosphatase, horseradish peroxidase, lysozyme, glucose-6-phosphate dehydrogenase, lactate dehydrogenase and urease. For a detailed discussion of enzymes in immunoassays see Engvall, Enzyme Immunoassay ELISA and EMIT, Methods of Enzymology, 70, 419-439 ( 1980).
A preferred immunoassay method for use according to the invention is a double antibody technique for measuring the level of the marker proteins in the t0 patient's body fluid. According to this method one of the antibodies is a "captwe"
antibody and the other is a "detector" antibody. The captwe antibody is immobilized on a solid support which may be any of various types which are known in the art such as, for example, microtiter plate wells, beads, tubes and porous materials such as nylon, glass fibers and other polymeric materials. In this method, a solid support, t 5 e.g., microtiter plate wells, coated with a captwe antibody, preferably monoclonal, raised against the particular marker protein of interest, constitutes the solid phase.
Diluted patient body fluid, e.g., serum or plasma, typically about 25 p1, standards and controls are added to separate solid supports and incubated. When the marker protein is present in the body fluid it is captwed by the immobilized antibody which is 20 specific for the protein. After incubation and washing, an anti-marker protein detector antibody, e.g., a polyclonal rabbit anti-marker protein antibody, is added to the solid support. The detector antibody binds to marker protein bound to the capture antibody to form a sandwich structwe. After incubation and washing an anti-IgG
antibody, e.g., a polyclonal goat anti-rabbit IgG antibody, labeled with an enzyme such as 25 horseradish peroxidase (I-IRP) is added to the solid support. After incubation and washing a substrate for the enzyme is added to the solid support followed by incubation and the addition of an acid solution to stop the enzymatic reaction.
The degree of enzymatic activity of immobilized enzyme is determined by measuring the optical density of the oxidized enzymatic product on the solid support 3o at the appropriate wavelength, e.g., 450 nm for I-IRP. The absorbance at the wavelength is proportional to the amount of marker protein in the fluid sample. A set of marker protein standards is used to prepare a standard curve of absorbance vs.
marker protein concentration. This method is preferred since test results can be provided in 45 to 50 minutes and the method is both sensitive aver the concentration range of interest for each marker and is highly specific.
The assay methods used to measure the marker proteins should exhibit sufficient sensitivity to be able to_ measure each protein over a concentration range from normal values found in healthy persons to elevated levels, i.e., 2SD
above normal and beyond. Of course, a normal value range of the marker proteins can be found by analyzing the body fluid of healthy persons. For the S 1 OOb isoform where +2SD = 0.02 ng/mL the upper limit of the assay range is preferably about 5.0 ng/mL.
For NSE where +2SD = 9.9 ng/mL the upper limit of the range is preferably about 60 ng/mL. For MBP, which has an elevated level cutoff value of 0.02 ng/mL, the upper limit of the assay range is preferably about 5.0 ng/mL and for Tm, which has an elevated level cutoff value of about 73 ng/mL, the assay range upper limit is preferably about 500 ng/mL.
The assays can be carried out in various assay device formats including those described in United States Patents 4,06,439; 5,051,237 and 5,147,609 to PB
Diagnostic Systems, lnc.
The assay devices used according to the invention can be arranged to provide a semiquantitative or a quantitative result. By the term "semiquantitative" is meant the ability to discriminate between a level which is above the elevated marker protein value, and a level which is not above that threshold.
The assays may be carried out in various formats including, as discussed previously, a microtiter plate format which is preferred for canrying out the assays in a batch mode. The assays may also be carried out in automated immunoassay'analyzers which are well known in the art and which can carry out assays on a number of different samples. These automated analyzers include continuous/random access types. Examples of such systems are described in United States Patents 5,207,987 and 5,518,688 to PB Diagnostic Systems, Inc. Various automated analyzers that are 3o commercially available include the OPUS~ and OPUS MAGNUM~ analyzers.

Another assay format which can be used according to the invention is a rapid manual test which can be administered at the point-of care at any location.
Typically, such point-of care assay devices will provide a result which His above or below a threshold value, i.e., a semiquantitative result as described previously.
s It should be recognized also that the assay devices used according to the invention can be provided to carry out one single assay for a particular marker protein or to cant' out a plurality of assays, from a single volume of body fluid, for a corresponding number of different marker proteins. A preferred assay device of the latter type is one which can provide a semiquantitative result for the four primary marker proteins measured according to the invention, i.e., S 1 OOb, NSE, MBP
and a brain endothelial marker protein, e.g., Tm. These device typically are adapted to provide a distinct visually detectable colored band at the location where the capture antibody for the particular marker protein is located when the concentration of the marker protein is above the threshold level. For a detailed dic~necinn of acea,r +«.,o~
is which can be utilized according to the invention as well as various assay formats and automated analyzer apparatus see U.S. Patent 5,747,274 to Jackowski.
The invention will now be described further in detail with respect to specific preferred embodiments, it being understood that these are intended to be illustrative only and the invention is not limited to the materials, procedures, etc.
recited therein.
2o EXAMPLE
A prospective observational pilot study was carried out at two tertiary care hospitals. The study evaluated thirty three patients admitted with a clinical and computed tomographic (CT) diagnosis of acute ischemic stroke. The mean age of the patients presenting with stroke was approximately 66 years (66.4 116.4) with an age 2s range of from 27 to 90 years. The mean delay between the onset of symptoms and presentation to the hospital was 22 hours with a range of from 1 to 72 hours.
Admission National Institutes of Health Stroke Scale and Discharge modified Rankin scale scores were recorded. Blood samples were obtained on days 1 (presentation), 3, S and 7 at one hospital and days l, 2 and 3 at the second hospital. All blood samples were centrifuged and aliquots of serum were frozen and stored at -80°C
until analysis for S 100, NSE, MBP and Tm.
Control subjects included one hundred three healthy bloPd donors (age range from 18 to 78 years; mean age 54.6115.2 years) whose blood samples were used to determine reference values for concentrations of S 100 and NSE and twenty four healthy blood donors who provided samples for reference measurements of MBP
and Tm concentrations.
All reference values are reported as mean +2SD unless otherwise stated. The reference value for 5100 in serum was 0.0067 ng/mL with a 98th percentile of 0.020 ng/mL. An elevated S 100 value was taken as any concentration greater than the 98th percentile (0.02 ng/mL) of normal (normal +2SD = 0.02 ng/mL).
The reference value for NSE in serum was 5.03 X2.40 ng/mL. An elevated NSE value was any concentration greater than 2SD above normal, 9.85 ng/mL.
The reference value for MBP in serum was 0.0162 (0.0019 ng/mL. An ~ 5 elevated MBP value was any concentration greater than 2SD above normal, 0.02 ng/mL.
The reference value for Tm in serum was 50.52 (13.62 ng/mL. An elevated Tm value was any concentration greater than +2SD above normal, 76.14 ng/mL.
The levels of S 100 and NSE were analyzed using Exact S 100 and Exact NSE
20 Elisa Assay Kits, respectively, available from Skye PharmaTech Inc., Mississauga, Canada. The levels of Tm were analyzed with an ELISA assay available from Diagnostica Stago, 9 rue des Freres Chausson, 92600 Asneres Sur Seine, France.
The level of MBP concentration was analyzed with an ELISA immunoassay from Diagnostic Systems Laboratories, Webster, Texas, United States.
25 In the tables showing the data obtained "D1" indicates the first day with the first blood sample being taken at the time of presentation. Subsequent days of sample collection are indicated by D2, D3, etc. For the values of the concentrations of the markers, +2SD are above the normal range. "ND" signifies that no data was obtained.

TABLE
II
NSE, 5100, MBP ND
Tm CONCENTRATIONS
IN
CLINICAL
SERUM
SAMPLES

CODE # AGE GENDER NSE 5100 ~ MBp Tm (ng/mL) (ng/mL) (ng/mL) (ng/mL) + 2SI~9.9+ 2SD~.02+ 25D=0.02+ 25D=73 SM-1 D1 42 Female 8.342 0.028 0.000 43.535 SM-I D3 13.300 1.098 ND 61.946 SM-1 DS 9.622 0.060 0.238 65.859 SM-I D7 10.710 0.066 1.725 62.177 DIAGNOSIS Left internal carotid.
CEREBRAL
INFARCT
(arteroembolic).
5h from onset of symptoms.

OUTCOME GOOD.
Mild aphasia.

SM-2 D1 SS Female 9.420 0.053 0.032 ND

SM-2 D3 5.430 0.015 0.105 ND

SM-2 DS 7.360 0.011 0.341 ND

SM-2 D7 9.906 0.008 0.124 ND

DIAGNOSIS CEREBRAL
INFARCT.
Posterior circulation infarction (unknown mechanism).
20 h from onset of symptoms.

OUTCOME MODERATE.
Dysarthia and hemiparesis.

5M-3 D 78 Male 12.670 0.112 0.000 92.324 I

SM-3 D3 14.980 0.719 1.420 101.990 SM-3 DS 28.570 1.301 4.845 I 19.251 DIAGNOSIS CEREBRAL
INFARCT.
Total anterior circulation infarction (cardioembolic).

OUTCOME DEATH

SM-4 D 58 Male 8.520 0.008 0.000 ' 73.913 SM-4 D3 4.406 0.028 0.147 78.286 SM-4 DS 4.888 0.024 0.265 85.881 DIAGNOSIS CEREBRAL
INFARCT.
Lacunar circulation infarction (lacune).

OUTCOME GOOD.
Mild ataxic hemiparesis.

_ 17_ TABLE
II
NSE, 5100, MBP ND
Tm CONCENTRATIONS
IN
CLINICAL
SERUM
SAMPLES

CODE # AGE GENDER NSE 5100 ~ MBp 'I~n (ng/mL) (ng/mL) (ng/mL) (ng/mL) + 2SI~9.9+ 2SD~.02+ 2SD~.02 + 2SD=73 SM-5 D2 27 Male 9.139 0.099 2.301 59.415 SM-5 D3 5.492 0.000 0.090 53.892 SM-S DS 11.730 0.079 7.682 68.850 SM-S D7 11.540 0.018 10.382 68.620 DIAGNOSIS CEREBRAL
INFARCT
(fibromuscular dysplasia).
48h fibm onset of symptoms.

OUTCOME MODERATE.
Aphasia and hemiparesis.

5M-6 D1 63 Male 7.029 0.000 0.000 56.883 SM-6 D3 6.455 0.020 0.000 75.985 DIAGNOSIS CEREBRAL
INFARCT
(unknown mechamism).
22 h from onset of symptoms.

OUTCOME MODERATE

SM-7 D 64 Female 8.566 0.021 0.013 105.212 SM-7 D3 5.061 0.024 0.000 129.146 SM-7 DS 6.783 0.021 0.017 129.607 SM-7 D8 7.377 0.015 0.000 162.746 DIAGNOSIS CEREBRAL
INFARCT.
Lacunar circulation infarction (lacune).

OUTCOME MODERATE.
Hemiparetic.

SM-8 D1 45 Male 15.740 0.053 0.009 37.092 SM-8 D3 21.010 0.112 0.082 35.711 DM-8 DS 15.060 0.095 0.112 38.703 DIAGNOSIS CEREBRAL
INFARCT
(Right vertebral dissection).

OUTCOME GOOD.
Minimal deficit.

TABLE

NSE, 5100, MBP
ND
Tm CONCENTRATIONS
IN
CLINICAL
SERUM
SAMPLES

CODE # AGE GENDER NSE S100 ~ MBP Tm (ng/mL) (ng/mL) (ng/mL) (ag/mL) + 2519.9 + 2SD~.02+ 2SD~.02+ 25D=73 SM-9 D1 35 Male 11.530 0.015 0.101 ND

SM-9 DS 8.033 0.021 0.040 ND

SM-9 D7 7.336 0.002 0.000 ND

DIAGNOSIS CEREBRAL
INFARCT
(unknown mechanism).

OUTCOME GOOD.
Minimal deficit.

TABLE
III

CODE # AGE GENDER NSE S100 MBP 'Ib, (ng/mL) (ng/mL) (ng/mL) (ng/mL) + 2SI1~9.9+ 2SD~.02+ 2SD~.02+ 25D=73 SJ-O1 83 MALE 6.803 0.091 0.000 185.760 SJ-O1 8.566 0.235 0.000 166.659 SJ-O1 8.689 1.143 0.000 209.234 DIAGNOSIS CEREBRAL
INFARCT
(recurrent).
I BP, renal insufficiency, MI

OUTCOME Severe impairment developed on second day.

5J-02 61 MALE 14.040 0.054 0.433 476.193 SJ-02 13.430 0.110 1.199 403.010 SJ-02 12.890 0.247 2.625 501.739 DIAGNOSIS CEREBRAL
INFARCT
(parietal infarction), renal failure, MI, CA. 48 h from onset of symptoms OUTCOME First CT negative.
Second CT positive (Day 3). DEATH
(day 5) SJ-03 83 MALE 10.700 0.000 0.000 75.064 SJ-03 8.926 0.000 0.000 81.968 SJ-03 9.000 0.000 0.000 89.793 DIAGNOSIS CEREBRAL
INFARCT
(lacune).
T BP, DM

OUTCOME CT positive (Day 2) TABLE
III

CODE # AGE GENDER NSE 5100 MBP 'Ilm (ng/mL) (ng/mL) (ng/mL) (ng/mL) + 2SI~9.9+ 2SD~.02+ hSD~.02+ 25D=73 SJ-04 70 FEMALE 10.270 0.000 0.000 134.209 DIAGNOSIS TIA. I
BP, DM

OUTCOME

SJ-OS 72 MALE 6.639 0.000 0.326 185.760 Dl SJ-OS 10.870 0.000 0.219 136.281 SJ-OS 8.197 0.000 0.387 132.598 DIAGNOSIS CEREBRAL
INFARCT
(lacune), renal impairment OUTCOME First CT negative SJ-06 81 FEMALE 10.440 0.001 0.086 ND
Dl DIAGNOSIS CEREBRAL
INFARCT.
Renal impairment (dialysis).
36 h from onset of symptoms OUTCOME

SJ-07 90 FEMALE 12.540 0.001 0.162 ND
DI

DIAGNOSIS CEREBRAL
INFARCT.
36 h from onset of symptoms OUTCOME

SJ-08 81 MALE 12.450 0.749 0.017 82.198 DIAGNOSIS HAEMORRHAGIC.
1 h from onset of symptoms OUTCOME CT positive.
DEATH
2 h later.

SJ-09 46 MALE 4.891 0.000 0.000 88.182 SJ-09 3.913 0.000 0.000 87.722 SJ-09 1.848 0.000 0.000 105.903 DIAGNOSIS STROKE
(clinically).
PA within 3 h of onset of symptoms OUTCOME CT negative TABLE
III

CODE # AGE GENDER NSE 5100 MBP Tm (g/mL) 1P~mL) ~~~) ~~~) + 2SI~9.9+ 2SD~.02+ 2SD~.02+ 2SI~73 SJ-10 69 FEMALE 8.303 0.000 0.000 79.437 SJ-10 6.000 0.000 0.000 74.144 SJ-10 3.939 0.000 0.000 68.850 DIAGNOSIS ~12 h from onset of symptoms - numbness in arms - R side facial droop;
difficulty swallowing - no past Hx CVA

- patient diabetic;
has Hx high BP

OUTCOME Initial CT negative.
All symptoms resolved;
except patient still unable to swallow.

5J-11 39 MALE 10.770 0.058 0.063 65.398 SJ-11 12.050 0.047 0.128 69.311 SJ-11 17.330 0.068 0.189 76.675 DIAGNOSIS CEREBRAL
INFARCT.
~24 h from onset of symptoms - found unconscious with R-sided neglect OUTCOME CT positive (Day I) - 3 lesions present ~2 cm - basal ganglia L side Patient still has severe weakness R side with speech impairment SJ-12 S1 FEMALE 11.700 0.000 0.067 286.100 DI

SJ-12 8.788 0.000 0.055 270.911 SJ-12 I 1.800 0.002 0.124 226.264 INFARCT
(lacune).

~ 12 h from onset of symptoms - weakness L side, esp.
L arm - facial droop and pronounced slurring of speech - Bell's Palsy L side - renal dialysis patient OUTCOME CT positive (Day - developed thrombocytopenia Day 2 TABLE
III

CODE # AGE GENDER NSE SI00 MBP '15n ~01~) + 2SI~9.9+ 2SD~.02+~2SD~02 + 25173 SJ-13 78 FEMALE 10.090 0.000 0.000 46.297 SJ-13 40.040 0.768 0.433 41.924 (Haemolytic) SJ-13 4.667 0.103 0.000 36.861 DIAGNOSIS CEREBRAL
INFARCT
(Left MCA CVA) + CAD
+ Diabetic, Hx HTN, + family Hx CVA.
~ 19 h from onset of symptoms OUTCOME Initial CT negative.
Initial symptoms worsened over 48 h to R
hemiplegia.

SJ-14 72 MALE 7.303 0.087 0.299 NC

SJ-14 5.697 0.007 0.055 NC

DIAGNOSIS CEREBRAL
INFARCT
(Left CVA).
9 h from onset of symptoms - prior - Hx strial fib., anticoagulated OUTCOME Symptoms improving SJ-15 79 MALE 5.667 0.000 0.013 ND
DI

INFARCT
(Left CVA) - symptoms progressive over 2 wk period;
worsened over day period just prior to presentation at hospital.

OUTCOME CT negative Day 1 - condition worsening at discharge (discharged at family's request for palliative care at home) SJ-16 90 FEMALE 20.940 0.81 I 5.142 52.281 SJ-16 12.220 0.498 5.459 ! 55.733 SJ-l6 9.424 0.253 3.377 55.503 DIAGNOSIS Large intracerebral bleed with smaller subdural hematoma and intraventricular hemorrhage - Onset of symptoms unknown (6 to 29 h prior) - previously well;
no Hx other than colon Ca 20 yr prior;
on no meds at home;
found collapsed OUTCOME Patient continues to worsen TABLE
III

CODE # AGE GENDER NSE SI00 MBP Tm O~mL) I~mL) 0~~) Off) + 25I)=,9.9+ 25110.02+~SD~.02 + 25D=73 SJ-17 77 MALE 10.660 0.042 0.002 ND
Dl SJ-17 8.758 0.095 0.006 ND

SJ-17 12.510 0.261 0.417 ND

DIAGNOSIS CEREBRAL
INFARCT
(Right CVA) - old left cerebellar infarct - sudden onset;
slurred speech and L-sided weakness 15 h from onset of symptoms OUTCOME CT showed old CVA
and new right MCA infarct SJ-18 79 MALE 21.560 0.008 0.000 61.946 SJ-18 14.390 0.218 0.814 48.598 SJ-18 11.050 0.102 0.698 55.963 DIAGNOSIS Initial CT showed bleed or cerebral edema.
2 h from onset of symptoms OUTCOME Aphasia and R-sided weakness SJ-19 82 FEMALE 9.948 0.000 ND 64.248 DI

SJ-19 9.781 0.008 ND 58.955 SJ-19 I 1.720 0.023 ND 64.248 DIAGNOSIS TIA ~
24 h from onset of symptoms OUTCOME Slurred speech, difl'lculty swallowing which persists.

SJ-20 ND MALE 26.400 0.122 0.000 32.719 DIAGNOSIS Haemorrhagic stroke OUTCOME

SJ-21 74 MALE 5.828 0.016 ND 74.374 SJ-21 7.423 0.063 ND 75.985 SJ-21 8.436 0.286 ND 71.382 DIAGNOSIS CEREBRAL
INFARCT
(left CVA) OUTCOME R-sided weakness TABLE

CODE # AGE GENDER NSE 5100 MBP Tm O~~) ~~~~) ~n~mL) + 2SD=9.9+ 2SD~.02 + dSD=0.02+ 2S1~73 SJ-22 63 FEMALE 18.600 0.000 0.000 ND

(Haemolytic) SJ-22 9.540 0.008 0.000 ND

DIAGNOSIS CEREBRAL
INFARCT
(left CVA), initial CT negative OUTCOME weakness (resolving) SJ-23 79 MALE 14.530 2.009 5.478 ND

SJ-23 23.980 >3.200 8.155 ND

SJ-23 27.670 2.218 7.309 ND

DIAGNOSIS CEREBRAL
INFARCT, CT positive OUTCOME CT showed multiple cerebral infarcts.

SJ-24 73 MALE 20.630 0.000 0.000 74.160 SJ-24 17.880 0.000 0.000 89.750 SJ-24 17.880 0.000 0.000 83.290 DIAGNOSIS TIA
- sudden decrease in ability to function, word difficulties OUTCOME CT negative - Discharged with diagnosis of TIA

The analysis of S 100, NSE and MBP levels in serum samples from healthy control subjects showed no relationship of levels of these proteins to age or sex. In the case of Tm, the concentrations were higher in serum samples from healthy males than in females (54.62 113.62 ng/mL, 2SD above normal = 81.86 ng/mL and 43.63 +11.18 ng/mL, 2SD above normal = 68.74 ng/mL, respectively).
Of the thirty three stroke patients twenty six were infarcts (79%) and of these five were lacunar ( 15%) and four had hemorrhagic stroke ( 12%). Of the hemorrhagic stroke patients three had subarachnoid hemorrhage and one had an intracerebral bleed.
Three patients (9%) had transient ischemic attacks (TIA).

On presentation the levels of S 100 were elevated in 44% of the patients, NSE
levels were elevated in 59%, MBP levels were elevated in 40% and Tm levels were elevated in 57%.
The data indicate that by measuring the four marker proteins in accordance with the invention, where any one marker was elevated, 94% of the patients could be identified on presentation. Nineteen of the twenty one non-lacunar infarcts (90%) could be identified on presentation. The remaining two patients arrived at the hospital at 22 and 72 hours respectively after onset of symptoms.
Each of Figs. 3-10 is a graphical illustration of the data obtained from a different patient of the study. The concentration levels are expressed as multiples of a reference value and were obtained by dividing the actual measured concentration values by the defined reference value for each respective marker protein, i.e., the 2SD
value.
All lacunar infarcts, hemorrhagic and TIA patients were identified on ~ 5 presentation with 100% accuracy. All five lacunar infarcts had elevated levels of Tm on presentation. In some patients the only elevated marker protein was Tm.
Referring now to Fig. 3 it can be seen that, for patient SM7, the only elevated marker protein was Tm indicating a lacunar infarct.
The three TIA patients had elevated NSE levels and normal S 100 and MBP
20 levels that stayed within the normal range. Tm was elevated in one of the TIA
patients. Referring now to Fig. 4 it can be seen that for patient SM-24, Tm was slightly elevated and NSE was elevated indicating a TIA. The patient was discharged with diagnosis of TIA. Referring now to Fig. 5 it can be seen that patient SM-3 had greatly elevated levels of MBP and 5100 as well as elevated levels of NSE and Tm 25 indicating a cerebral infarct with damage spreading into the base of the brain.
In the four hemorrhagic stroke patients, the three subarachnoid hemorrhagic patients had elevated levels of 5100 and NSE and a normal Tm level. In the patient with an intracerebral hemorrhagic stroke the levels of 5100 and NSE were elevated and the level of MBP was elevated about 250 times. Fig. 6 illustrates that patient SJ-30 16 had a 250 fold increased level of MBP upon presentation as well as elevated levels of S 100 and NSE and had suffered an intracerebral hemorrhage.

Fig. 7 illustrates that patient SJ-2 had elevated MBP, Tm and S 100 upon presentation and that the MBP and S100 levels continued to increase with time indicating a cerebral infarct with the stroke increasing over times An initial CAT scan upon presentation was negative and became positive only days later.
Fig. 8 illustrates that patient SJ-18 presented with a TIA which evolved into a stroke. Tm was in the normal range indicating that the cerebral vasculature was not compromised and thus indicating that the patient was a good candidate for thrombolysis.
Fig. 9 illustrates that patient SM-8 presented with a cerebral infarct and, with so Tm in the normal range, was a good candidate for thrombolysis since the endothelial vasculature was not compromised.
Fig. 10 illustrates that patient SJ-1 had a cerebral infarct and because of the elevated Tm level was at risk of hemorrhage if given thrombolytics because of the endothelial vasculature being compromised.
~ 5 For the second serum sample obtained the levels of 5100 were elevated in 73% of the stroke patients, the NSE levels in 54%, MBP levels in 64% and Tm levels in 55%. These data indicated that by measuring the four marker proteins in accordance with the invention, where any one marker was elevated 96% of the patients could be identified from the second serum sample obtained.
2o The data indicate that the levels of the protein markers in subsequent serum samples either increased or decreased depending upon whether the stroke was evolving in severity or subsiding.
Eighteen (54%) of the thirty three stroke patients had a CAT scan performed on presentation. All four hemorrhagic stroke patients were CAT positive at 25 presentation. Nine (50%) of the eighteen patients had a normal CAT at presentation which became positive days later. Eight of these nine patients who had a normal CAT
on presentation had elevated levels of one or more of the four protein markers on presentation. All of the nine CAT positive patients on presentation also had elevated levels of one or more protein markers on presentation.

Peak S100, NSE and MBP levels were significantly correlated (Pearson's) with admission NIHSS scores (p <0.05) and discharge modified Rankin scores (p <0.05).
i The data show that levels of 5100, NSE, MBP and Tm can be easily and reliably measured in acute ischemic stroke patients and that by measuring these four marker proteins in accordance with the invention, when any one marker protein is elevated a 94% sensitivity for acute ischemic stroke can be achieved upon presentation. Further, in the hyperacute period of the evolving stroke, elevated levels of one or more of these four marker proteins appear to precede irreversible tissue 1o damage and brain edema prior to detection of such damage by CAT.
Although the invention has been described with respect to various preferred embodiments it is not intended to be limited thereto but rather those skilled in the art will recognize that variations and modifications may be made therein which are within the spirit of the invention and the scope of the appended claims.

Claims (33)

1. A method for diagnosing the occurrence of an ischemic cerebral event or a hemorrhagic cerebral event and for distinguishing the type of cerebral event which has occurred, said method comprising a. analyzing a body fluid of a patient to detect the presence and concentration of four markers of cerebral event wherein i. a first marker is myelin basic protein, ii. a second marker is the beta isoform of S 100 protein, iii. a third marker is neuronal specific enolase, iv. a fourth marker is a brain endothelial cell membrane protein, and b. comparing said concentration of each marker detected to specific threshold values to determine the presence of statistically significant concentrations thereof, wherein the presence of said markers in statistically significant concentrations is indicative that an ischemic cerebral event or a hemorrhagic cerebral event has occurred and differentiates which type of cerebral event has occurred.
2. A method as defined in claim 1 wherein said body fluid is selected from the group consisting of blood, blood products and cerebrospinal fluid.
3. A method as defined in claim 1 wherein each of said analyses is carried out on the same sample of body fluid.
4. A method as defined in claim 1 wherein at least one of said analyses is carried out on a first sample of body fluid and at least another of said analyses is carried out on a second sample of body fluid.
5. A method as defined in claim 4 wherein said first and said second samples of body fluid are taken at different time periods.
6. A method as defined in claim 1 wherein said brain endothelial cell membrane protein is selected from the group consisting of Thrombomodulin, Glucose Transporter 1 in the dimeric or tetrameric form, Neurothelin/HT7, Gamma Glutamyl Transpeptidase, P-glycoprotein and combinations thereof.
7. A method as defined in claim 1 wherein at least one of said analyses comprises contacting said body fluid with an antibody which is specific for said marker.
8. A method as defined in claim 7 wherein at least one of said analyses is carried out with an enzyme-labeled immunoassay method.
9. A method as defined in claim 1 and further comprising the step of analyzing said body fluid for a fifth marker protein, wherein said fifth marker protein has the same specific cell type as one of said first, second or third markers and has a higher molecular weight than said first, second or third marker which has the same specific cell type.
10. A method as defined in claim 9 wherein at least one of said analyses comprises contacting said body fluid with an antibody which is specific for said marker.
11. A method as defined in claim 10 wherein at least one of said analyses is carried out with an enzyme-labeled immunoassay method.
12. A method as defined in claim 1 and further comprising the step of analyzing a second sample of body fluid from said patient for said four markers, said second sample of body fluid being taken at a time subsequent to said body fluid analyzed in step a.
13. A method as defined in claim 1 wherein said steps of determining whether an ischemic or hemorrhagic cerebral event has occurred and differentiating which particular type of cerebral event has occurred comparing the concentration level detected in said analysis for each of said four markers to a predefined threshold level for each said marker.
14. A diagnostic kit for diagnosing the occurrence of an ischemic cerebral event or a hemorrhagic cerebral event and distinguishing the type of cerebral event which has occurred comprising at least four antibodies which are specific for each of four different marker proteins, said antibodies immobilized on a solid support, wherein a. a first marker protein is myelin basic protein and a first antibody is specific therefor, b. a second marker protein is the beta isoform of 5100 protein and a second antibody is specific therefor, a third marker protein is neuronal specific enolase and a third antibody is specific therefor, and d. a fourth marker protein is a brain endothelial cell membrane protein and a fourth antibody is specific therefor and at least four labeled antibodies, each of said labeled antibodies binding to one of said marker proteins, and means for comparing said four marker proteins to specific threshold values of each of the marker proteins to determine the presence of statistically significant concentrations thereof, wherein the presence of said marker protein(s) in statistically significant concentrations is indicative that a cerebral event has occurred and differentiates which type of cerebral event has occurred.
15. A diagnostic kit as defined in claim 14 wherein each of said four antibodies is immobilized on the same solid support.
16. A diagnostic kit as defined in claim 14 wherein at least one of said four antibodies is immobilized on a first solid support and at least another of said four antibodies is immobilized on a second solid support.
17. A diagnostic kit as defined in claim 14 wherein at least one of said labeled antibodies comprises an enzyme-labeled antibody.
18. A diagnostic kit as defined in claim 14 wherein said brain endothelial cell marker protein is selected from the group consisting of Thrombomodulin, Glucose Transporter 1 in the dimeric or tetrameric form, Neurothelin/HT7, Gamma Glutamyl Transpeptidase, P-glycoprotein and combinations thereof.
19. A diagnostic kit as defined in claim 14 and further comprising a fifth antibody which is specific for a fifth masker protein, wherein said fifth marker protein has the same specific cell type as one of said first, second or third markers and has a higher molecular weight than said first, second or third marker which has the same specific cell type, and a fifth labeled antibody which binds to said fifth marker protein.
20. A diagnostic kit as defined in claim 19 wherein said fifth labeled antibody comprises an enzyme-labeled antibody.
21. A method for the differential diagnosis of ischemic and hemorrhagic cerebral events comprising a. analyzing a body fluid of a patient to detect the presence and concentration level of one or more ischemic marker proteins selected from the group consisting of myelin basic protein, the beta isoform of S 100 protein, neuronal specific enolase and combinations thereof, b. analyzing a body fluid of said patient to detect the presence and concentration level of a brain endothelial cell membrane protein, and c. comparing said concentration of each protein detected in steps a and b to specific threshold values to determine the presence of statistically significant concentrations thereof, wherein the presence of said proteins in statistically significant concentrations is indicative that an ischemic cerebral event or a hemorrhagic cerebral event has occurred and differentiates which type of cerebral event has occurred.
22. A method as defined in claim 21 wherein said steps of determining whether an ischemic or hemorrhagic cerebral event has occurred and differentiating which particular type of cerebral event has occurred comprise comparing the concentration levels detected in said analyses for said one or more ischemic marker proteins and for said brain endothelial cell membrane protein to a predefined threshold level for each said ischemic marker protein and for said brain endothelial cell membrane protein.
23. A method as defined in claim 21 wherein said body fluid is selected from the group consisting of blood, blood products and cerebrospinal fluid.
24. A method as defined in claim 21 wherein said brain endothelial cell membrane protein is selected from the group consisting of Thrombomodulin, Glucose Transporter 1 in the dimeric or tetrameric form, Neurothelin/HT7, Gamma Glutamyl Transpeptidase, P-glycoprotein and combinations thereof.
25. A method as defined in claim 24 wherein said brain endothelial cell membrane protein is Thrombomodulin.
26. A method as defined in claim 21 further comprising analyzing said body fluid to detect the presence and concentration level of a secondary marker protein having the same specific cell type as one of said myelin basic protein, beta isoform of S100 protein or neuronal specific enolase whereby the time of onset of a hemorrhagic or ischemic cerebral event can be determined.
27. A method as defined in claim 26 wherein said secondary marker protein has a higher molecular weight than said myelin basic protein, beta isoform of S100 protein or neuronal specific enolase which has the same specific cell type.
28. A method as defined in claim 21 wherein each of said analyses is carried out on a single sample of body fluid.
29. A method as defined in claim 21 wherein at least one of said analyses is carried out on a first sample of body fluid and at least another of said analyses is carried out on a second sample of body fluid.
30. A method as defined in claim 29 wherein said first and said second samples of body fluid are taken at different time periods.
31. A method as defined in claim 21 wherein a plurality of samples of said body fluid are obtained at predefined time intervals and analyzed and the information from said analyses compared as a function of time whereby the progression of an ischemic or hemorrhagic cerebral event can be determined.
32. A method as defined in claim 21 wherein each of said analyses comprises contacting said body fluid with an antibody which is specific for said protein.
33. A method as defined in claim 32 wherein at least one of said analyses is carried out with an enzyme-labeled immunoassay method.
CA002263063A 1999-02-26 1999-02-26 Method for diagnosing and distinguishing stroke and diagnostic devices for use therein Expired - Fee Related CA2263063C (en)

Priority Applications (18)

Application Number Priority Date Filing Date Title
CA002263063A CA2263063C (en) 1999-02-26 1999-02-26 Method for diagnosing and distinguishing stroke and diagnostic devices for use therein
DK00906097T DK1155325T3 (en) 1999-02-26 2000-02-22 Procedures for diagnosing and distinguishing types of stroke
AU27884/00A AU765923B2 (en) 1999-02-26 2000-02-22 Method for diagnosing and distinguishing stroke
PCT/CA2000/000182 WO2000052476A1 (en) 1999-02-26 2000-02-22 Method for diagnosing and distinguishing stroke
PT00906097T PT1155325E (en) 1999-02-26 2000-02-22 METHOD FOR DIAGNOSIS AND DISTINGUISH CEREBRAL VASCULAR ACCIDENTS
EP00906097A EP1155325B1 (en) 1999-02-26 2000-02-22 Method for diagnosing and distinguishing stroke
BR0008317-8A BR0008317A (en) 1999-02-26 2000-02-22 Process to diagnose and distinguish stroke
EP04027808A EP1521083A3 (en) 1999-02-26 2000-02-22 Method for diagnosing and distinguishing stroke
US09/510,700 US6235489B1 (en) 1999-02-26 2000-02-22 Method for diagnosing and distinguishing stroke and diagnostic devices for use therein
NZ513005A NZ513005A (en) 1999-02-26 2000-02-22 Method for diagnosing and distinguishing stroke
JP2000602637A JP4560212B2 (en) 1999-02-26 2000-02-22 Methods for diagnosing and distinguishing stroke
CN00803524.5A CN1339108A (en) 1999-02-26 2000-02-22 Method for diagnosing and distinguishing stroke
AT00906097T ATE283485T1 (en) 1999-02-26 2000-02-22 METHOD FOR DIAGNOSIS AND CHARACTERIZATION OF STROKE
ES00906097T ES2232424T3 (en) 1999-02-26 2000-02-22 METHOD FOR DIAGNOSING AND DISTINGUISHING AN ICTUS.
DE60016178T DE60016178T2 (en) 1999-02-26 2000-02-22 METHOD FOR DIAGNOSIS AND CHARACTERIZATION OF STROKE
US09/621,592 US6780606B1 (en) 1999-02-26 2000-07-21 Method for diagnosing and distinguishing stroke and diagnostic devices for use therein
US10/924,283 US7655424B2 (en) 1999-02-26 2004-08-23 Method for diagnosing and distinguishing stroke and diagnostic devices for use therein
US12/698,032 US7955811B2 (en) 1999-02-26 2010-02-01 Method for diagnosing and distinguishing stroke and diagnostic devices for use therein

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CA002263063A CA2263063C (en) 1999-02-26 1999-02-26 Method for diagnosing and distinguishing stroke and diagnostic devices for use therein

Publications (2)

Publication Number Publication Date
CA2263063A1 CA2263063A1 (en) 2000-08-26
CA2263063C true CA2263063C (en) 2004-08-10

Family

ID=4163334

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002263063A Expired - Fee Related CA2263063C (en) 1999-02-26 1999-02-26 Method for diagnosing and distinguishing stroke and diagnostic devices for use therein

Country Status (14)

Country Link
US (1) US6235489B1 (en)
EP (2) EP1521083A3 (en)
JP (1) JP4560212B2 (en)
CN (1) CN1339108A (en)
AT (1) ATE283485T1 (en)
AU (1) AU765923B2 (en)
BR (1) BR0008317A (en)
CA (1) CA2263063C (en)
DE (1) DE60016178T2 (en)
DK (1) DK1155325T3 (en)
ES (1) ES2232424T3 (en)
NZ (1) NZ513005A (en)
PT (1) PT1155325E (en)
WO (1) WO2000052476A1 (en)

Families Citing this family (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE19837387A1 (en) 1998-08-18 2000-02-24 Aventis Res & Tech Gmbh & Co New nucleic acid analogs useful in therapeutic or diagnostic agents or electronic components, consist of 3'-deoxypentopyranosyl nucleosides
US7070937B1 (en) 1998-10-02 2006-07-04 Ischemia Technologies, Inc. Marker useful for detection and measurement of free radical damage and method
US7449338B2 (en) 1998-10-02 2008-11-11 Ischemia Technologies, Inc. Tests for the rapid evaluation of ischemic states and kits
US20030215952A1 (en) * 1998-10-02 2003-11-20 Ischemia Technologies, Inc. Tests for the rapid evaluation of ischemic states and kits
US6475743B1 (en) 1998-10-02 2002-11-05 Ischemia Technologies, Inc. Marker useful for detection and measurement of free radical damage and method
US20030215359A1 (en) * 1998-10-02 2003-11-20 Ischemia Technologies, Inc. Tests for the rapid evaluation of ischemic states and kits
US20050142613A1 (en) * 1998-10-02 2005-06-30 David Bar-Or Test for the rapid evaluation of ischemic states and kits
US6780606B1 (en) 1999-02-26 2004-08-24 Synx Pharma, Inc. Method for diagnosing and distinguishing stroke and diagnostic devices for use therein
CA2263063C (en) * 1999-02-26 2004-08-10 Skye Pharmatech Incorporated Method for diagnosing and distinguishing stroke and diagnostic devices for use therein
WO2002082970A2 (en) * 2001-04-06 2002-10-24 The Research Foundation Of The City University Of New York Diagnosis and treatment of neural disease and injury using microvoltammetry
US20070026440A1 (en) 2001-04-06 2007-02-01 Broderick Patricia A Identification, diagnosis, and treatment of neuropathologies, neurotoxicities, tumors, and brain and spinal cord injuries using electrodes with microvoltammetry
US20030219734A1 (en) * 2001-04-13 2003-11-27 Biosite Incorporated Polypeptides related to natriuretic peptides and methods of their identification and use
US7713705B2 (en) 2002-12-24 2010-05-11 Biosite, Inc. Markers for differential diagnosis and methods of use thereof
US20040176914A1 (en) * 2001-04-13 2004-09-09 Biosite Incorporated Methods and compositions for measuring biologically active natriuretic peptides and for improving their therapeutic potential
US7524635B2 (en) * 2003-04-17 2009-04-28 Biosite Incorporated Methods and compositions for measuring natriuretic peptides and uses thereof
US20030199000A1 (en) * 2001-08-20 2003-10-23 Valkirs Gunars E. Diagnostic markers of stroke and cerebral injury and methods of use thereof
US6451547B1 (en) * 2001-04-25 2002-09-17 Syn X Pharma Process for differential diagnosis of Alzheimer's dementia and device therefor
US20020160420A1 (en) * 2001-04-30 2002-10-31 George Jackowski Process for diagnosis of physiological conditions by characterization of proteomic materials
CA2414073A1 (en) 2001-05-04 2002-11-14 Gunars E. Valkirs Diagnostic markers of acute coronary syndromes and methods of use thereof
WO2002089656A2 (en) * 2001-05-04 2002-11-14 Ischemia Technologies, Inc. Diagnosis of acute myocardial infarction
JP4268347B2 (en) * 2001-05-30 2009-05-27 富士フイルム株式会社 Control serum for dry analytical elements
US7608406B2 (en) * 2001-08-20 2009-10-27 Biosite, Inc. Diagnostic markers of stroke and cerebral injury and methods of use thereof
US20040209307A1 (en) * 2001-08-20 2004-10-21 Biosite Incorporated Diagnostic markers of stroke and cerebral injury and methods of use thereof
EP1419388B1 (en) * 2001-08-20 2009-10-07 Biosite Incorporated Diagnostic markers of stroke and cerebral injury and methods of use thereof
US20040219509A1 (en) * 2001-08-20 2004-11-04 Biosite, Inc. Diagnostic markers of stroke and cerebral injury and methods of use thereof
US20030040660A1 (en) * 2001-08-27 2003-02-27 George Jackowski Method for diagnosing and distinguishing traumatic brain injury and diagnostic devices for use therein
WO2003046556A2 (en) * 2001-11-23 2003-06-05 Syn.X Pharma, Inc. Globin biopolymer markers indicative of insulin resistance
US20050153360A1 (en) * 2002-02-18 2005-07-14 Hochstrasser Denis F. Diagnostic method for stroke
GB0203768D0 (en) * 2002-02-18 2002-04-03 Univ Geneve Diagnostic method for stroke
SE0200860D0 (en) * 2002-03-20 2002-03-20 Monica Almqvist Microfluidic cell and method for sample handling
US7115103B2 (en) * 2002-08-31 2006-10-03 Peter Trexler Keith Stroke symptom recognition devices and methods
EP1867734A1 (en) * 2002-12-24 2007-12-19 Biosite Incorporated Markers for differential diagnosis and methods of use thereof
JP2006526140A (en) * 2002-12-24 2006-11-16 バイオサイト インコーポレイテッド Marker for differential diagnosis and method of using the same
EP1601795A4 (en) * 2003-03-07 2008-03-26 Threshold Pharmaceuticals Inc Method for determining susceptibility of tumor to treatment with anti-neoplastic agent
GB2404981A (en) * 2003-08-15 2005-02-16 Univ Geneve Diagnostic method for stroke
EP1519194A1 (en) 2003-09-24 2005-03-30 Roche Diagnostics GmbH Use of gfap for identification of intracerebral hemorrhage
CN100373820C (en) * 2003-10-08 2008-03-05 松下电器产业株式会社 Road-vehicle communication system, and roadside apparatus, mobile apparatus which are used for the same
US8148057B2 (en) 2005-06-21 2012-04-03 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services, Centers For Disease Control And Prevention Methods, immunoassays and devices for detection of anti-lipoidal antibodies
US20100221740A1 (en) * 2005-06-21 2010-09-02 Castro Arnold R Methods, immunoassays and devices for detection of anti-lipoidal antibodies
GB2428240A (en) * 2005-07-14 2007-01-24 Univ Gen Ve Diagnostic method for brain damage-related disorders
DE102007004991B4 (en) 2006-08-14 2018-08-30 Varicula Biotec GmbH Use of RiV preparations for the treatment of cerebral infarction
DE102007026910A1 (en) 2007-06-12 2008-12-18 Siemens Ag Examination unit with integrated mini laboratory analysis unit
WO2008155718A2 (en) 2007-06-20 2008-12-24 Koninklijke Philips Electronics N.V. Detecting haemorrhagic stroke in ct image data
DE102007042986A1 (en) 2007-09-10 2009-04-23 Siemens Ag Medical device and method for operating a medical device
RU2491291C2 (en) * 2009-02-16 2013-08-27 Общество С Ограниченной Ответственностью "Герофарм" Modified peptide and using it for cns cancer screening and therapeutic effectiveness tests
US20100228101A1 (en) * 2009-03-04 2010-09-09 Michael Maschke Workflow for stroke diagnosis combining in-vitro diagnosis (ivd) with an imaging modality
US20100331673A1 (en) * 2009-06-26 2010-12-30 Michael Maschke System and method for reducing patient risk of allergic reaction to contrast agents or medical material
US20100332254A1 (en) * 2009-06-26 2010-12-30 Michael Maschke In-vitro device support for x-ray based kidney function test
US20100331833A1 (en) * 2009-06-26 2010-12-30 Michael Maschke In-vitro device monitoring during minimally invasive ablation therapy
GB201008541D0 (en) * 2010-05-21 2010-07-07 Univ Geneve Diagnostic methods
WO2012137832A1 (en) 2011-04-05 2012-10-11 オリンパス株式会社 Pancreas test method, and pancreas test kit
EP2825893A4 (en) 2012-03-13 2016-03-16 Univ Johns Hopkins Citrullinated brain and neurological proteins as biomarkers of brain injury or neurodegeneration
US9562911B2 (en) * 2015-01-30 2017-02-07 Cyrex Laboratories, Llc Simultaneous characterization of IgG and IgA antibodies to multiple food antigens and C1Q-food protein immune complexes
CN113970640B (en) * 2020-07-24 2022-07-08 首都医科大学附属北京天坛医院 Biomarkers for ICH prognosis evaluation and uses thereof
CN117054669A (en) * 2023-10-11 2023-11-14 北京美联泰科生物技术有限公司 Diagnostic or prognostic markers, products and methods for acute ischemic stroke

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2825283B2 (en) * 1989-08-21 1998-11-18 日本臓器製薬株式会社 Vascular disorder judgment method
JPH0736019B2 (en) * 1989-12-27 1995-04-19 富士薬品工業株式会社 Method for quantifying human thrombomodulin and its degradation products by a novel anti-human thrombomodulin monoclonal antibody
JP3057093B2 (en) * 1990-09-30 2000-06-26 日本臓器製薬株式会社 Cell injury defense ability measurement method
US5710008B1 (en) * 1990-10-12 1999-09-07 Spectral Diagnostics Inc Method and device for diagnosing and distinguishing chest pain in early onset thereof
US5604105B1 (en) * 1990-10-12 1999-08-24 Spectral Diagnostics Inc Method and device for diagnosingand distinguishing chest pain in early onset thereof
CA2027434C (en) * 1990-10-12 1999-01-05 George Jackowski Diagnostic kit for diagnosing and distinguishing chest pain in early onset thereof
RU1802337C (en) * 1990-12-21 1993-03-15 Центральный Военный Клинический Госпиталь Им.А.А.Вишневского Method for determination of volume of cerebral infarction during ischemic insult
JPH05219982A (en) * 1992-02-07 1993-08-31 Toshiba Corp Monoclonal antibody to protein distributing in brain
JP3073667B2 (en) * 1995-06-01 2000-08-07 オリエンタル酵母工業株式会社 A new marker for stroke
JPH09322783A (en) * 1996-04-05 1997-12-16 Takeda Chem Ind Ltd New protein and its dna
SE9602677D0 (en) 1996-07-05 1996-07-05 Sangtec Medical Ab Methods for determining brain antigens
CA2263063C (en) * 1999-02-26 2004-08-10 Skye Pharmatech Incorporated Method for diagnosing and distinguishing stroke and diagnostic devices for use therein
US20030199000A1 (en) * 2001-08-20 2003-10-23 Valkirs Gunars E. Diagnostic markers of stroke and cerebral injury and methods of use thereof
EP1419388B1 (en) * 2001-08-20 2009-10-07 Biosite Incorporated Diagnostic markers of stroke and cerebral injury and methods of use thereof

Also Published As

Publication number Publication date
ES2232424T3 (en) 2005-06-01
JP2002538463A (en) 2002-11-12
EP1155325B1 (en) 2004-11-24
EP1521083A3 (en) 2008-06-25
AU2788400A (en) 2000-09-21
BR0008317A (en) 2002-01-29
US6235489B1 (en) 2001-05-22
CN1339108A (en) 2002-03-06
NZ513005A (en) 2003-07-25
JP4560212B2 (en) 2010-10-13
DE60016178T2 (en) 2005-12-01
CA2263063A1 (en) 2000-08-26
EP1155325A1 (en) 2001-11-21
AU765923B2 (en) 2003-10-02
DK1155325T3 (en) 2005-04-04
PT1155325E (en) 2005-03-31
ATE283485T1 (en) 2004-12-15
WO2000052476A1 (en) 2000-09-08
DE60016178D1 (en) 2004-12-30
EP1521083A2 (en) 2005-04-06

Similar Documents

Publication Publication Date Title
CA2263063C (en) Method for diagnosing and distinguishing stroke and diagnostic devices for use therein
US7955811B2 (en) Method for diagnosing and distinguishing stroke and diagnostic devices for use therein
US20120149042A1 (en) Methods for predicting outcome in traumatic brain injury
JP4523587B2 (en) Method for distinguishing between type A and type B acute aortic dissection and acute myocardial infarction and kit for differentiation
JP2004527754A (en) Differential diagnostic processing of Alzheimer&#39;s dementia and apparatus therefor
EP1899726B1 (en) Methods for diagnosing and treating cerebrovascular events based on nr2 peptides
KR20180083909A (en) For the diagnosis or prediction of Alzheimer &amp;apos; s disease, or for the diagnosis of Parkinson &amp;apos;
US20230393154A1 (en) Method of treating large vessel occlusion stroke
US20210270848A1 (en) Method for diagnosing dementia or determining the risk of developing dementia
US20020137108A1 (en) CD45 isoform alteration in CD4+ T cells as a potential diagnostic marker in alzheimer&#39;s disease
RU2426130C2 (en) Instant diagnostic test for alzheimer&#39;s disease
MXPA01008092A (en) Method for diagnosing and distinguishing stroke
Buh et al. Serum Biomarker Concentrations on Admission in Acute Traumatic Brain Injury: Associations with, TBI Severity, Toxoplasma gondii Infection and Outcome in a Referral Hospital Setting, Cameroon
EP3730942A1 (en) Diagnosis markers for atrial fibrillation
EP3982123A1 (en) Markers and their use in brain injury
US20080188435A1 (en) Methods of dignosing or treating depression on basis of increased cerebrospinal fluid levels of neurotrodhin 3
Schwartz et al. Evaluation of the cardiac STATus™ CK-MB/Myoglobin Card Test to diagnose acute myocardial infarctions in the ED
WO2022093274A1 (en) Methods, systems, and kits for prediction, detection, monitoring, and treatment of alzheimer&#39;s disease
MXPA06004451A (en) Quick test for the diagnosis of alzheimer&#39;disease

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed