Biomarkers of vascular disease

Information

  • Patent Grant
  • 11821905
  • Patent Number
    11,821,905
  • Date Filed
    Wednesday, September 8, 2021
    2 years ago
  • Date Issued
    Tuesday, November 21, 2023
    5 months ago
  • Inventors
  • Original Assignees
  • Examiners
    • Bowers; Erin M.
    Agents
    • Haliday; Emily M.
    • Weaver Austin Villeneuve & Sampson LLP
Abstract
A biomarker panel including a four-panel test for clotting that detects soluble fibrin (SF), thrombin-antithrombin complex (TAT), antithrombin III (ATIII), and plasminogen activator inhibitor (PAI-1). A biomarker panel including a three-panel test for glycocalyx integrity that detects syndecan-1 (SDC1), heparan sulfate (HS), and hyaluronidase (HAD). A biomarker panel including a test that detects a biomarker chosen from soluble fibrin (SF), thrombin-antithrombin complex (TAT), antithrombin III (ATIII), plasminogen activator inhibitor (PAI-1), syndecan-1 (SDC1), heparan sulfate (HS), hyaluronidase (HAD), and combinations thereof. A kit including a biomarker panel, instructions for use, materials to take and apply samples to the panel, and descriptions of biomarker levels and their meaning. Methods of detecting the presence of vascular disease, determining the stage of vascular disease, monitoring the progress of vascular disease treatments, and monitoring the efficacy of drugs during drug development.
Description
BACKGROUND OF THE INVENTION
1. Technical Field

The present invention relates to biomarkers of vascular diseases or thromboembolism. More specifically, the present invention relates to panels of biomarkers that can detect the health status of blood vessels in the general population and in vascular diseases or thromboembolism.


2. Background Art

Chronic diseases are the leading cause of morbidity with thromboembolic diseases on top of the list. Thromboembolic disease is a blood vessel or vascular abnormality common to both arteries and veins. It involves formation of a blood clot, which can subsequently dislodge to form a thrombus or embolus that flows downstream in the blood vessel tree and impairs blood flow: a process called thrombosis. Thromboembolic disease is the leading cause of morbidity and mortality in the developed world with arterial thromboses being the most common underlying cause of coronary heart disease (CHD), acute myocardial infarction (MI), stroke, hypertension, atrial fibrillation, congestive heart failure (CHF), congenital heart condition, and peripheral arterial disease (PAD); and, venous thromboses, which include chronic venous insufficiency (CVI), deep venous thrombosis (DVT) affecting approximately two million Americans annually, and pulmonary embolism (PE) accounting for 60,000 annual deaths in the United States.


Current medical textbooks consider these vascular diseases as separate and different etiologies with treatment options including the cholesterol-lowering statins, anti-hypertensives, and anticoagulants. However, despite decades of statin and anti-hypertensive therapies, vascular diseases continue to be leading killers because these drugs are not addressing the root cause of thromboembolism. Advances in biochemistry now point to clot formation as common to the pathology of thromboembolism and the risk factors for clot formation include abnormalities in the blood vessel wall, blood flow dynamics, and blood constituents.


Blood clots or thrombosis is the cause of death in various vascular diseases. To understand the etiology of clot formation, one must look at blood circulation: an average human has 60-100,000 miles of blood vessels consisting of straight and bifurcated segments. The dynamics of blood flow through these vessels is unimpeded in the straight segments but slows down at bends or bifurcations forming a ‘whirlpool’ flow. Whirlpool blood flow alone could cause clot formation where overcrowding produce fibrinogen, which binds to the sialic acid on the surface of red blood cells (RBC) making them stick together and acquire a ‘Roleaux’ formation (stacking due to flat RBC surface). This Roleaux formation subsequently gets trapped by fibrin along with white blood cells and platelets to form native clots without vessel wall injury.


Conditions that increase blood viscosity further slow down blood flow to stagnation. Thus, excessive consumption of fats contributes to increased blood viscosity. Fats or triglycerides (TG) in the diet are packaged by the liver in very low density lipoprotein (VLDL—55% TG: 9% C) for delivery to muscles as energy source. Excess dietary fat means an increased VLDL level, causing hypertriglyceridemia if fat lingers or stays in the blood stream for an extended period; or obesity after VLDL unloads its fat cargo to adipocytes. Once TG is released from VLDL, the package is reduced and becomes LDL (7% TG: 50% C), which is the source of cholesterol for the body's need; excess LDL goes back to liver for recycling or for bile.


Stagnant blood flow concentrates blood contaminants (bacterial infections, pollutants, etc.), which attracts monocytes and white blood cells (WBC), subsequently provoking an inflammatory response with the release of inflammatory cytokines and free radicals or reactive oxygen species (ROS). Over time, chronic inflammation disrupts the protective endothelial glycocalyx lining, and creates ‘gaps’ and osmotic imbalance in the blood vessel resulting in edema and infiltration of blood debris, e.g., dead cells, chemicals, calcium, etc.


Moreover, the denuded endothelium produces a sticky or adhesive material called e-selectin, causing activated macrophages to accumulate to form sticky ‘foam cell’, which matures into plaque. The processes leading to plaque build-up or atheroma (atherosclerosis) generally begin in the early years of life, as young as 5 years old, but the symptoms generally do not become apparent until after the age of 40 years.


More importantly, the endothelial glycocalyx offers a ‘nest’ for protective enzymes including anticoagulant anti-thrombin (AT-III), anti-oxidant (SOD), and anti-high blood viscosity lipoprotein lipase (LpL). Thus, the loss of endothelial glycocalyx results in a build up of fibrin, a suppression of fibrinolysis and the promotion of clot formation. Further inflammation predisposes the plaque to rupture, and ruptured plaque triggers clots formation: this clot can be exacerbated by the seed clot formed by Roleaux cells thus becoming a significant thrombus. Loose thrombi can wedge on a rigid vessel narrowed by plaque, particularly in individuals who already have atherosclerosis, causing a stroke (clogged artery to the brain), heart attack (clogged artery to the heart), or PAD (clogged artery to the arms or legs).


Meanwhile, the straight vessel segment with high blood flow has high shear stress, which is important in maintaining a thick protective endothelial glycocalyx layer. The venous system returns ‘used’ blood to the heart by means of the skeletal pump. The mechanism involves the contraction-relaxation motion of muscles surrounding the veins (relaxation, draws blood from superficial veins; contraction, propels blood to heart) pushing blood through the one-way valve system (preventing back-flow). Poor muscle contraction or dysfunctional valve system, due to inactivity or muscle degeneration, create stagnation and blood pooling. Concentration of blood cells is the main mechanism for clot formation, although injury to vein wall may trigger formation of blood clot as well. A loose clot becomes an embolus, which travels to the lung or heart causing a DVT.


In summary, interruptions to arterial blood flow causes blood stagnation where ‘blood debris’ gravitates (e.g., dead cells, ‘microbial contaminants’, etc.). These attract monocytes, which produce inflammatory factors (e.g., cytokines, free radicals or ROS) and eventually chronic inflammation leads to destruction of the protective endothelial glycocalyx lining. Destruction of the endothelial glycocalyx creates an osmotic imbalance causing edema and infiltration of blood components resulting in plaque formation. More importantly, loss of endothelial glycocalyx results in the shedding of built-in protective enzymes including the anti-oxidant superoxide dismutase (SOD) and catalase, anticoagulant anti-thrombin AT-III, and ‘anti-viscosity’ lipoprotein lipase (LpL). All these factors lead to clot formation and thrombosis. Thrombosis is typically fatal when a clot wedges on a rigid or inelastic vessel, which is narrowed by a stenotic plaque.


On the venous system, a faulty ‘skeletal muscle-pump’ causes stagnation or blood pooling: concentrated blood cells are trapped in fibrin and subsequently activate platelets to form clots. Loose clots become emboli that travel back to heart or lung causing deadly deep vein thrombosis (DVT). Alternatively, pooled blood attract inflammatory monocytes that disrupts the endothelial glycocalyx causing protein infiltration (extravasation), edema and varicose or spider veins.


There are several treatments available for blood clots, i.e. antithrombotics. These treatments fall into two classes, anticoagulants (that slow down clotting) and antiplatelets (that prevent clumping of platelets and clot formation). Heparin is an anticoagulant that is commonly used and administered before surgery in order to prevent clots forming. Heparin has the side effect of causing excessive bleeding and bruising and cannot be used on a long term basis. Dicumarol, and its derivative warfarin, are other anticoagulants that act as a vitamin K depleter and prevent the formation of coagulant enzymes. Fondaparinux is a synthetic anticoagulant related to low molecular weight heparin that is indicated for deep vein thrombosis and can be given daily. Bivalirudin is a synthetic anticoagulant that is a specific and reversible direct thrombin inhibitor and is indicated for patients with unstable angina undergoing percutaneous transluminal coronary angioplasty or percutaneous coronary intervention. Aspirin is an antiplatelet and widely used in low doses to reduce the risk of stroke and heart attack. Clopidogrel is an antiplatelet that irreversibly inhibits P2Y12 receptor on platelet cell membranes and can be administered along with or instead of aspirin in preventing heart disease. Dipyridamole is an antiplatelet that inhibits thrombus formation, and is also administered along with aspirin to prevent stroke and heart attack. Abciximab is another antiplatelet administered to patients undergoing percutaneous coronary intervention. While each of these antithrombotics are useful in different situations, there are still side effects and there remains a need for more effective therapeutics and diagnostics in detecting risk of blood clots.


There are several tests that can be performed to determine if an individual has vascular disease. For example, a carotid duplex is an ultrasound scan of the carotid artery that can detect stenosis or narrowing of the carotid arteries. An abdominal duplex is an ultrasound scan of the aorta that can detect abnormalities. An ankle brachial index can detect reduced blood flow in the leg. Venous ultrasound can be used to detect blood flow through vessels for detecting deep vein thrombosis and superficial vein thrombosis.


Biomarkers have been identified that can be useful for identifying individuals at risk of vascular diseases. For example, biomarkers of inflammation can indicate the presence of atherosclerosis or plaques (C-reactive protein, IL-18, IL-6). Biomarkers of lipid accumulation can indicate the presence of plaques (lipoprotein-associated phospholipase A2). Biomarkers of thrombosis can indicate the presence of plaque instability or carotid disease progression (tissue plasminogen activator (t-PA), fibrinogen, plasminogen activator inhibitor-1 (PAI-1)). However, no such biomarkers are currently in use by medical practitioners as a diagnostic tool.


U.S. Patent Application No. 2007/0269836 to McPherson, et al. discloses methods and compositions for diagnosis of venous thromboembolic disease, pulmonary embolism, and/or deep vein thrombosis, and for risk stratification in such conditions. An assay can be performed from test samples obtained from a subject to diagnose a subject, including markers used individually or in combination, such as TAT, ATIII, and PAI-1. McPherson, et al. does not disclose an assay including the marker soluble fibrin (SF).


U.S. Pat. No. 8,759,095 to Vink, et al. discloses diagnostic and therapeutic tools for diseases altering vascular function. In particular, endothelial glycocalyx perturbation can be diagnosed in samples from subjects by detecting heparan sulfate (HS) (heparan sulphate therein), hyaluronidase (HAD), and syndecan-1. Vink, et al. does not provide any particular advantages to using these particular three markers together but merely provides them as examples of relevant markers.


U.S. Patent Application No. 2013/0273096 to Daniels discloses methods of treating disorders affecting the endothelial glycocalyx. Characteristics of the endothelial glycocalyx can be determined by detecting markers in a sample from a subject, such as heparan sulfate (HS), hyaluronidase (HAD), and syndecan-1. Daniels does not provide any particular advantages to using these particular three markers together but merely provides them as examples of relevant markers.


As vascular diseases remain a large problem with society, there remains a need for a diagnostic device that can detect diseases accurately as well as indicate the stage of disease.


SUMMARY OF THE INVENTION

The present invention provides for a biomarker panel including a four-panel test for clotting that detects soluble fibrin (SF), thrombin-antithrombin complex (TAT), antithrombin III (ATIII), and plasminogen activator inhibitor (PAI-1).


The present invention provides for a biomarker panel including a three-panel test for endothelial glycocalyx integrity that detects syndecan-1 (SDC1), heparan sulfate (HS), and hyaluronidase (HAD).


The present invention provides for a biomarker panel including a test that detects a biomarker chosen from soluble fibrin (SF), thrombin-antithrombin complex (TAT), antithrombin HI (ATM), plasminogen activator inhibitor (PAI-1), syndecan-1 (SDC1), heparan sulfate (HS), hyaluronidase (HAD), and combinations thereof.


The present invention provides for a kit including a biomarker panel chosen from the four-panel test, the three-panel test, both the four-panel and three panel tests, a test that detects a biomarker chosen from soluble fibrin (SF), thrombin-antithrombin complex (TAT), antithrombin III (ATIII), plasminogen activator inhibitor (PAI-1), syndecan-1 (SDC1), heparan sulfate (HS), hyaluronidase (HAD), and combinations thereof, instructions for use, materials to take and apply samples to the panel, and descriptions of biomarker levels and their meaning.


The present invention also provides for a method of detecting the presence of vascular disease or propensity to develop vascular disease, by taking a sample, applying the sample to a biomarker panel that detects clotting, endothelial glycocalyx integrity, or combinations thereof, detecting the presence of at least one biomarker, comparing levels of the biomarker to a baseline, and determining if the individual has vascular disease or the propensity to develop vascular disease.


The present invention further provides for a method of determining the stage of vascular disease, by taking a sample, applying the sample to a biomarker panel that detects clotting, endothelial glycocalyx integrity, or combinations thereof, detecting the presence of at least one biomarker, comparing levels of the biomarker to known stage levels, and determining the stage of vascular disease.


The present invention provides for a method of monitoring the progress of vascular disease treatments by taking a sample from an individual receiving treatment for vascular disease, applying the sample to a biomarker panel chosen from a panel that detects clotting, endothelial glycocalyx integrity, or combinations thereof, detecting the presence of at least one biomarker, comparing levels of the biomarker to a baseline, and determining if the treatment is working to reverse or prevent vascular disease.


The present invention further provides for a method of monitoring the efficacy of drugs during drug development against cardiovascular diseases or other diseases involving inflammation, disruption of blood vessels, removal of plaques, or treatment of clot formation, by taking a sample from an individual receiving a drug being tested, applying the sample to a biomarker panel chosen from a panel that detects clotting, endothelial glycocalyx integrity, or combinations thereof, detecting the presence of at least one biomarker, comparing levels of the biomarker to a baseline, and determining the efficacy of the drug.


The present invention also provides for a method of detecting and monitoring vascular health by determining the status of internal vascular architecture.





DESCRIPTION OF THE DRAWINGS

Other advantages of the present invention are readily appreciated as the same becomes better understood by reference to the following detailed description when considered in connection with the accompanying drawings wherein:



FIGS. 1A and 1B are charts of experiment protocols;



FIGS. 2A and 2B are photomicrographs of sections of group 3 and group 4;



FIG. 3 is a graph of total plasminogen activation inhibitor-1 levels;



FIG. 4 is a graph of heparan sulfate levels;



FIG. 5 is a graph of hyaluronan synthase 1 levels;



FIG. 6 is a graph of syndecan-1 levels;



FIG. 7 is a graph of thrombin-anti-thrombin III levels;



FIG. 8 is a graph of anti-thrombin levels;



FIG. 9 is a graph of average disease scores for treatment groups and sacrifice times;



FIG. 10 is a graph of hyaluronan synthase levels;



FIG. 11 is a graph of heparan sulfate levels;



FIG. 12 is a graph of total plasminogen activation inhibitor-1 levels,



FIG. 13 is a graph of syndecan-1 levels; and



FIG. 14A is a photograph of plaque at 10×, FIG. 14B is a photograph of plaque at 40×, and FIG. 14C is a photograph of a normal arterial wall.





DETAILED DESCRIPTION OF THE INVENTION

Most generally, the present invention is directed to panels of biomarkers used to detect vascular diseases, and especially in detecting biomarkers that indicate abnormal biochemical elements responsible for the blood clotting cascade and biomarkers that indicate abnormal levels of enzymes and structural components of the blood vessel surface.


The term “assay” as used herein refers to a procedure that determines the amount of a particular constituent of a mixture or sample. “Assay” can interchangeably be used with the term “test” herein.


The term “biomarker” as used herein refers to a substance, such as, but not limited to, a protein, DNA sequence, RNA sequence, or other biological substance or substances that, when detected, indicates a particular healthy or unhealthy state of an individual with respect to vascular disease.


“Vascular disease”, as used herein, refers to any disease affecting the circulatory system of arteries, veins, capillaries, and lymph vessels in the body. Vascular disease can include, but is not limited to, peripheral artery disease, aneurysms, renal artery disease, Raynaud's disease, Buerger's disease, peripheral venous disease, varicose veins, blood clots (thromboembolism), blood clotting disorders, lymphedema.


“Thromboembolism”, as used herein, refers to a family of vascular diseases including coronary heart disease (CHD), acute myocardial infarction (MI), stroke, hypertension, atrial fibrillation, congestive heart failure (CHF), congenital heart condition, peripheral arterial disease (PAD), chronic venous insufficiency (CVI), deep venous thrombosis (DVT), and pulmonary embolism (PE).


The term “healthy” as used herein refers to a state of an individual who is free from vascular disease, is in good health, and has relatively low risk of developing vascular disease.


The term “sample” as used herein refers to a biological sample from an individual, and can be, but is not limited to, blood, plasma, urine, saliva, tears, or cerebral spinal fluid (CSF).


The present invention generally provides for a method of detecting and monitoring vascular health, by determining the status of internal vascular architecture. This status is determined by monitoring products of vascular oxidation damage and clot formation, as further described below.


Most generally, the biomarker panels include of a set of chemical, immunochemical and/or enzymatic assays or tests that can be used together for monitoring the levels of a set of biomarkers. The biomarker panels can be used to determine the presence of disease, or the propensity of an individual to develop disease. The biomarker panels can also be used to mark the progression of disease. Evaluation of different stages or components of vascular disease is important for intervention or reversal of the effects of the disease.


The biomarker panel can include a four-panel test for endothelial glycocalyx health that detects soluble fibrin (SF), thrombin-antithrombin complex (TAT), antithrombin III (ATIII), and plasminogen activator inhibitor (PAI-1).


Soluble fibrin (SF) is composed of fibrin monomer and fibrinogen derivatives, exist in the circulating blood in patients with thrombosis. Its detection and quantification are useful for obtaining information about the condition and degree of intravascular coagulation in early-stage thrombosis. The level of SF increases on coagulation, which is related to the production of blood factor VIII. Thus, factor VIII circulates in the plasma bound to von Willebrand factor (vWf). Thrombin cleaves and activates factor VIII and releases vWf. The vWf is then free to bind to ruptured endothelial cell surfaces where it activates platelet aggregation. The released FVIIIa acts as a cofactor of factor IXa to generate factor Xa. In the presence of Ca2+ and phospholipids, FX is activated to FXa by FIXa. Since FVIIIa is a cofactor to FIXa, it greatly stimulates the reaction. By using optimal amounts of Ca2+, phospholipid, and FIXa, and an excess of FX, the rate of activation of FX is linearly related to the amount of FVIII. FXa hydrolyses the chromogenic substrate S-2765 which releases the chromophoric group pNA. The color can be read at 405 nm, and generated FXa and thus the intensity of color, is proportional to the FVIII activity in the sample. A baseline level for soluble fibrin can be established that reflects the levels in a healthy individual. A healthy individual should have lower levels of SF than a diseased individual. If levels are detected with the biomarker panel that are above the baseline level, it can be determined that the individual has vascular disease or is at risk of developing vascular disease and especially thrombosis. Other levels can determine the stage or progression of vascular disease.


Another blood component that reflects blood coagulation is the formation of thrombin-antithrombin complex (TAT). TAT complex is a parameter of coagulation and fibrinolysis. Elevated concentrations have been associated with vascular disease. Antithrombin deficiency promotes clot formation in the arteries and/or veins and is associated with a high risk of thromboembolic disorders. An operational detection of TAT involves microtiter plates, which are commercially available. Thus, microtiter plates precoated with antibody specific to thrombin are commercially available. Calibrators or samples are then added to the appropriate microtiter plate wells with a biotin-conjugated polyclonal antibody preparation specific for ATIII. Next, Avidin conjugated to Horseradish Peroxidase (HRP) is added to each microplate well and incubated. Then a TMB substrate solution is added to each well. Only those wells that contain TAT, biotin-conjugated antibody and enzyme-conjugated Avidin will exhibit a change in color. The enzyme-substrate reaction is terminated by the addition of a sulfuric acid solution and the color change is measured spectrophotometrically at a wavelength of 450 nm±10 nm. The concentration of TAT in the samples is then determined by comparing the O.D. of the samples to the calibration curve. A baseline level for TAT can be established that reflects the levels in a healthy individual. A healthy individual should have lower levels of TAT than diseased individual. If levels are detected with the biomarker panel that are above the baseline level, it can be determined that the individual has vascular disease or is at risk of developing vascular disease. Other levels can determine the stage or progression of vascular disease.


Antithrombin III (AT III) is a non-vitamin K-dependent protease enzyme, which serves as a natural blood thinner and inhibits coagulation. AT III deficiency leads to increased risk of developing life-threatening clots that block blood flow. For example, deep vein thrombosis (DVT) occurs when a clot, or thrombus, develops in one of the deep veins, most common in the legs. The level of AT III is reduced when blood coagulates, which is determined by commercially available test kits. One such example of a test kit is the AssayMax Mouse AT III ELISA kit (supplier): this is designed for detection of mouse AT III in plasma, serum and cell culture supernatants. This assay employs a quantitative sandwich enzyme immunoassay technique, which measures AT III in 4 hours. Thus, a microliter plate pre-coated with polyclonal antibody specific for mouse AT III is commercially available. Mouse AT III in standards and samples is sandwiched by the immobilized antibody and biotinylated polyclonal antibody specific for mouse AT III, which is recognized by a streptavidin-peroxidase conjugate. All unbound material is then washed away and a peroxidase enzyme substrate is added. The color development is stopped and the intensity of the color is measured. A baseline level for AT III can be established that reflects the levels in a healthy individual. A healthy individual should have lower levels of AT III than a diseased individual. If levels are detected with the biomarker panel that are above the baseline level, it can be determined that the individual has vascular disease or is at risk of developing vascular disease. Other levels can determine the stage or progression of vascular disease.


Plasminogen activator inhibitor (PAI-1) is a protein, also known as endothelial plasminogen activator inhibitor or serpin E1, and is a central regulator of the blood fibrinolytic system and its production precedes thrombosis. In other words, increased PAI-1 levels increase the risk for thrombosis, whereas decreased levels cause recurrent bleeding. PAI-1 is the main inhibitor of the plasminogen activators and thus an important component of the coagulation system that down-regulates fibrinolysis. Reduced PAI-1 levels result in increased fibrinolysis and an associated bleeding diathesis. The other PAI, plasminogen activator inhibitor-2 (PAI-2), is secreted by the placenta and only present in significant amounts during pregnancy. Test kits for murine PAI-1 are available commercially: thus, free, latent or complex PAI-1 present in plasma reacts with the capture antibody coated and dried on a microtiter plate. Any unbound PAI-1 is washed away and an anti-PAI-1 primary antibody is added. Excess primary antibody is washed away and bound antibody, which is proportional to the total PAI-1 present in the samples, is then reacted with the secondary antibody. Following an additional washing step, TMB is then used for color development at 450 nm. The amount of color development is directly proportional to the concentration of total PAI-1 in the sample. A baseline level for PAI-1 can be established that reflects the levels in a healthy individual. A healthy individual should have lower levels of PAI-1 than a diseased individual. If levels are detected with the biomarker panel that are above the baseline level, it can be determined that the individual has vascular disease or is at risk of developing vascular disease. Other levels can determine the stage or progression of vascular disease.


The biomarker panel can alternatively include a three-panel test for endothelial glycocalyx integrity that detects syndecan-1 (SDC1), heparan sulfate (HS), and hyaluronidase (HAD).


Syndecans are transmembrane domain proteins that carry three to five heparan sulfate and chondroitin sulfate chains, which harbor a variety of important ligands including fibroblast growth factors, vascular endothelial growth factor, transforming growth factor-beta, fibronectin, and antithrombin-1. Syndecan-1 (SDC1) is a cell surface heparan sulfate proteoglycan, which is an important component of the protective endothelial glycocalyx lining the luminal surface of blood vessels. Key roles for SDC1 is in endothelial mechano-sensing and regulation of endothelial integrity and function. Shedding of syndecan-1 and heparan sulfate into the circulation is associated with inflammatory disease and atherosclerosis. Test kits for syndecan, for example test kit precoated with monoclonal antibody specific to SDC1 are available commercially. Thus, samples are then added to the appropriate microtiter plate wells with a biotin-conjugated polyclonal antibody preparation specific for SDC1. Next, Avidin conjugated to Horseradish Peroxidase (HRP) is added to each microplate well and incubated. Then a TMB substrate solution is added to each well. Only those wells that contain SDC1, biotin-conjugated antibody and enzyme-conjugated Avidin will exhibit a change in color. The enzyme-substrate reaction is terminated by the addition of a sulphuric acid solution and the color change is measured spectrophotometrically at a wavelength of 450 nm±10 nm. The concentration of SDC1 in the samples is then determined by comparing the O.D. of the samples to the standard curve. A baseline level for SDC1 can be established that reflects the levels in a healthy individual. A healthy individual should have lower levels of SDC1 than a diseased individual. If levels are detected with the biomarker panel that are above the baseline level, it can be determined that the individual has vascular disease or is at risk of developing vascular disease. Other levels can determine the stage or progression of vascular disease.


Heparan sulfates (HSs) are highly negatively charged polysaccharides with 1→4-linked sulfated glucosamine and uronic acid repeating disaccharide units. HSs are present on the cell surface as well as in the extracellular matrix and bind to proteins involved in anticoagulation, angiogenesis, microbial infection, and monocyte adhesion. HSs are glycoproteins with the common characteristic of containing one or more covalently attached chains, including syndecans and glycosylphosphatidylinositol-anchored proteoglycans (glypicans), the secreted extracellular matrix HSPGs (agrin, perlecan, type XVIII collagen), and the secretory vesicle proteoglycan, serglycin. HSs are implicated in the pathogenesis of atherosclerosis by their ability to trap plasma lipoproteins in the arterial wall and by their influence on cellular migration, adhesion and proliferation. Intact HS chains are anti-atherogenic. ELISA test kits for heparan sulfate are available commercially. The test includes pretreatment of serum with proteinase (actinase E) to digest serum proteins. One volume of dissolved Actinase E (20 mg/mL in actinase E dissolution buffer) can be added against ten volumes of serum and then mixed. Proteins can be digested at 55° C. for 16-20 hours in a water bath. After digestion, the mixture can be boiled for 5 minutes to stop digestion. After boiling, the mixture can be brought to room temperature (15-25° C.) and then centrifuged 3,000 rpm, for 10 minutes. After centrifugation, the supernatant can be taken and mixed well. The supernatant is applied to Heparan Sulfate ELISA kit. HS values can be calculated in pretreated samples according to the Heparan Sulfate ELISA kit procedure. The calculated HS values must be multiplied dilution factors as below to determine the HS concentration in serum. HS concentration=calculated HS value×dilution factor×1.1. A baseline level for HS can be established that reflects the levels in a healthy individual. A healthy individual should have lower levels of HS than a diseased individual. If levels are detected with the biomarker panel that are above the baseline level, it can be determined that the individual has vascular disease or is at risk of developing vascular disease. Other levels can determine the stage or progression of vascular disease.


Hyaluronic acid (HA, also called hyaluronan or hyaluronate) is a negatively charged, nonsulfated large linear glycosaminoglycan (a class of negatively charged polysaccharides) of repeating disaccharide structure D-Glucuronic acid (UDP-GlcA) and N-acetylglucosamine (UDP-GlcNac), which is a principal component of endothelial glycocalyx. HA is distributed widely throughout connective, epithelial, and neural tissues. HA is the simplest glycosaminoglycan that provides compression strength, lubrication and hydration. A disturbed HA is atherogenic. The removal of HA-rich glycocalyx with hyaluronidase is associated with increased vascular permeability leading to atherogenic insults. Increased plasma HA and hyaluronidase levels are found associated with endothelial glycocalyx damage, presence of microvascular diseases and carotid intima-media thickness. A coated well immunoenzymatic assay for the quantitative measurement of hyaluronidase (HAD) utilizes a polyclonal anti-HAD antibody and an HAD-HRP conjugate. The assay sample and buffer are incubated together with HAD-HRP conjugate in pre-coated plate for one hour. After the incubation period, the wells are decanted and washed five times. The wells are then incubated with a substrate for HRP enzyme. The product of the enzyme-substrate reaction forms a blue colored complex. Finally, a stop solution is added to stop the reaction, which will then turn the solution yellow. The intensity of color is measured spectrophotometrically at 450 nm in a microplate reader. The intensity of the color is inversely proportional to the HAD concentration since HAD from samples and HAD-HRP conjugate compete for the anti-HAD antibody binding site. Since the number of sites is limited, as more sites are occupied by HAD from the sample, fewer sites are left to bind HAD-HRP conjugate. Standards of known HAD concentrations are run concurrently with the samples being assayed and a standard curve is plotted relating the intensity of the color (O.D.) to the concentration of HAD. The HAD concentration in each sample is interpolated from this standard curve. A baseline level for HA can be established that reflects the levels in a healthy individual. A healthy individual should have lower levels of HA than a diseased individual. If levels are detected with the biomarker panel that are above the baseline level, it can be determined that the individual has vascular disease or is at risk of developing vascular disease. Other levels can determine the stage or progression of vascular disease.


The biomarker panel can also include any combination of the above-described biomarkers, i.e. they are not limited to being used in combination in just the three-panel test and four-panel test. For example, another preferred combination can include PAI-1, HS, and HAS-1 as a blood test that defines vascular leakage and clot onset to correlate with plaque formation. As shown in the examples below, these three biomarkers are highly correlative with plaque formation.


The biomarker panels can be used alone or in combination. For example, the biomarker panels can be used individually if there is a strong correlation established for any of the biomarkers in that particular panel, or the combination of biomarker panels can be used to ensure reliability.


In general, the biomarker panel can use a support structure such as a flat microwell plate (such as an ELISA plate) that has multiple wells to hold samples. Various enzymes or antibodies can be applied to the wells as needed for each test, such as those described above. A housing can enclose the biomarker panel to prevent contamination or unwanted spread of samples, in plastic or another suitable material.


The biomarker panels of the present invention can be included in a kit. The kit can include the biomarker panels (the four-panel test, the three-panel test, both the four-panel and three panel tests, or any combination of the above-described biomarkers) instructions for use, materials to take and apply samples to the panel (such as, but not limited to, swabs, syringes, or vials), and descriptions of biomarker levels and their meaning (such as normal values). The kit can include various antibodies as needed to detect the biomarkers.


The biomarker panels can be used to detect the presence of vascular disease or propensity to develop vascular disease in the following method. A sample is taken (preferably by a healthcare practitioner) and applied to the biomarker panel (either the four-panel test for clotting, the three-panel test for glycocalyx integrity, both as described above, or any combination of the above-described biomarkers). The sample reacts with various reagents in the panel based on the presence of the biomarkers described above. Preferably, the sample applied to the panel is sent to a lab for analysis. Results of the reaction can provide a colorimetric result and can be read in a colorimeter. Alternatively, any other method of detection and quantification can be used, such as, but not limited to, Western blot, immunoprecipitation, immunohistochemistry, Enzyme-linked immunosorbent assay (ELISA), Radio Immuno Assay (RIA), radioreceptor assay, proteomics methods (such as mass spectrometry), or quantitative immunostaining methods. If any or all of the biomarkers are detected, it can be determined if the individual has vascular disease or the propensity to develop vascular disease by comparing the biomarker levels to known baseline levels for healthy individuals. In other words, if levels of the biomarkers are detected that are above the baseline level, it can be determined that the individual has vascular disease or is at risk of developing vascular disease.


The biomarker panel can also be used to determine the stage of vascular disease in an individual (i.e. the progression of vascular disease in the individual). A sample is taken (preferably by a healthcare practitioner) and applied to the biomarker panel (either the four-panel test for clotting, the three-panel test for endothelial glycocalyx integrity, both as described above, or any combination of the above-described biomarkers). The sample reacts with various reagents in the panel based on the presence of the biomarkers described above. Preferably, the sample applied to the panel is sent to a lab for analysis. Results of the reaction can provide a colorimetric result and can be read in a colorimeter or by any other method described above. If any or all of the biomarkers are detected, it can be determined what stage of vascular disease the individual has by comparing the results to known stage levels. Based on the results of the stage of vascular disease, the individual can be proscribed medication that is appropriate for that particular stage.


The biomarker panels can also be used in a method of monitoring the efficacy of drugs during drug development against cardiovascular diseases or other diseases involving inflammation, disruption of blood vessels, removal of plaques, or treatment of clot formation. A sample can be taken (preferably by a healthcare practitioner) from an individual receiving a drug being tested. The sample can be applied to a biomarker panel (either the four-panel test for clotting, the three-panel test for endothelial glycocalyx integrity, both as described above, or any combination of the above-described biomarkers). The sample reacts with various reagents in the panel based on the presence of the biomarkers described above. Preferably, the sample applied to the panel is sent to a lab for analysis. Results of the reaction can provide a colorimetric result and can be read in a colorimeter or by any other method described above. If the presence of any or all of the biomarkers are detected, indicating the presence of inflammation, disruption of blood vessels, plaques, or clot formation (among any other symptoms described above), it can be determined if the drug is effective by comparing the results to known baseline levels for healthy individuals. A higher than baseline level would indicate that the drug has not been effective in treatment.


The biomarkers included in the panels of the present invention measure factors produced early on in the clot formation process. Therefore, each of these biomarkers alone are significant as well as together in the panels in predicting the initiation of the biologic process (oxidation and immunogenic and/or inflammatory process) that leads to the formation of the clots. Also, the “lipid panel” devised by the American Heart Association is not predictive of cardiovascular disease: they measure cholesterol and triglycerides. Therefore, the biomarker panel of the present invention can replace this lipid panel for routine diagnostics.


The biomarker panel can also be used in a method of monitoring the progress of various vascular disease treatments. A sample is taken from an individual currently receiving treatment for vascular disease and applied to the biomarker panel (either the four-panel test for clotting, the three-panel test for endothelial glycocalyx integrity, or both as described above). The sample reacts with various reagents in the panel based on the presence of the biomarkers described above. Results can be read by any method described above. Based on the detection of any or all of the biomarkers by comparing the results to a predetermined baseline, it can be determined if the treatments are working to reverse or prevent vascular disease. The treatments that can be monitored include, but are not limited to, anti-inflammatories (such as non-steroidal anti-inflammatory drugs (NSAIDS), steroids, or immune selective anti-inflammatory derivatives (ImSAIDs)), anticoagulants (such as alteplase, ardeparin, dalteparin, danaparoid, enoxaparin, fondaparinux, lepirudin, urokinase, or warfarin), antioxidants (such as glutathione, alpha-lipoic acid, CoQ10, resveratrol, carotenoids, astaxanthin Vitamin C, or Vitamin E), supplements, and any other suitable therapeutics.


Detecting various biomarkers used in the biomarker panels of the present invention is shown in EXAMPLE 1 below.


The invention is further described in detail by reference to the following experimental examples. These examples are provided for the purpose of illustration only, and are not intended to be limiting unless otherwise specified. Thus, the invention should in no way be construed as being limited to the following examples, but rather, should be construed to encompass any and all variations which become evident as a result of the teaching provided herein.


EXAMPLE 1

Materials and Methods


Mice


For the pilot study, 48 ten week old male C57/BI6 mice were obtained from Jackson Laboratories. Three mice were raised from 6 weeks on a regular diet and served as controls, and the remaining 45 were raised on a 60% fat diet (D12451, DIO series diet, Opensource Diets). For the bacterial dosage test, 16 ten week old male C57/BI6 mice were divided into 4 treatment groups and maintained on a normal diet.


Treatments


3,3′,4,4′-Tetrachlorobiphenyl (PCB-77) was obtained from Neosyn Laboratories. 100 mg of the dry chemical was suspended in 15.22 ml of corn oil to deliver 150 μmol/kg in 0.2 ml per mouse. Porphyromonas gingivalis 381 (ATCC 33277) was obtained from ATCC. The bacteria were stored frozen prior to use. The bacteria were cultured in multiple sterile tubes in 40 ml of supplemented tryptic soy broth at 37° C. under anaerobic conditions. The cultures were centrifuged, the medium removed, and the samples combined. A 100 μl sample of the bacteria was mixed with 100 μl of medium, and added to a 96 well plate. A microplate reader was used to measure the optical density at 600 nm to determine the concentration of bacteria. Based on the bacterial concentration, the samples were diluted to appropriate concentration with 2% carboxymethylcellulose in sterile phosphate buffered saline (PBS).


Gavage


A 20 gauge curved feeding needle was used to administer 0.2 ml of the treatment into the stomach of each mouse. Light Isoflurane gas anesthesia was utilized to facilitate introduction of the needle to the esophagus, and to decrease the risk of animal injury due to movement during gavage.


ELISA


Six test kits were used to analyze the collected plasma samples: Thrombin-Anti-Thrombin Complex ELISA (Kamiya Biomedical Company, Thousand Oaks, Calif.), Anti-Thrombin III ELISA (ABCam), Total Plasminogen Activation Inhibitor-1 ELISA (Molecular-Innovive), Syndecan-1 ELISA (USCN, Houston, Tex.), and Heparan Sulfate ELISA and Hyaluronan Synthase 1 ELISA (antibodies-online). All tests were performed on plasma, diluted to the fall within the standard curve if necessary, and carried out according to the manufacturer's instructions.


Methods


The gavage schedule was carried out as listed in FIG. 1A. Sufficient bacteria were unable to be grown to produce the suggested dosage of 3×1011 bacteria per mouse, which is much higher than the dosages used in the scientific literature, so the dosage was decreased to 5×109. There were 3 deaths overnight following the first bacterial gavage, so the second bacterial gavage was delayed to day 6, and reduced to 1.5×109.


For the dosing test, the mice received a gavage of bacteria on day 1 and day 6, to simulate the gavage schedule employed in the pilot study. FIG. 1B lists the bacterial dosages.


Sacrifice and Harvest


The mice were sacrificed on days 10, 15, or 20 according to the experimental plan (three each from groups 1-5). The animals were anesthetized by intraperitoneal injection of 90 mg/kg Ketamine and 8 mg/kg Xylazine, and Isoflurane gas anesthesia. Blood was collected by retro-orbital bleeding or from the heart and mixed with 50 mg/ml heparin to prevent clotting. The thorax was opened to expose the heart, and saline was injected into the left ventricle, with the right atrium opened to allow the drainage of blood and saline. The heart was perfused with at least 5 ml of saline and until no blood was observed in the drainage from the atrium. The heart was carefully dissected and frozen for histological sectioning. Plasma was collected from the blood samples by centrifuging at 1000 rpm for 15 minutes, and collecting the supernatant. The samples were stored at −80° C. until analysis.


Histology


The hearts were prepared as frozen sections. They were mounted in blocks and 10 μm thick sections were cut through the aortic valve, with 30 sections per mouse. Oil Red O staining was used to visualize the lipid content of the plaques. Multiple 10 μm sections at the level of the aortic sinus were analyzed for the presence of oil Red O lipid staining, plaque size, amount of fibrous tissue, and inflammation. The percentage of the lumen occupied by the first three features was calculated using Image Pro Plus. The average percentage of each feature was used to score the three features with the following scale: For fibrous tissue, lipid staining, and plaque size: 0=<2%, 1=≥2%, 2=≥4%, 3=≥6%, 4=≥10%. The level of inflammation in each section was scored on the following scale: 0=no inflammatory cells observed, 1=few macrophages with no giant cells, 2=foam cells present, 3=foam cells with cholesterol, 4=foam cells, giant cells, and cholesterol present. The inflammation score was averaged over all the sections, then converted to an overall score: For inflammation: 0=<0.2, 1=≥0.2, 2=≥0.4, 3=4=≥1.


Results


Pathology


No inflammation or plaques were found in Groups 1, 2, 5 and 6; however, inflammatory cells as indicated by general lipid staining throughout Group 3 (FIG. 2A) and well-defined atherosclerotic plaque in Group 4 (FIG. 2B).


Correlation of biomarkers with plaque formation or vascular inflammation:


Blood was drawn from the animals at various intervals and analyzed for biomarkers. Of the different markers evaluated, three showed significant levels in groups 3 and 4 animals (particularly in day 15 and 20), indicating high correlations to inflammation or plaque formation (statistically analyzed by independent T-test). These biomarkers include plasminogen activator inhibitor-1 (PAI-1), heparan sulfate, and hyaluronan synthase; syndecan-1 is a marginally predictive biomarker.


A. Highly Correlative Biomarkers


1. Plasminogen Activator Inhibitor-1 (PAI-1):



FIG. 3 shows that PAI-1 was significantly elevated in the 20 day sacrifice than control (group 6: normal food).


2. Heparan Sulfate (HS)



FIG. 4 shows that HS levels at 20 day sacrifice were significantly higher than in the control group.


3. Hyaluronan Synthase 1 (HAS-1)



FIG. 5 shows that hyaluronan synthase at 20 day sacrifice was significantly higher than the same time point in the control group.


B. Marginal Biomarker


1 Syndecan-1



FIG. 6 shows that syndecan-1 at 10 days was significantly higher than the same time point in the control group.


C. Poorly prognostic of inflammation or plaque formation:


1 Thrombin-Anti-Thrombin (TAT)


As shown in FIG. 7, no correlations were observed with the TAT complexes.


2. Antithrombin III


As shown in FIG. 8, there was no significant difference in the levels of Anti-Thrombin III between the treatment groups at any time point, or between time points within the groups, as measured by independent T-test.


Summary



FIG. 9 shows graphically the average scores for each group and time point, indicating PCB77 treatment as the most significant risk factor to producing inflammation and plaque.


EXAMPLE 2

Materials and Methods


A novel model of atherosclerosis in mice was developed, using a high fat diet and administration of a polychlorinated biphenyl (3,3′,4,4′-Tetrachlorobiphenyl; PCB-77) that promotes both obesity and atherosclerosis and the additional oral administration of a bacteria responsible for tooth decay, Porphyromonas gingivalis 381 (ATCC 33277). The objective of this study was to examine the association of biomarkers with the murine model of atherosclerosis in mice treated with a strategy to protect and repair endothelial glycocalyx. Eighty-four (84) ten week old male C57/BI6 mice were obtained from Jackson Laboratories. Thirty-two mice were raised from 6 weeks on a regular diet and served as controls, and the remaining mice were raised on a 60% fat diet (D12451, DIO series diet, Opensource Diets). 3,3′,4,4′-Tetrachlorobiphenyl (PCB-77) was obtained from Neosyn Laboratories. The dry chemical was suspended in 15.22 ml of corn oil to deliver 200 μmol/kg in 0.2 ml by gavage per mouse.


ELISA


Four test kits were used to analyze the collected plasma samples: Heparan Sulfate ELISA and Hyaluronan Synthase 1 (HAS-1) ELISA (Antibodies-Online), Total Plasminogen Activation Inhibitor-1 (PAI-1) ELISA (Molecular-Innovive) and Syndecan-1 (SDC1) ELISA (USCN, Houston, Tex.). All tests were performed on plasma, diluted to the fall within the standard curve if necessary, and carried out according to the manufacturer's instructions.


Sacrifice and Harvest


The mice were sacrificed on days 4, 11, or 18 according to the experimental plan (three each from groups). The animals were anesthetized by intraperitoneal injection of 90 mg/kg Ketamine and 8 mg/kg Xylazine, and Isoflurane gas anesthesia. Blood was collected by retro-orbital bleeding or from the heart and mixed with 50 mg/ml heparin to prevent clotting. The thorax was opened to expose the heart, and saline was injected into the left ventricle, with the right atrium opened to allow the drainage of blood and saline. The heart was perfused with at least 5 ml of saline and until no blood was observed in the drainage from the atrium. The heart was carefully dissected and frozen for histological sectioning. Plasma was collected from the blood samples by centrifuging at 1000 rpm for 15 minutes, and collecting the supernatant. The samples were stored at −80° C. until analysis.


Results


Hyaluronan Synthase 1 (HAS-1)



FIG. 10 shows the results for Hyaluronan Synthase 1 (HAS-1). It was observed that the highest HAS-1 levels occurred in the mice on the high fat diet treated with PCB on Day 1 and 3 and sacrificed at Day 4. Reductions in HAS-1 levels have been observed in mice treated to with compounds designed to restore and repair the endothelial glycocalyx.


Heparan Sulfate (HS)



FIG. 11 shows the results for Heparan Sulfate. An elevation in HS was observed in mice treated PCB-77 and Porphyromonas gingivalis.


Total Plasminogen Activation Inhibitor-1 (PAI-1)



FIG. 12 shows the results for PAI-1. It was observed that high PAI-1 levels occurred in all mice treated with an insult designed to provoke an atherosclerosis response.


Syndecan-1 (SDC1)



FIG. 13 shows the results for SDC1. An elevation in SDC-1 was observed in mice treated PCB-77 and Porphyromonas gingivalis, although a high degree of variability was seen in the results for this assay.


Pathology


The positive control group (High fat diet, PCB) revealed presence of a pathology consistent with plaque (FIGS. 14A, 10×; and 14B, 40×); fibrous material loosely attached to the surface of the arterial wall was observed in this sample. In contrast, the negative control group (Normal diet, no PCB, no treatment) exhibited the typical features of a normal arterial wall (14C).


Conclusion


The three biomarkers that were found highly correlative to plaque production are Hyaluronan Synthase (HAS-1), Heparan Sulfate (HS), and Plasminogen Activation Inhibitor-1 (PAI-1). The biochemical changes that define cardiovascular disease (CVD) are difficult to quantitate. In this regard, simplified and predictive biomarkers were developed involving blood samples to monitor onset of cardiovascular disease and its progression. These biomarkers are developed to provide a reliable predictor of cardiovascular events.


Significant smaller endothelial glycocalyx dimensions and amounts for two of its major constituents heparan sulfate and hyaluronan at the atherogenic sinus region of the carotid artery bifurcation compared with the common carotid region; perturbed endothelial glycocalyx content at pre-lesion areas within the arterial vascular tree contributes to local loss of EC barrier properties. A possible role of the endothelial glycocalyx in control of vascular wall permeability emerged, as suggested from increased local intima-to-media ratio at sites of reduced endothelial glycocalyx dimension at atherogenic risk areas. These early changes in local intima-to-media ratio were without evidence of blood cell or monocyte accumulation within the extended intimal layer, indicating a minimal inflammatory response at this very early stage.


In conclusion, predisposed arterial vascular regions have lower amounts of carbohydrate structures such as heparin sulfate and hyaluronan present within their luminal surface endothelial glycocalyx that results in locally reduced permeability barrier properties. In the present study, we reveal the endothelial cell glycocalyx as a complex 3D matrix, vulnerable to atherogenic risk factors, which, through preexisting differences in local architecture, results locally predisposed vulnerable arterial sites


A reasonable number of compounds tested exhibited an influence upon the biomarkers in the murine model of atherosclerosis, with marker changes in Hyaluronan Synthase 1 (HAS-1) and Total Plasminogen Activation Inhibitor-1 (PAI-1) in the curative model and Heparan Sulfate (HS) and Total Plasminogen Activation Inhibitor-1 (PAI-1) in the preventative protocol.


Throughout this application, various publications, including United States patents, are referenced by author and year and patents by number. Full citations for the publications are listed below. The disclosures of these publications and patents in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this invention pertains.


The invention has been described in an illustrative manner, and it is to be understood that the terminology, which has been used is intended to be in the nature of words of description rather than of limitation.


Obviously, many modifications and variations of the present invention are possible in light of the above teachings. It is, therefore, to be understood that within the scope of the appended claims, the invention can be practiced otherwise than as specifically described.

Claims
  • 1. A method of assaying a biological sample, the method comprising: assaying the biological sample for a biomarker panel comprising the biomarkers hyaluronan synthase-1, plasminogen activator inhibitor (PAI-1), and heparan sulfate (HS), and combinations thereof.
  • 2. The method of claim 1, wherein the biomarker panel additionally comprises the biomarker syndecan-1 (SDC1).
  • 3. The method of claim 1, wherein the sample is from an individual receiving treatment for cardiovascular disease.
  • 4. The method of claim 3, wherein the treatment is selected from the group consisting of anti-inflammatories, steroids, or immune selective anti-inflammatory derivatives, anticoagulants, antioxidants, and supplements.
  • 5. The method of claim 1, wherein the sample is from an individual receiving a drug being tested.
  • 6. The method of claim 1, wherein said assaying comprises using reagents for detecting the biomarkers.
  • 7. The method of claim 6, wherein the reagents comprise antibodies for detecting the biomarkers.
  • 8. The method of claim 7, wherein the antibodies are labeled or are capable of becoming labeled during an immunoassay.
  • 9. The method of claim 1, wherein the sample is from an individual suspected of having, or known to have, a perturbation of the glycocalyx.
  • 10. The method of claim 9, wherein the pertubation of the glycocalyx comprises a cardiovascular disease.
  • 11. The method of claim 10, wherein the cardiovascular disease comprises plaque formation or vascular inflammation.
  • 12. The method of claim 3, wherein the cardiovascular disease comprises plaque formation or vascular inflammation.
  • 13. The method of claim 1, wherein the sample is from an individual, and the method additionally comprises treating the individual for a perturbation of the glycocalyx based on the results of the biomarker panel assay.
  • 14. The method of claim 13, wherein the pertubation of the glycocalyx comprises a cardiovascular disease.
  • 15. The method of claim 14, wherein the cardiovascular disease comprises plaque formation or vascular inflammation.
  • 16. A method of treating a perturbation of the glycocalyx, wherein the method comprises treating an individual selected for treatment of the glycocalyx, wherein the individual is selected based on the results of a biomarker panel assay previously carried out according to the method of claim 8, wherein the method comprises treating the individual with an an agent selected from the group consisting of an anticoagulant, an antiplatelet agent, an anti-inflammatory agent, a steroid, an immune selective anti-inflammatory derivative, and an antioxidant or a combination thereof.
  • 17. The method of claim 16, wherein pertubation of the glycocalyx comprises a cardiovascular disease.
  • 18. The method of claim 17, wherein the cardiovascular disease comprises plaque formation or vascular inflammation.
  • 19. The method of claim 1, wherein the biomarker panel additionally comprises one or more of the biomarkers soluble fibrin, thrombin-antithrombin complex, antithrombin III, and hyaluronidase.
  • 20. The method of claim 16, wherein the biomarker panel additionally comprises the biomarker syndecan-1 (SDC1).
  • 21. The method of claim 20, wherein the biomarker panel additionally comprises one or more of the biomarkers soluble fibrin, thrombin-antithrombin complex, antithrombin III, and hyaluronidase.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a divisional of U.S. Ser. No. 16/060,840, filed on Jun. 8, 2018, U.S. Pat. No. 11,143,659, which is a 371 US National Phase of PCT/US2016/015015, filed Jan. 27, 2016, which claims benefit of and priority to U.S. Ser. No. 62/108,146, filed on Jan. 27, 2015, all of which are incorporated herein by reference in their entireties for all purposes.

US Referenced Citations (190)
Number Name Date Kind
5281700 Schnitzer et al. Jan 1994 A
5453359 Gargan et al. Sep 1995 A
5811250 Solum et al. Sep 1998 A
5843690 Gargan Dec 1998 A
5922302 Goldenberg et al. Jul 1999 A
6156530 Rånby Dec 2000 A
8759095 Vink et al. Jun 2014 B2
9086412 Taylor et al. Jul 2015 B2
11143659 Tunac Oct 2021 B2
20010051351 Racis Dec 2001 A1
20020086282 Pillarisetti et al. Jul 2002 A1
20020106634 Adams et al. Aug 2002 A1
20020132370 Lassen et al. Sep 2002 A1
20020182587 Pillarisetti Dec 2002 A1
20030003515 Farrell et al. Jan 2003 A1
20030008911 Evans et al. Jan 2003 A1
20030036103 Pillarisetti et al. Feb 2003 A1
20030040505 Fogelman et al. Feb 2003 A1
20030124536 McCarthy Jul 2003 A1
20030166004 Gyuris et al. Sep 2003 A1
20030199000 Valkirs et al. Oct 2003 A1
20030219813 Yang et al. Nov 2003 A1
20040002124 Lau et al. Jan 2004 A1
20040038856 Chakravarty et al. Feb 2004 A1
20040047861 Kehrel et al. Mar 2004 A1
20040121343 Buechler et al. Jun 2004 A1
20040142334 Schacht Jul 2004 A1
20040142496 Nicholson et al. Jul 2004 A1
20040203083 Buechler et al. Oct 2004 A1
20040213789 Yacoby-Zeevi et al. Oct 2004 A1
20040215087 Genero et al. Oct 2004 A1
20040253637 Buechler et al. Dec 2004 A1
20050032140 Kurosawa et al. Feb 2005 A1
20050065184 Wolf Mar 2005 A1
20050069969 Berg et al. Mar 2005 A1
20050089914 Yamasaki Apr 2005 A1
20050107601 Loeb May 2005 A1
20050181386 Diamond et al. Aug 2005 A1
20050250156 Shebuski et al. Nov 2005 A1
20060003338 Deng et al. Jan 2006 A1
20060039863 Schirner et al. Feb 2006 A1
20060105323 Whitelaw et al. May 2006 A1
20060269552 Yacoby-Zeevi et al. Nov 2006 A1
20060275214 Gregor et al. Dec 2006 A1
20060286681 Lehmann et al. Dec 2006 A1
20070065879 Conover et al. Mar 2007 A1
20070134814 Kajander et al. Jun 2007 A1
20070141055 Kajander et al. Jun 2007 A1
20070141632 Xu et al. Jun 2007 A1
20070196926 Soito et al. Aug 2007 A1
20070213255 Hastings et al. Sep 2007 A1
20070225222 Chiquet-Ehrismann et al. Sep 2007 A1
20070225261 Miller et al. Sep 2007 A1
20070239483 Chandler et al. Oct 2007 A1
20070269836 McPherson Nov 2007 A1
20080009684 Corsetti et al. Jan 2008 A1
20080010024 Diamond Jan 2008 A1
20080057516 Saarma et al. Mar 2008 A1
20080121025 Okazaki May 2008 A1
20080124277 Arap et al. May 2008 A1
20080160007 Powell Jul 2008 A1
20080300797 Tabibiazar et al. Dec 2008 A1
20080300798 McDevitt et al. Dec 2008 A1
20080311606 Chapman-Montgomery et al. Dec 2008 A1
20090011055 Lawrence et al. Jan 2009 A1
20090054256 Dogulu et al. Feb 2009 A1
20090104121 Madasamy Apr 2009 A1
20090155827 Zeiher et al. Jun 2009 A1
20090197344 Villard-Saussine et al. Aug 2009 A1
20090215042 Sella-Tavor et al. Aug 2009 A1
20090246810 Maier et al. Oct 2009 A1
20090263827 Johansen Oct 2009 A1
20100028335 Lu et al. Feb 2010 A1
20100068705 Helgadottir et al. Mar 2010 A1
20100105046 Epstein et al. Apr 2010 A1
20100112587 Hare et al. May 2010 A1
20100130403 Pfuetzner et al. May 2010 A1
20100158896 Brown et al. Jun 2010 A1
20100159474 Bergmann et al. Jun 2010 A1
20100209350 Pfuetzner et al. Aug 2010 A1
20100233085 Kwon et al. Sep 2010 A1
20100248288 Hess et al. Sep 2010 A1
20100249064 Singleton et al. Sep 2010 A1
20100261284 Spanuth Oct 2010 A1
20100267025 Young Oct 2010 A1
20100267062 Frey et al. Oct 2010 A1
20100286053 Kuan et al. Nov 2010 A1
20100291614 Nitz et al. Nov 2010 A1
20100304424 Vink et al. Dec 2010 A1
20100310646 Oxvig et al. Dec 2010 A1
20110003297 Liew Jan 2011 A1
20110008346 Duckers Jan 2011 A1
20110070601 Kastrup Mar 2011 A1
20110076692 Sista et al. Mar 2011 A1
20110083199 Essers et al. Apr 2011 A1
20110104735 Buehrer et al. May 2011 A1
20110107821 Hess et al. May 2011 A1
20110111527 Hess et al. May 2011 A1
20110117589 Bergmann et al. May 2011 A1
20110136157 Cooper Jun 2011 A1
20110137131 Adourian et al. Jun 2011 A1
20110177610 Matsuo et al. Jul 2011 A1
20110229911 Bergmann et al. Sep 2011 A1
20110262444 Kim Oct 2011 A1
20110274749 Gaillard et al. Nov 2011 A1
20120003751 Bergmann et al. Jan 2012 A1
20120028880 Pasqualini et al. Feb 2012 A1
20120149131 Struck et al. Jun 2012 A1
20120164669 Hess et al. Jun 2012 A1
20120208715 McDevitt et al. Aug 2012 A1
20120208762 Dudley Aug 2012 A1
20120219943 Ky et al. Aug 2012 A1
20120231472 Anderberg et al. Sep 2012 A1
20120264636 Holm et al. Oct 2012 A1
20130004487 Zeiher et al. Jan 2013 A1
20130052637 Kovar et al. Feb 2013 A1
20130137632 Pfuetzner et al. May 2013 A1
20130164284 Lu et al. Jun 2013 A1
20130171649 Mayr Jul 2013 A1
20130190197 Liew Jul 2013 A1
20130210041 Anderberg et al. Aug 2013 A1
20130261177 Johansson et al. Oct 2013 A1
20130273096 Daniels Oct 2013 A1
20130302841 Struck et al. Nov 2013 A1
20140024551 Mayr Jan 2014 A1
20140044797 Johansson et al. Feb 2014 A1
20140065648 Wienhues-Thelen et al. Mar 2014 A1
20140100128 Narain et al. Apr 2014 A1
20140141986 Spetzler et al. May 2014 A1
20140147867 Arnold et al. May 2014 A1
20140206698 Dudley Jul 2014 A1
20140271464 Garcia-Martinez et al. Sep 2014 A1
20140315752 Anderberg et al. Oct 2014 A1
20140324460 Caffrey et al. Oct 2014 A1
20140357505 Mohler, III et al. Dec 2014 A1
20150010929 Anderberg et al. Jan 2015 A1
20150057325 Johansson et al. Feb 2015 A1
20150064139 Shoemaker et al. Mar 2015 A1
20150079615 Wienhues-Thelen et al. Mar 2015 A1
20150087727 Bergmann et al. Mar 2015 A1
20150099311 Holmes et al. Apr 2015 A1
20150160229 Schaal et al. Jun 2015 A1
20150175979 Bottini et al. Jun 2015 A1
20150268251 Zaugg et al. Sep 2015 A1
20150308939 Oberleithner Oct 2015 A1
20150346217 Spanuth Dec 2015 A1
20150376704 Harrington et al. Dec 2015 A1
20160084849 Chojkier et al. Mar 2016 A1
20160109464 Horsch et al. Apr 2016 A1
20160121023 Edelman et al. May 2016 A1
20160169911 Block et al. Jun 2016 A1
20160252526 Bergmann et al. Sep 2016 A1
20160282362 Karsdal et al. Sep 2016 A1
20160320411 Struck et al. Nov 2016 A1
20160320416 Pugia et al. Nov 2016 A1
20160327548 Crawford et al. Nov 2016 A1
20170010280 Tanaka et al. Jan 2017 A1
20170010283 Karl et al. Jan 2017 A1
20170138961 Hess et al. May 2017 A1
20170218091 Ambrosi Aug 2017 A1
20170227552 Latini et al. Aug 2017 A1
20170234853 Contant et al. Aug 2017 A1
20170269106 Everett et al. Sep 2017 A1
20170304389 Mann Oct 2017 A1
20180000972 Lee Jan 2018 A1
20180003685 Cummings et al. Jan 2018 A1
20180003725 Kline Jan 2018 A1
20180031483 Singamaneni et al. Feb 2018 A1
20180036285 Tunac et al. Feb 2018 A1
20180088131 Ostrowski et al. Mar 2018 A1
20180100862 Goix et al. Apr 2018 A1
20180291374 Bloch et al. Oct 2018 A1
20180292414 Amir et al. Oct 2018 A1
20180298341 Elliman Oct 2018 A1
20180364257 Tunac Dec 2018 A1
20190010223 Smith Jan 2019 A1
20190112582 Redondo Moya et al. Apr 2019 A1
20190224135 Struck et al. Jul 2019 A1
20190227074 Denk et al. Jul 2019 A1
20190227081 Struck et al. Jul 2019 A1
20190250126 Hall et al. Aug 2019 A1
20190369114 Van Eyk et al. Dec 2019 A1
20200003759 Vath et al. Jan 2020 A1
20200018747 King et al. Jan 2020 A1
20200041496 Kershner et al. Feb 2020 A1
20200049721 Bergmann Feb 2020 A1
20200166523 Gill et al. May 2020 A1
20200199510 Luo et al. Jun 2020 A1
20200241019 Kim Jul 2020 A1
20200271670 Block et al. Aug 2020 A1
Foreign Referenced Citations (83)
Number Date Country
1352029 Jun 2002 CN
102353789 Feb 2012 CN
102863530 Jan 2013 CN
104914247 Sep 2015 CN
109055368 Dec 2018 CN
20220351 Aug 2003 DE
10247356 Apr 2004 DE
10316059 Nov 2004 DE
2439535 Apr 2012 EP
2597467 May 2013 EP
2003227837 Aug 2003 JP
2008188016 Aug 2008 JP
2011038858 Feb 2011 JP
2015047141 Mar 2015 JP
2018021797 Feb 2018 JP
2019158753 Sep 2019 JP
20060105654 Oct 2006 KR
20090048056 May 2009 KR
20180055319 May 2018 KR
2154977 Aug 2000 RU
2158532 Nov 2000 RU
2248744 Mar 2005 RU
2533836 Nov 2014 RU
2550722 May 2015 RU
2557916 Jul 2015 RU
2592237 Jul 2016 RU
2617418 Apr 2017 RU
2720672 May 2020 RU
57001 Feb 2011 UA
60581 Jun 2011 UA
92604 Aug 2014 UA
WO-9310261 May 1993 WO
WO-9810293 Mar 1998 WO
WO-9826092 Jun 1998 WO
WO-9826292 Jun 1998 WO
WO-9918442 Apr 1999 WO
WO-9948916 Sep 1999 WO
WO-9964627 Dec 1999 WO
WO-0050639 Aug 2000 WO
WO-0111064 Feb 2001 WO
WO-0123426 Apr 2001 WO
WO-0173445 Oct 2001 WO
WO-0202593 Jan 2002 WO
WO-2004001421 Dec 2003 WO
WO-2006052924 May 2006 WO
WO-2006115047 Nov 2006 WO
WO-2007070021 Jun 2007 WO
WO-2008009869 Jan 2008 WO
WO-2008040328 Apr 2008 WO
WO-2009033095 Mar 2009 WO
WO-2009058168 May 2009 WO
WO-2009100907 Aug 2009 WO
WO-2009128917 Oct 2009 WO
WO-2010018203 Feb 2010 WO
WO-2010026272 Mar 2010 WO
WO-2010047767 Apr 2010 WO
WO-2010054810 May 2010 WO
WO-2010133173 Nov 2010 WO
WO-2012009547 Jan 2012 WO
WO-2012020045 Feb 2012 WO
WO-2012066140 May 2012 WO
WO-2013045570 Apr 2013 WO
WO-2013188787 Dec 2013 WO
WO-2015073709 May 2015 WO
WO-2015110957 Jul 2015 WO
WO-2016123163 Aug 2016 WO
WO-2016126662 Aug 2016 WO
WO-2016130802 Aug 2016 WO
WO-2017136652 Aug 2017 WO
WO-2018042072 Mar 2018 WO
WO-2018136825 Jul 2018 WO
WO-2018178386 Oct 2018 WO
WO-2018208846 Nov 2018 WO
WO-2019183671 Oct 2019 WO
WO-2020018005 Jan 2020 WO
WO-2020053355 Mar 2020 WO
WO-2020074777 Apr 2020 WO
WO-2020081866 Apr 2020 WO
WO-2020115288 Jun 2020 WO
WO-2020146263 Jul 2020 WO
WO-2020148769 Jul 2020 WO
WO-2020167735 Aug 2020 WO
WO-2021062298 Apr 2021 WO
Non-Patent Literature Citations (34)
Entry
Alexander et al. (2011) “Association between γ' fibrinogen levels and inflammation” Thromb Haemost 105:605-9 [NIH Public Access—Author Manuscript—10 pages].
Allender et al. (2008) “Patterns of coronary heart disease mortality over the 20th century in England and Wales: Possible plateaus in the rate of decline” BMC Public Health 8:148 (12 pages).
Appiah et al. (2015) “Association of Plasma γ' Fibrinogen With Incident Cardiovascular Disease The Atherosclerosis Risk in Communities (ARIC) Study” Arterioscler Thromb Vasc Biol. 35(12):2700-2706.
Canadian Office Action dated Jan. 6, 2022 issued in CA 3,012,985.
Co-pending U.S. Appl. No. 17/762,703, filed Mar. 22, 2022.
Cosin-Sales et al. (2004) “Pregnancy-Associated Plasma Protein A and Its Endogenous Inhibitor, the Proform of Eosinophil Major Basic Protein (proMBP), Are Related to Complex Stenosis Morphology in Patients With Stable Angina Pectoris” Circulation 109(14):1724-1728.
Danesh et al. (2005) “Plasma Fibrinogen Level and the Risk of Major Cardiovascular Diseases and Nonvascular Mortality An Individual Participant Meta-analysis” JAMA 294(14):1799-1809.
International Preliminary Report on Patentability dated Apr. 7, 2022, in Application No. PCT/US2020/052912.
Kalousová et al. (2014) “Pregnancy-associated plasma protein A associates with cardiovascular events in diabetic hemodialysis patients.” Atherosclerosis 236(2):263-269.
Lindahl, Bertil (2013) “The Story of Growth Differentiation Factor 15: Another Piece of the Puzzle” Clinical Chemistry 59(11): 1550-1552.
Lovely (2010) “γ' Fibrinogen: Evaluation of a New Assay for Study of Associations with Cardiovascular Disease” Clin Chem 2010; 56(5):781-8.
Lovely et al. (2011) “Assessment of Genetic Determinants of the Association of γ' Fibrinogen in Relation to Cardiovascular Disease” Arterioscler Thromb Vasc Biol. 31(10): 2345-2352.
Mannila et al. (2007) “Elevated plasma fibrinogen gamma' concentration is associated with myocardial infarction: effects of variation in fibrinogen genes and environmental factors” J Thromb Haemost 5(4):766-73 doi: 10.1111/j.1538-7836.2007.02406.x. Epub Jan. 22, 2007.
Meh et al. (1996) “Identification and Characterization of the Thrombin Binding Sites on Fibrin” J Biol Chem 271(38):23121-23125.
Mueller et al. (2006) “Increased pregnancy-associated plasma protein-A as a marker for peripheral atherosclerosis: results from the Linz Peripheral Arterial Disease Study” Clin Chem 52(6):1096-1103.
Papageorgiou et al. (2017) “Coronary Artery Atherosclerosis in Hypertensive Patients: The Role of Fibrinogen Genetic Variability” Rev Esp Cardiol. 70:34-41.
Parveen et al. (2015) “Pregnancy Associated Plasma Protein-A (PAPP-A) Levels in Acute Coronary Syndrome: A Case Control Study in a Tertiary Care Centre” Indian J Clin Biochem. 30(2): 150-154.
PCT International Preliminary Report on Patentability and Written Opinion dated Aug. 1, 2017 issued in PCT/US2016/015015.
PCT International Search Report and Written Opinion dated Feb. 24, 2021 issued in PCT/US2020/052912.
PCT International Search Report and Written Opinion dated Jul. 18, 2016 issued in PCT/US2016/015015.
Robbers et al. (2013) “Magnetic resonance imaging-defined areas of microvascular obstruction after acute myocardial infarction represent microvascular destruction and haemorrhage” Eur Heart J. 34:2346-2353.
Rohatgi et al. (2012) “Association of Growth Differentiation Factor-15 with Coronary Atherosclerosis and Mortality in a Young, Multiethnic Population: Observations from the Dallas Heart Study” Clinical Chemistry 58(1):172-182.
Tunac (2021) “Curative and Preventive Treatment for Cardiovascular Disease (CVD) Targeting Multiple Etiology” Cardiology and Cardiovascular Research 5(2): 103-125.
U.S. Notice of Allowance dated Feb. 18, 2021 issued in U.S. Appl. No. 16/060,840.
U.S. Notice of Allowance dated Jun. 9, 2021 issued in U.S. Appl. No. 16/060,840.
U.S. Office Action dated May 18, 2020 issued in U.S. Appl. No. 16/060,840.
Van den Herik et al. (2011) “γ'/total fibrinogen ratio is associated with short-term outcome in ischaemic stroke” Thromb Haemost 105(3):430-4; doi: 10.1160/TH10-09-0569. Epub Dec. 6, 2010.
Van den Herik et al. (2012) “Fibrinogenγ'levels in patients with intracerebral hemorrhage” Thrombosis Research 129(6): 807-809.
Wallentin et al. (2014) “Growth Differentiation Factor 15, a Marker of Oxidative Stress and Inflammation, for Risk Assessment in Patients With Atrial Fibrillation” Circulation 130:1847-1858.
Wiklund et al. (2010) “Macrophage inhibitory cytokine-1 (MIC-1/GDF15): a new marker of all-cause mortality” Aging Cell 9: 1057-1064.
Wollert et al. (2017) “Growth Differentiation Factor 15 as a Biomarker in Cardiovascular Disease” Clinical Chemistry 63(1): 140-151.
Wu et al. (2016) “Level of Pregnancy-associated Plasma Protein-A Correlates With Coronary Thin-cap Fibroatheroma Burden in Patients With Coronary Artery Disease: Novel Findings From 3-Vessel Virtual Histology Intravascular Ultrasound Assessment” Medicine (Baltimore) 95(3): e2563 (7 pages).
Zengin et al. (2015) “The utility of pregnancy-associated plasma protein A for determination of prognosis in a cohort of patients with coronary artery disease” Biomark Med. 9:731-741 ((Epub ahead of print—10.2217/BMM.15.41 ISSN 1752-0363 11 pages).
Zimmers et al. (2005) “Growth differentiation factor-15/macrophage inhibitory cytokine-1 induction after kidney and lung injury” Shock 23(6): 543-8.
Related Publications (1)
Number Date Country
20220057410 A1 Feb 2022 US
Provisional Applications (1)
Number Date Country
62108146 Jan 2015 US
Divisions (1)
Number Date Country
Parent 16060840 US
Child 17469714 US