US20110195491A1 - Devices and systems for biomarker detection - Google Patents

Devices and systems for biomarker detection Download PDF

Info

Publication number
US20110195491A1
US20110195491A1 US12/906,168 US90616810A US2011195491A1 US 20110195491 A1 US20110195491 A1 US 20110195491A1 US 90616810 A US90616810 A US 90616810A US 2011195491 A1 US2011195491 A1 US 2011195491A1
Authority
US
United States
Prior art keywords
diagnostic marker
stabilizing agent
microfluidic device
present disclosure
saliva
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/906,168
Inventor
Richard H. Selinfreund
Rakesh Vig
Richard P. Gill
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.)
Companion Diagnostics Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US12/906,168 priority Critical patent/US20110195491A1/en
Priority to PCT/US2011/031935 priority patent/WO2011127467A1/en
Publication of US20110195491A1 publication Critical patent/US20110195491A1/en
Assigned to COMPANION DIAGNOSTICS INC. reassignment COMPANION DIAGNOSTICS INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VIG, RAKESH, GILL, RICHARD P., SELINFREUND, RICHARD H.
Priority to US13/911,784 priority patent/US20140155282A1/en
Assigned to BMO HARRIS BANK N.A. reassignment BMO HARRIS BANK N.A. SECURITY INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: COMPANION DIAGNOSTICS, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54393Improving reaction conditions or stability, e.g. by coating or irradiation of surface, by reduction of non-specific binding, by promotion of specific binding
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61BDIAGNOSIS; SURGERY; IDENTIFICATION
    • A61B10/00Other methods or instruments for diagnosis, e.g. instruments for taking a cell sample, for biopsy, for vaccination diagnosis; Sex determination; Ovulation-period determination; Throat striking implements
    • A61B10/0045Devices for taking samples of body liquids
    • 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
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • C12Q1/40Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase involving amylase
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/10Composition for standardization, calibration, simulation, stabilization, preparation or preservation; processes of use in preparation for chemical testing
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/10Composition for standardization, calibration, simulation, stabilization, preparation or preservation; processes of use in preparation for chemical testing
    • Y10T436/105831Protein or peptide standard or control [e.g., hemoglobin, etc.]
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/25Chemistry: analytical and immunological testing including sample preparation
    • Y10T436/2525Stabilizing or preserving

Definitions

  • Biomarkers are molecular indicators of a specific biological property, a biochemical feature or facet that can be used to measure the progress of a disease or the effects of a treatment.
  • serum low-density lipoprotein (LDL) is a biomarker of cholesterol and blood pressure
  • P53 gene is a biomarker for cancer.
  • LDL low-density lipoprotein
  • accurate diagnosis is particularly important, especially where the side effects of a treatment are severe.
  • biomarkers as molecular indicators not only detect the presence or absence of the biomarker, but often must measure the exact concentration of a biomarker to determine whether an abnormal condition exists. Because of the requirement for accuracy, the process of sample collection, preparation, and analysis are often complicated and time consuming. Currently, blood-based assays for biomarker presence or activity are considered to be the “gold standard” for biomarker-type assays.
  • the disclosure of the present application provides various devices for analyzing the presence or absence of a diagnostic marker to be detected in a body fluid.
  • the device comprises a container, wherein the container houses a sample reservoir having a sample input port in fluid connection with at least one detector array, wherein the detector array comprises at least one detector comprising a reaction chamber comprising an immobilized capture molecule, a stabilizing agent contained within the sample reservoir, the stabilizing agent useful to completely or substantially prevent degradation or inactivation of a diagnostic marker, and a reagent contained within the detector array, the reagent capable of undergoing a colorimetric reaction or displaying an optically detectable signal and capable of reacting with the diagnostic marker.
  • a device for analyzing the presence or absence of a diagnostic marker to be detected in a body fluid the device further comprising filter capable of separating at least one unwanted component from a body fluid.
  • an embodiment of the device of the present disclosure may further comprise a results display, wherein a result from the detector array is visually detectable.
  • the results display of an embodiment of a device of the present disclosure, may comprise a plurality of displays capable to visualizing a plurality of detected diagnostic markers.
  • the device further comprises a control display, wherein the detection of at least one control reagent by the at least one detector is visualized.
  • the stabilizing agent is selected from the group consisting of a protease inhibitor, a DNase inhibitor, and a RNase inhibitor.
  • the stabilizing agent is useful to completely or substantially prevent degradation or inactivation of a diagnostic marker selected from the group consisting of a protein, a glycoprotein, a nucleic acid, an enzyme, an enzyme inhibitor, and a metabolite.
  • the stabilizing agent is useful to completely or substantially prevent degradation or inactivation of a diagnostic marker indicative of a disease.
  • the disease state is selected from the group consisting of a cancer, a metabolic disease, a cardiac disease, diabetes, a bacterial infection, a viral infection, a fungal infection, and a parasitic infection.
  • the stabilizing agent is selected from the group consisting of Fixanal® Buffer 6.0, acetic acid, aluminum hydroxide bentonite, aluminum sulfate hydrate, benzoic acid, aluminum potassium sulfate dodecahydrate, caffeine, and 3-tert-butyl-hydroxyanisole, or a combination thereof.
  • the device comprises a sample reservoir having a sample input port in fluid connection with at least one detector array, wherein said detector array comprises at least one detector comprising a reaction chamber having an immobilized capture molecule, a stabilizing agent useful to completely or substantially prevent degradation or inactivation of a diagnostic marker, a reagent capable of undergoing a colorimetric reaction or displaying an optically detectable signal and capable of reacting with the diagnostic marker, a filter capable of separating at least one unwanted component from the body fluid, and a results display, wherein a result from the detector array is visualized.
  • said detector array comprises at least one detector comprising a reaction chamber having an immobilized capture molecule, a stabilizing agent useful to completely or substantially prevent degradation or inactivation of a diagnostic marker, a reagent capable of undergoing a colorimetric reaction or displaying an optically detectable signal and capable of reacting with the diagnostic marker, a filter capable of separating at least one unwanted component from the body fluid, and a results display, wherein a result from the detector array is
  • FIGS. 1A and B show a diagram of a detection system, according to at least one embodiment of the present disclosure
  • FIGS. 2-4 shows a diagram of a method of producing a bead system, according to at least one embodiment of the present disclosure
  • FIG. 5 shows a flowchart of a method for creating a bead system, according to at least one embodiment of the present disclosure
  • FIGS. 6A and B show a diagram of a test strip, according to at least one embodiment of the present disclosure
  • FIG. 6C show a diagram of a microarray, according to at least one embodiment of the present disclosure.
  • FIG. 6D shows a diagram of a microfluidic system, according to at least one embodiment of the present disclosure
  • FIG. 7 shows a flowchart of a method of stabilizing diagnostic markers, according to at least one embodiment of the present disclosure
  • FIG. 8 shows a flowchart of a method analyzing a diagnostic markers, according to at least one embodiment of the present disclosure
  • FIG. 9 shows a an exemplary system framework, according to at least one embodiment of the present disclosure.
  • FIG. 10 shows a graphical depiction of ⁇ -amylase inhibition according to at least one embodiment of the present disclosure
  • FIG. 11 shows a graphical depiction of ⁇ -amylase inhibition according to at least one embodiment of the present disclosure
  • FIG. 12 shows a graphical depiction of lysozyme inhibition according to at least one embodiment of the present disclosure
  • FIG. 13 shows a graphical depiction of a galactose oxidase screen according to at least one embodiment of the present disclosure
  • FIG. 14A shows a three dimensional plot of glycated Hemoglobin A1c (HbA1c) analyzed by high pressure liquid chromatography (HPLC) according to at least one embodiment of the present disclosure
  • FIG. 14B shows a ultraviolet (UV) image of HbA1c analyzed by HPLC according to at least one embodiment of the present disclosure
  • FIG. 14C shows a chromatogram of HbA1c analyzed by HPLC according to at least one embodiment of the present disclosure
  • FIG. 15A shows a three dimensional plot of HbA1c analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure
  • FIG. 15B shows a UV image of HbA1c analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure
  • FIG. 15C shows a chromatogram of HbA1c analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure
  • FIG. 16A shows a three dimensional plot of HbA1c analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure
  • FIG. 16B shows a UV image of HbA1c analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure
  • FIG. 16C shows a chromatogram of HbA1c analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure
  • FIG. 17 shows a comparison of chromatograms from HPLC analyzed HbA1c ( FIG. 17A ), HbA1c+Saliva ( FIG. 17B ), and HbA1c+Saliva+Stabilizing mixture ( FIG. 17C ) according to at least one embodiment of the present disclosure;
  • FIG. 18 shows a comparison of UV images from HPLC analyzed HbA1c ( FIG. 18A ), HbA1c+Saliva ( FIG. 18B ), and HbA1c+Saliva+Stabilizing mixture ( FIG. 18C ) according to at least one embodiment of the present disclosure;
  • FIG. 19 shows a comparison of three dimensional plots from HPLC analyzed HbA1c ( FIG. 19A ), HbA1c+Saliva ( FIG. 19B ), and HbA1c+Saliva+Stabilizing mixture ( FIG. 19C ) according to at least one embodiment of the present disclosure;
  • FIG. 20A shows a three dimensional plot of Cancer Antigen 19-9 (CA 19-9) analyzed by high pressure liquid chromatography (HPLC) according to at least one embodiment of the present disclosure
  • FIG. 20B shows a ultraviolet (UV) image of CA 19-9 analyzed by HPLC according to at least one embodiment of the present disclosure
  • FIG. 21A shows a three dimensional plot of CA 19-9 analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure
  • FIG. 21B shows a UV image of CA 19-9 analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure
  • FIG. 22A shows a three dimensional plot of CA 19-9 analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure
  • FIG. 22B shows a UV image of CA 19-9 analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure
  • FIG. 23 shows a comparison of three dimensional plots from HPLC analyzed CA 19-9 ( FIG. 23A ), CA 19-9+Saliva ( FIG. 23B ), and CA 19-9+Saliva+Stabilizing mixture ( FIG. 23C ) according to at least one embodiment of the present disclosure;
  • FIG. 24 shows a comparison of UV images from HPLC analyzed CA 19-9 ( FIG. 24A ), CA 19-9+Saliva ( FIG. 24B ), and CA 19-9+Saliva+Stabilizing mixture ( FIG. 24C ) according to at least one embodiment of the present disclosure;
  • FIG. 25 shows a graphical depiction of Peroxidase activity inhibition by an inhibitor cocktail according to at least one embodiment of the present disclosure
  • FIG. 26 shows a graphical depiction of Peroxidase activity inhibition by caffeine according to at least one embodiment of the present disclosure
  • FIG. 27 shows a graphical depiction of Peroxidase activity inhibition by 3-tert-butyl-hydroxyanisole according to at least one embodiment of the present disclosure
  • FIG. 28 shows a graphical depiction of Peroxidase activity inhibition by benzoic acid according to at least one embodiment of the present disclosure
  • FIG. 29 shows a graphical depiction of Peroxidase activity inhibition by aluminum potassium sulfate dodecahydrate according to at least one embodiment of the present disclosure
  • FIG. 30 shows a schematic of a diagnostic system, according to at least one embodiment of the present disclosure
  • FIG. 31 shows a graphical representation of an assay component, according to at least one embodiment of the present disclosure.
  • FIG. 32 shows a graphical depiction of a fluid collection device, according to at least one embodiment of the present disclosure.
  • compositions, systems, and methods for biomarker stabilization and analysis provide various compositions, systems, and methods for biomarker stabilization and analysis.
  • formulations are disclosed herein, which function to preserve the level of diagnostic markers, such as biomarkers, in body fluids, from enzymatic alteration, or degradation.
  • methods for the collection and analysis of body fluids are disclosed.
  • the methods and formulations disclosed herein can be used to improve the sampling and testing of a body fluid by conditioning the body fluids for the stabilization of specific biomolecules and/or drug metabolites.
  • body fluids includes fluids produced by the body, such as saliva, or fractions thereof, mucous secretions, tears, sweat, bile, semen, urine, vaginal secretions, exhalations, anal secretions, blood, plasma, serum and mixtures of thereof.
  • Body fluids may also comprise cancer cells, peripheral blood mononuclear cells, lymphocytes, lymph fluid, and other tissue secretions or fluid.
  • Saliva is clear, viscous fluid with a slightly alkaline pH and a pI range from 11.5-3.0. It is hypotonic, composed of about 99.5% water, and also contains ions (e.g., K + , Na + , Ca 2+ , Mg 2+ , H + , Cl ⁇ , HCO 3 ⁇ , I ⁇ , F ⁇ , HPO 4 2 ⁇ ), and small organic molecules (e.g., ureas, hormones, lipids, DNA, and RNA). There are multiple contributors to the composition of saliva. Saliva has a complex “proteome”—10 6 D glycoproteins to 1000D peptides.
  • salivary glands It contains secretory products of salivary glands, products of B cells, PMNs, epithelial cells, and bacteria.
  • Major e.g., parotid, submandibular, and sublingual
  • minor e.g., palatine and retromolar
  • salivary glands contribute to the composition of saliva, along with extraneous contributors such as gingival crevicular fluid, serum proteins, white blood cells and their byproducts, oral epithelial cells, oral bacteria, food debris and dissolved food components.
  • Saliva from different glands may differ in composition. For example, saliva from the parotid gland is dominated by serous secretory cells, whereas saliva from the submandibular and sublingual glands and minor glands are mixed serous or mostly mucous. There can also be qualitative and quantitative differences in saliva output that affect its composition. Glandular contribution to saliva is affected by level of gland activity. The amount of saliva secreted per minute or the salivary flow rate influences the concentration of the constituents as well as the proportion of the constituents from each of the three pairs of major salivary glands and the minor salivary glands.
  • conditioning of body fluids for the preservation of diagnostic markers may be accomplished through the use of stabilizing agents, which may be incorporated into one or more carrier vehicles, such as rinses, gums, beverages, and confectionaries.
  • stabilizing agents such as rinses, gums, beverages, and confectionaries.
  • specific rinses and/or pre-rinses specially formulated according to the specific molecule or molecules (the diagnostic marker) to be detected in the body fluid may contain a stabilization agent.
  • the use of rinses and pre-rinses of the present disclosure to condition the body fluid may enhance the sensitivity for detection of specific diagnostic markers for clinical diagnosis. Additionally, this process may improve the signal-to-noise ratio for a better diagnostic yield. Further, each rinse/pre-rinse can be specifically formulated to reduce or prevent false positive and false negative results.
  • the stabilizing agent may act to prevent/decrease the degradation, or reduction of activity, of diagnostic markers.
  • the stabilizing agent may be comprised of one or more Generally Regarded as Safe (GRAS) compound, as determined by the Food and Drug Administration of the United States of America, such as those listed in Table I.
  • GRAS Generally Regarded as Safe
  • An exemplary embodiment of a stabilization agent may act to stabilize a diagnostic marker, and/or to bind to a destructive component to prevent the destructive component from degrading or decreasing the activity of the diagnostic marker.
  • the stabilizing agent may comprise at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, or at least 25 compounds which acts to stabilize the diagnostic marker, and/or to bind to a destructive component to prevent the destructive component from degrading or decreasing the activity of the diagnostic marker.
  • the stabilizing agent of the present disclosure may be present in at a level of between about 200ppm to about 2,000 ppm, about 400ppm to about 1600 ppm, about 600ppm to about 1400 ppm, about 800 ppm to about 1200 ppm.
  • the stabilizing agent may be present at a level of approximately 500 ppm.
  • the stabilizing agent may block at least one enzymatic activity of a destructive component.
  • the destructive component may be one or more component of a bodily fluid which acts to degrade, or decrease the activity of a diagnostic marker.
  • the destructive component may be one or more Amylases, Lysozymes, Peroxidases, Glycosidases, Esterases, Proteases, and/or Peptidases.
  • the stabilizing agent may be a naturally occurring or artificial protease inhibitor, DNase inhibitor, or RNase inhibitor.
  • a natural stabilizing agent may include any component found from food products (including, but not limited to, the Solanaceae family), herbs, or spices that acts to decrease the effect of a destructive component.
  • exemplary embodiments of the natural stabilizing agents may be any of the inhibitors found in lima beans, soybeans, or avian eggs (such as ovomucoid glycoprotein protease inhibitors) (See Table II).
  • an embodiment of the stabilizing agent may also comprise a molecule which specifically inhibits the activity of a molecule which targets the diagnostic marker for degradation or inactivation.
  • the stabilizing agent may act to alter the composition of the body fluid (such as pH, or ion concentration) to decrease the degradation or inactivation of the diagnostic marker.
  • diagnostic marker may be any biological molecule whose presence, specific concentration, or specific activity may be indicative of a disease state or heath/lifestyle characteristic.
  • indicator of a disease state refers to the presence, progression, prevention, remission, amelioration, or prognosis of a disease. This also includes, but is not limited to, the effects of a therapeutic on a disease.
  • the disease state analyzed through use of the diagnostic marker may in an exemplary embodiment be directed towards any cancer, metabolic disease (such as diabetes I or II), cardiovascular disease, or the predisposition of any of the same.
  • a disease state may also include the infection of the patient by a bacterial, virus, yeast, fungus, or parasite.
  • Bacterial infections may include a mammalian infection by any bacterial species.
  • Embodiments of the bacterial species may include, but are not limited to, Bacillus anthracis, Bacillus cereus, Staphylococcus aureus, Listeria monocytogenes, Streptococcus pneumoniae, Streptococcus pyogenes, Clostridium botulinum, Clostridium difficile, Clostridium perfringens, Clostridium tetani, Borrelia burgdorferi, Treponema pallidum, Chlamydia trachomatis, Chlamydophila psittaci, Corynebacterium diphtherias, Mycobacterium tuberculosis, and Mycobacterium avium, Rickettsia prowazekii, Rickettsia rickettsii, Rickettsia typhi,
  • pseudomallei Neisseria gonorrhoeae, Neisseria meningitides, Campylobacter jejuni, Helicobacter pylori, Legionella pneumophila, Acinetobacter baumannii, Moraxella catarrhalis, Pseudomonas aeruginosa, Aeromonas sp., Vibrio cholerae, Vibrio parahaemolyticus, Thiotrichales sp., Haemophilus influenzae, Klebsiella pneumoniae, Proteus mirabilis, Yersinia pestis, Yersinia enterocolitica, Shigella flexneri, Salmonella enterica, and Escherichia coli.
  • Viral infections may include a mammalian infection by any viral species.
  • Embodiments of the viral species may include, but are not limited to, single-stranded DNA viruses, including the genus Parvoviridae, double-stranded DNA viruses, including the genus Papillomaviridae, Polyomaviridae, Poxviridae, Herpesviridae (ex. Herpes Simplex Virus 1, 2, 6, Varicella-zoster virus, Epstein-Barr virus EBV, Human cytomegalovirus, and human herpesvirus 7), single-stranded RNA viruses, Reo- and Retroviruses (ex. Hepatitis B virus, Human immunodeficiency virus 1 and 2, and Human T-lymphotropic virus 1).
  • Fungal infections may include a mammalian infection by any bacterial species.
  • Embodiments of the fungal species, as used herein, may include, but are not limited to, Fusarium oxysporum, Pneumocystis jirovecii, Aspergillus spp., Coccidioides immitis/posadasii, Candida albicans, Filobasidiella neoformans, Trichosporon, Encephalitozoon cuniculi, Enterocytozoon bieneusi, Mucor circinelloides, Rhizopus oryzae, and Lichtheimia corymbifera.
  • Parasite infections may include a mammalian infection by any parasite.
  • Embodiments of the parasite may be, but are not limited to, Entamoeba histolytica, Babesia microti, Babesia sp. WA1, Trypanosoma cruzi, Taenia solium, Echinococcus granulosus, Leishmania braziliensis, L. donovani, L. tropica, Plasmodium falciparum, Plasmodium vivax, Plasmodium ovale and Plasmodium malariae, Plasmodium knowlesi, Paragonimus westermani, chistosoma sp., S. mansoni, S.
  • Exemplary embodiments of diagnostic markers include those listed in Table III. Further exemplary embodiments for the diagnosis of type-II diabetes include, but are not limited to, Albumin, Alpha-1 antitrypsin (A1AT)—G Protein, Cystatin C, Cystatin A, alpha-2-macroglobulin (A2MG), Uteroglobin, Transthyretin (TTR), Annexin A1, Annexin A2, Annexin A3 and Calnexin.
  • A1AT Alpha-1 antitrypsin
  • A2MG alpha-2-macroglobulin
  • TTR Transthyretin
  • Annexin A1, Annexin A2, Annexin A3 and Calnexin Calnexin.
  • Diagnostic markers directed towards cardiovascular disease may include, but are not limited to, any isoform of creatine kinase, troponin I and T, LD, Myoglobin, ALT/AST, H-FABP, and Glycogen phosphorylase B.
  • Diagnostic markers directed towards cancer may include, but are not limited to, Aldose reductase, Angiogenin, Annexin A1, B-cell activating factor (BAFF), B-cell lymphoma 2 (BCL2)-like 2, Beta Human chorionic gonadotropin, Ca15-3, Calcyclin, Calvasculin, Cathepsin D, Caveolin-1, Chromogranin A, Alpha-crystallin B chain (CRYAB), Endostatin, Eotaxin-2, Epithelial cell adhesion molecule (EpCAM), Ezrin, fatty acid binding protein 4 (FABP4), Galectin-3, ⁇ -glutamylcysteine ligase regulatory chain (GCLR), Gelsolin, Glucose 6-phosphate (G6P), Glycoprotein 130 (gp130), Glutathione S-transferase Mu 1 (GSTM1), Hepsin, High-mobility group protein B1 (HMGB-1), Insulin
  • BAFF B-cell activating
  • Diagnostic markers directed towards a bacterial or virus may include any surface or secreted antigen from the bacteria or virus, or a conserved nucleotide sequence.
  • diagnostic markers may include one or more of: 1,25 dihydroxy-vitamin D, 17-Hydroxyprogesterone, 25-hydroxy-vitamin D, Antineutrophil Cytoplasmic Antibodies, 5-Hydroxy Tryptamine, 5-hydroxyindoleacetic acid, Acetoacetate, Activated Partial Thromboplastin Time, Adrenocorticotropic Hormone, Alanine aminotransferase, Alanine transaminase, Albumin, Albumin-to-Creatinine ratio, Albumin/Globulin ratio, Alcohol, Aldolase, Aldosterone, Aldosterone and plasma renin activity, Aldosterone and Renin, Alkaline Phosphatase, Allergen-specific IgE, Alpha tryptase, Alpha-1 Antitrypsin, Alpha-fetoprotein, Alpha1-antitrypsin, Alzheimer biomarkers (including Tau protein and Amyloid Beta 42 peptides
  • Amylase is an enzyme that breaks down starch into sugar, and is found in such places as human saliva, where it begins the chemical process of digestion.
  • the ⁇ -amylases are calciummetalloenzymes that are completely unable to function in the absence of calcium. Through acting at random locations on the starch chain, ⁇ -amylase breaks down carbohydrates into maltotriose and maltose from amylase. In animals, ⁇ -amylase is a major digestive enzyme that has an optimum pH of 6.7 to 7.0.
  • the stabilizing agent may be an inhibitor of ⁇ -amylase. Further, the stabilizing agent may be active in a biological fluid, such as in saliva.
  • a stabilizing agent may comprise one or more of Fixanal® Buffer 6.0 (Sigma-Aldrich Co.), aluminum sulfate hydrate, aluminum hydroxide, bentonite, and aluminum potassium sulfate dodecahydrate.
  • Lysozyme also known as muramidase or N-acetylmuramide glycanhydrolase, is a glycoside hydrolase, that damage bacterial cell walls by catalyzing hydrolysis of 1,4-beta-linkages between N-acetylmuramic acid and N-acetyl-D-glucosamine residues in a peptidoglycan and between N-acetyl-D-glucosamine residues in chitodextrins. Lysozyme is abundant in a number of secretions, such as tears, saliva, human milk, and mucus. It is also present in cytoplasmic granules of the polymorphonuclear neutrophils (PMN).
  • PMN polymorphonuclear neutrophils
  • a stabilizing agent of the present disclosure may be an inhibitor of lysozyme. Further, the stabilizing agent may be active in a biological fluid, such as saliva.
  • a stabilizing agent may comprise one or more of Fixanal® Buffer 6.0 (Sigma-Aldrich Co.), bentonite, benzoic acid, and acetic acid.
  • Peroxidase are a class of oxidoreductase enzymes that catalyze the oxidation of a compound by the decomposition of hydrogen peroxide or an organic peroxide. These peroxidases may be found in many different bodily fluids. For example, saliva contains both salivary peroxidase (SPX) and myeloperoxidase (MPO). These peroxides may act on a number of different diagnostic markers found in bodily fluids to mask the detection or analysis of the markers.
  • SPX salivary peroxidase
  • MPO myeloperoxidase
  • the stabilizing agent may be an inhibitor peroxidase activity. Further, the stabilizing agent may be active in a biological fluid, such as saliva, blood, serum, or cancer cells.
  • a stabilizing agent may comprise one or more of aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate.
  • the stabilizing agent may preserve a diagnostic marker for insulin resistance and/or glucose intolerance.
  • the stabilizing agent may be active in a biological fluid, such as saliva.
  • a diagnostic marker may be one or more of ⁇ -hydroxybutyrate, 1-linoleoyl-GPC, palmitate, Glycine, 3-methyl-2-oxybutyrate.
  • a diagnostic marker may be any marker of early stage insulin resistance and glucose intolerance in a non-diabetic individual.
  • Galactose oxidase is a member of the family of oxidoreductaases, and has been shown to participate in galactose metabolism. Because of this activity, galactose oxidase may be used to detect mucin-like glycoproteins. These glycoproteins are a major component of mucus secreted by epithelial and glandular cells and are primarily responsible for the protective properties of the viscoeleastic mucous barrier. Mucins have been implicated in the process of cholesterol gallstone formation, and has been identified as having abnormal expression in some cancers.
  • the stabilizing agent may be an inhibitor of galactose oxidase. Further, the stabilizing agent may be active in a biological fluid, such as saliva.
  • a stabilizing agent may comprise one or more of aluminum potassium sulfate dodecahydrate, 3-tert-butyl-hydroxyanisole, benzoic acid, and acetic acid.
  • HbA1c Glycated hemoglobin
  • HbA1c is a form of hemoglobin that is indicative of the average plasma glucose concentration over a period of time. Due to the transport of components of the plasma into bodily fluids, such as saliva, the level of HbA1c may be determined in bodily fluids in addition to plasma.
  • the stabilizing agent may be an inhibitor of HbA1c degradation. Further, the stabilizing agent may be active in a biological fluid, such as saliva.
  • a stabilizing agent may comprise one or more of acetic acid, aluminum hydroxide bentonite, aluminum sulfate hydrate, aluminum potassium sulfate dodecahydrate, benzoic acid, caffeine, and 3-tert-butyl-hydroxyanisole, or a combination thereof.
  • Cancer Antigen 19-9 (CA 19-9), which may in some instances be referred to as Cancer Angigen 19.9, Cancer antigen-GI or CA-GI, is an antigen associated with various cancers, such as prostate and colon cancer. At least one use of CA 19-9 may be see whether a pancreatic tumor is secreting antigen. If that is the case, then the levels may decrease when the tumor is treated, and they may rise again if the disease recurs.
  • the stabilizing agent may be an inhibitor of CA 19-9 degradation. Further, the stabilizing agent may be active in a biological fluid, such as saliva, blood, serum, or cancer cells.
  • a stabilizing agent may comprise one or more of aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate.
  • the components of a stabilizing agent may be present at or about equal amounts, such as for example 1:1:1 in a stabilizing agent with three active components.
  • the stabilizing agents may in at least one embodiment inhibit degradation or inactivation of a diagnostic marker by at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99%.
  • the stabilizing agent may, in at least one embodiment, inhibit degradation of a diagnostic marker for at least about 1 minute, at least about 5 minutes, at least about 10 minutes, at least about 15 minutes, at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 4 hours, or at least about 8 hours.
  • stabilizing agents may be incorporated into various carriers for their use with mammals.
  • Such carriers may include various rinses, gums, lozenge, mouthwash, beverages, confectionary, washes, or other applicable vehicles to deliver a stabilizing agent to the site of a diagnostic marker.
  • Embodiments of a carrier, such as a rinse, according to the present disclosure may also comprise one or more additional component which may include an anti-caking agent, a chemical preservative, an emulsifying agent, a nutrient and dietary supplement, a sequestrant, a stabilizer, an additive, a synthetic and flavoring substance. Exemplary embodiments of these components are listed herein (See section entitled “Additional Components for Rinse”). Further, the one or more additional component may be a substance which has been labeled as Generally Recognized As Safe (GRAS) by the Food and Drug Administration of the United States of America.
  • GRAS Generally Recognized As Safe
  • a exemplary embodiment of a carrier of the present disclosure may be a mouthwash composition which may comprise water and pharmaceutically acceptable excipients or additives such as: one or more oils, such as an oil selected from the group comprising anethole, anisole, camphor, methyl salicylate, vanillin, eugenol, furaneol, linalool, menthol, thymol, cinnamaldehyde, citral, methyl butanoate, pentylbutanoate, pentylpentanoate, tea tree oil, peppermint oil, spearmint oil, pineapplemint oil and eucalyptus oil; sweetening agents, for example sorbitol; thickening agents, such as xanthan gum, carrageenan, carbomer, or HPMC (hydroxypropyl methyl cellulose); preservative agents, such as sodium benzoate, methyl paraben, or propyl paraben; emulsifiers, such as polysorbate
  • Gum compositions may be provided in a variety of different forms, such as, for example, slab, pellet, sticks, balls, cubes, center-fill gums, candy gum, multi-region gum, multi-layer gum, bubble gum, deposited gums and compressed gums.
  • the gum compositions also may include at least one flavor and a variety of optional additives.
  • the gum compositions described herein may be formed in a rope or sheet form and then processed into individual forms as described above.
  • a gum composition also may include a gum base.
  • the gum base may include any component known in the chewing gum art. Such components may be water soluble, water-insoluble or a combination thereof.
  • the gum base may include elastomers, bulking agents, waxes, elastomer solvents, emulsifiers, plasticizers, fillers and mixtures thereof.
  • a ingestible beverage composition of the present disclosure may comprise any formulation commonly known in the art.
  • a carrier of the present application may comprise a liquid.
  • the liquid in at least one embodiment, is a non-toxic liquid. Further, the liquid may comprise water, a beverage containing water, or glycerin. Moreover, in at least one embodiment, a stabilizing agent may be at least partially dissolved in the liquid.
  • the following components in at least one embodiment of the rinse of the present disclosure, may include:
  • ANTI-CAKING AGENTS aluminum calcium silicate, calcium silicate, magnesium silicate, sodium calcium aluminosilicate, and tricalcium silicate.
  • CHEMICAL PRESERVATIVES ascorbic acid, ascorbyl palmitate, benzoic acid, butylated hydroxyanisole, butylated hydroxytoluene, calcium ascorbate, calcium propionate, calcium sorbate, caprylic acid, dilauryl thiodipropionate, erythorbic acid, gum guaiac, methylparaben, potassium bisulfite, potassium metabisulfite, potassium sorbate, propionic acid, propyl gallate, propylparaben, sodium ascorbate, sodium benzoate, sodium bisulfite, sodium metahisulfite, sodium propionate, sodium sorbate, sodium sulfite, sorbic acid, stannous chloride, sulfur dioxide, thiodipropionic acid, tocopherols.
  • EMULSIFYING AGENTS cholic acid, desoxycholic acid, diacetyl tartaric acid esters of (M)mono- and diglycerides, glycocholic acid, mono- and diglycerides, monosodium phosphate derivatives of above, propylene glycol, ox bile extract, taurocholic acid.
  • NUTRIENTS AND DIETARY SUPPLEMENTS alanine, arginine, ascorbic acid, aspartic acid, biotin, calcium carbonate, calcium citrate, calcium glycerophosphate, calcium oxide, calcium pantothenate, calcium phosphate, calcium pyrophosphate, calcium sulfate, carotene, choline bitartrate, choline chloride, copper gluconate, cuprous iodide, cysteine, cystine, ferric phosphate, ferric pyrophosphate, ferric sodium pyrophosphate, ferrous gluconate, ferrous lactate, ferrous sulfate, glycine, histidine, inositol, iron (reduced), isoleucine, leucine, linoleic acid, lysine, magnesium oxide, magnesium phosphate, magnesium sulfate, manganese chloride, manganese citrate, manganese gluconate, manganese glycerophosphat
  • SEQUESTRANTS calcium acetate, calcium chloride, calcium citrate, calcium diacetate, calcium gluconate, calcium hexametaphosphate, calcium phosphate. monobasic, calcium phytate, citric acid, dipotassium phosphate, disodium phosphate, isopropyl citrate, monoisopropyl citrate, potassium citrate, sodium acid phosphate, sodium citrate, sodium diacetate, sodium gluconate, sodium hexametaphosphate, sodium metaphosphate, sodium phosphate, sodium potassium tartrate, sodium pyrophosphate, sodium pyrophosphate, tetra, sodium tartrate, sodium thiosulfate, sodium tripolyphosphate, stecaryl citrate, tartaric acid.
  • STABILIZERS acacia (gum arabic) , agar-agar, ammonium alginate, calcium alginate, carob bean gum, chondrus extract, ghatti gum, guar gum, potassium alginate, sodium alginate, sterculia (or karava) gum, tragacanth.
  • ADDITIVES acetic acid, adipic acid, aluminum ammonium sulfate, aluminum potassium sulfate aluminum sodium sulfate, aluminum sulfate, ammonium bicarbonate, ammonium carbonate, ammonium hydroxide, ammonium phosphate, ammonium sulfate, bees wax, bentonite, butane, caffeine, calcium carbonate, calcium chloride, calcium citrate, calcium gluconate, calcium hydroxide, calcium lactate, calcium oxide, calcium phosphate, caramel, carbon dioxide, carnauba wax, citric acid, dextrans, ethyl formate, glutamic acid, glutamic acid hydrochloride, glycerin, glyceryl monostearate, helium, hydrochloric acid, hydrogen peroxide, lactic acid, lecithin, magnesium carbonate, magnesium hydroxide, magnesium oxide, magnesium stearate, malic acid, methylcellulose, monoammonium glutamate, monopotassi
  • SYNTHETIC FLAVORING SUBSTANCES acetaldehyde, acetoin, aconitic acid, anethole, benzaldehyde, N-butyric acid, d- or l-carvone cinnamaldehyde, citral, decanal, diacetyl, ethyl acetate, ethyl butyrate, ethyl vanillin, eugenol, geraniol, geranyl acetate, glycerol tributyrate limonene, linalool, linalyl acetate, l-malic acid, methyl anthranilate, 3-methyl-3-phenyl glycidic acid, ethyl ester, piperonal, vanillin.
  • the carrier comprises an indicator compound capable of binding to and/or reacting with a target to produce an indicator signal.
  • the target in an exemplary embodiment may be a diagnostic marker, such as the presence of glucose or glucose in excess of a defined threshold, the presence of one or more glycosylated protein related to diabetes or pre-diabetes, the presence of cancer (or pre-cancer) markers, and the presence of cardiac (or pre-cardiac) markers.
  • the indicator compound in an exemplary embodiment of a carrier according to the present disclosure may be any compound, chemical, or biological component which may interact with a target or byproduct of a target.
  • an indicator compound may comprise an antibody, a reactive chemical compound, a labeled molecule, or any combination thereof.
  • Antibodies used in an embodiment of the present disclosure may be monoclonal or polyclonal and derived from any species (e.g. human, rat, mouse, rabbit, pig).
  • indicator molecules may be aptamers, proteins, peptides, small organic molecules, natural compounds (e.g. steroids), non-peptide polymers, WIC multimers (including MHC-dextramers, MHC-tetramers, MHC-pentamers and other MHC-multimers), or any other molecules that specifically and efficiently bind to other molecules are also marker molecules.
  • Labeled molecules for use as indicator compounds, may be any molecule that absorbs, excites, or modifies radiation, such as the absorption of light (e.g. dyes and chromophores) and the emission of light after excitation (fluorescence from flurochromes). Additionally, labeled molecules may have an enzymatic activity, by which it catalyzes a reaction between chemicals in the near environment of the labeling molecules, producing a signal which include production of light (chemi-luminescence) or precipitation of chromophors, dyes, or a precipitate that can be detected by an additional layer of detection molecules.
  • Fluorescence labels may produce the presence of light at a single wavelength, or a shift in wavelengths.
  • Exemplary fluorescent labels may include:
  • the target of an exemplary indicator compound of the present disclosure may be any diagnostic marker or diagnostic condition for a disease state (or pre-disease state) of an individual.
  • the disease state may be diabetes, pre-diabetes, cardiac disease, pre-cardiac conditions, cancers of any stage and type, or pre-cancerous conditions.
  • the target may be the presence, or level of, glucose found in saliva for diabetes.
  • the target may be the presence, or level of, glycosylated proteins, such as advanced glycosylation end products.
  • the target may be the presence or level of cardiac markers, such as Creatine kinase-MB (CK-MB), myoglobin, homocysteine, C-reactive protein (CRP), troponin T (cTnT), and troponin I (cTnI).
  • cardiac markers such as Creatine kinase-MB (CK-MB), myoglobin, homocysteine, C-reactive protein (CRP), troponin T (cTnT), and troponin I (cTnI).
  • the target may be the presence of cancer markers, such as Cancer Antigen 125, Cancer Antigen 15.3, Cancer Antigen 19.9, Prostate specific antigen, Carcinoembryonic Antigen, Alpha Feto-Protein, Epidermal growth factor receptor, Kallikrein 3, Vascular endothelial growth factor A (VEGF), Calcitonin, Chromogranin A, Gastrin, S100 alpha chain, Somatostatin, Thyroglobulin, V-erb-b2, cyckub-dependent kinase inhibitor 1 (p21), Breast Cancer Antigen 1 and 2 (BRCA1 and 2), MutL homolog 1 (MLH1), MutS homolog 2 (MSH2), MutS homolog 6 (MSH6), and postmeiotic segregation increased 1 and 2 (PMS1 and 2).
  • the target may be the presence or level of any diagnostic marker included herein.
  • the indictor signal in at least one embodiment of the present disclosure may comprise any detectable signal, including but not limited to, color change, fluorescence, and chemical/structural change of the target (and/or indicator compound) so as to be amenable to reacting with a secondary detection marker. Further, the detection of a signal may involve a secondary reactive molecule.
  • glucose may be detected through an indicator rinse by a chemical or enzymatic method.
  • exemplary embodiments of this method may include alkaline copper reduction, alkaline ferricyanide reduction, glucose oxidase, and/or hexokinase.
  • the rinse may further comprise one or more additional component as described herein.
  • additional components may include stabilizing agents, as well as GRAS components (such as those listed herein).
  • glycated Hemoglobin A1c may be detected through any known method for the detection of HbA1c or fragments thereof, such as high pressure liquid chromatography, immunoassays, and an indicator rinse by a chemical or enzymatic method.
  • the rinse may further comprise one or more additional component as described herein. These additional components may include stabilizing agents, as well as GRAS components (such as those listed herein).
  • cancer antigen 19-9 may be detected through any known method for the detection of CA 19-9 or fragments thereof, such as high pressure liquid chromatography, immunoassays, and an indicator rinse by a chemical or enzymatic method.
  • the rinse may further comprise one or more additional component as described herein. These additional components may include stabilizing agents, as well as GRAS components (such as those listed herein).
  • a diagnostic marker susceptible to degradation or alteration by a destructive component may be detected through any known method, including, but not limited to, high pressure liquid chromatography, immunoassays, and an indicator rinse by a chemical or enzymatic method. Further, the detection may be conducted in a micro well, deep well, microarray, microfluidic device, high throughput screen, or any applicable diagnostic device.
  • a detection platform 100 may comprise a platform 102 having a plurality of test wells 104 , where each well is capable of containing a plurality of test spots 106 .
  • the plurality of test wells 104 may include at least 2, at least 4, at least 8, at least 16, at least 32, at least 64, or at least 256 wells.
  • the plurality of tests spots 106 may include at least 2, at least 4, at least 8, at least 16, at least 32, or at least 64 test spots 106 . Further, each of the plurality of test spots 106 may comprise one or more diagnostic marker or control marker.
  • An embodiment of detection platform 100 may further be sized and shaped to allow for automated detection and analysis of a diagnostic or control marker.
  • the detection system 100 may comprise an identifier 108 , such as a barcode or Radio-frequency identification (RFID) tag, to allow for the identification of the detection system 100 and data collected from the analysis of test spots 106 .
  • RFID Radio-frequency identification
  • detection platform 100 may be comprised of any applicable material sufficient to house or define test wells 104 .
  • applicable materials for detection platform 100 may be a polymer, glass, metal, quartz, nylon, silica-based material, resin, or combination thereof.
  • embodiments of test wells 104 may include not only a depression defined or housed by platform 102 , but may also include a defined area or region of the platform 102 (such as may be found on a microarray based platform).
  • An exemplary embodiment of detection system 150 comprises an embodiment of detection platform 100 , a detection device 120 capable of determining a binding characteristic between a detection agent and a diagnostic marker on the detection platform 100 , and a computer processor 130 coupled to a computer database 135 and to the detection device 120 .
  • the computer processor 130 controlling the detection device 120 is able to (1) determine the binding characteristic of a detection agent to a diagnostic agent, (2) compare the binding characteristic among each of a plurality of samples tested, (3) generate a binding report using the compared binding characteristics, and deliver the binding report to a recipient or external computer (not shown) in communication with the computer processor 130 .
  • a binding characteristic as used herein may include any measurement of the attraction (or repulsion) of two molecules (such as a detection agent and a diagnostic marker).
  • Detection system 100 may also comprise an embodiment of a bead-based system of the present disclosure having at least one bead type.
  • a dual bead system may be used wherein at least one bead type is magnetic or paramagnetic.
  • These beads may also be bound to a carrier protein with a conjugated hapten. Further, the beads may have an analyte capture monoclonal antibody reagent bound to it. Additionally, the beads, in at least one embodiment, do not retain their magnetic properties when removed from a magnetic field.
  • the beads may be Europium micro-particles. The sizes of the Europium micro-particles may be varied as needed to accommodate the method of detection used.
  • At least one embodiment of a bead based system may comprise a latex particle (See FIGS. 2-4 ).
  • the latex particle may be a Carboxylate modified latex (CML) bead.
  • CML Carboxylate modified latex
  • an exemplary embodiment of a bead may include bovine serum albumin (BSA) and/or human serum albumin (HSA).
  • BSA bovine serum albumin
  • HSA human serum albumin
  • an embodiment of a bead based system of the present disclosure may include a linker coupled to HSA and/or BAS.
  • the linker according to an embodiment, may comprise a maleimide compound.
  • At least one exemplary embodiment of a maleimide compound may be Succinimidyl 4-[N-Maleimidomethyl] Cyclohexane-1-Carboxylate (SMCC) or N-Hydroxysuccinimide-activated hexa(ethylene glycol) undecane Thiol (NHS).
  • SMCC Cyclohexane-1-Carboxylate
  • NHS N-Hydroxysuccinimide-activated hexa(ethylene glycol) undecane Thiol
  • at least one exemplary embodiment of a bead based system may further comprise an antibody linked to maleimide on the bead.
  • at least one embodiment of the bead based system comprises a latex particle coupled to BAS or HSA, maleimide coupled to BAS or HSA, and an antibody coupled to maleimide.
  • the latex particle may be in a size range from about 0.02 ⁇ m to about 7.0 ⁇ m.
  • At least one at least one method 500 of coupling an antibody to a bead comprises the step 502 of modifying an embodiment of a bead (such as a latex bead/particle) with BSA or HAS, wherein the BSA or HSA may further comprise a maleimide containing molecule, such as SMCC or NHS. Additionally, an embodiment of the method 500 may further comprise the step 504 of attaching a diagnostic marker binding agent (such as an antibody) to the bead. Step 504 of attaching the diagnostic marker binding agent to the bead may be accomplished through binding the diagnostic marker binding agent to maleimide. Further, method 500 may also comprise the step 506 of attaching one or more stabilizing agent to an embodiment of a bead. Additionally, while an embodiment of method 500 may use latex beads for attachment of a maleimide containing molecule and/or a stabilizing agent, alternate embodiments of the bead may comprise silicon, a polymer, or other applicable materials.
  • one or more stabilizing agent may be attached or adsorbed to any one of an embodiment of a detection platform 100 , test strip, microchip, or microfluidic device.
  • An exemplary embodiment of diagnostic testing device 600 comprises housing structure 602 which comprises a collection chamber 604 capable of receiving body fluids, at least one membrane test strip 606 in fluid communication with collection chamber 604 , an immunoassay-based fingerprint acquisition pad 608 in fluid communication with the collection chamber, and a plurality of reaction zones 610 which may allow the visual display of the presence of a predetermined diagnostic marker.
  • reaction zone 610 may in an exemplary embodiment be capable of displaying a characteristic of the predetermined diagnostic marker, such as the presence of, the concentration of, or a concentration above or below a predetermined value for the predetermined diagnostic agent.
  • diagnostic testing device 600 further comprises a fluid collector 612 which is operable to collect body fluids from a subject, and wherein the fluid collector is capable of supply collected body fluid to collection chamber 604 .
  • test strip 606 may comprise any material capable of adsorbing or attaching a stabilizing agent and may bind a diagnostic marker. Further, exemplary embodiments of test strip 606 may comprise compositions comprising a polyvinyl chloride-silica combination, nitrocellulose, or any suitable synthetic, resinous material. At least one embodiment, housing 602 is capable of reducing the exposure of the test strip 606 at sites other than the collection chamber 604 to foreign material. Test strip 606 in at least one embodiment may be capable of interfacing with an analysis device 615 . The analysis device 615 in an exemplary embodiment may be capable of measuring the level of the predetermined diagnostic marker in the sample.
  • An exemplary embodiment of a microarray system 620 may comprise a microarray product 622 having a microarray identifier 624 , and a plurality of microarray sites 626 , a control microarray product 628 located on at least one microarray site 626 and bound to microarray product 622 , and operably coupled to a computer processor 130 for providing information regarding the identification and concentration of markers on the microarray product 622 based on the microarray identifier 624 .
  • microarray product 622 may further comprise, in at least one embodiment, an embodiment of a stabilization agent 630 located on at least one microarray site 626 and bound to microarray product 622 . Further, microarray product 622 may comprise one or more diagnostic marker 632 located on at least one microarray site 626 and bound to microarray product 622 . Moreover, microarray system 620 , in at least one embodiment, may further comprise a microarray detector 634 operable to detect a signal from at least one of the control microarray product 628 and/or diagnostic marker 632 .
  • a microchip or microarrays may refer to an array of distinct polynucleotides affixed to a substrate, such as glass, plastic, paper, nylon or other type of membrane, filter, chip, or any other suitable solid support.
  • the polynucleotides can be synthesized directly on the substrate, or synthesized separate from the substrate and then affixed to the substrate.
  • the microarray may be prepared and used at least according to the methods described in U.S. Pat. No. 5,837,832, Chee et al., PCT application WO95/11995 (Chee et al.), Lockhart, D. J. et al. (1996; Nat. Biotech. 14: 1675-1680) and Schena, M. et al. (1996; Proc. Natl. Acad. Sci. 93: 10614-10619), all of which are incorporated herein in their entirety by reference.
  • microfluidic device 640 comprises a sample reservoir 642 for receiving a body fluid (or other fluid sample) through a sample input 644 , where the sample reservoir 642 is fluidly connected to a detector array 646 .
  • detector array 646 comprises a reaction site 647 and a results display 648 .
  • an exemplary embodiment of microfluidic device 640 may further comprise a filter device 650 capable of separating at least one unwanted component from a fluid sample and fluidly coupled between sample reservoir 642 and detector array 646 .
  • Detector array 646 may also, in at least one embodiment comprise a control reagent display 652 , where at least one control reagent may be visually detected.
  • microfluidic device 640 may also, in at least one embodiment, be able to couple to a processor 655 .
  • Processor 655 may be able to compare at least one reading, such as from results display 648 or control reagent display 652 from an embodiment of microfluidic device 640 to at least one additional stored reading on a computer database 657 of the processor 655 .
  • An embodiment of microfluidic system 660 of the present disclosure may comprise one or more microfluidic device 640 operationally coupled to a processor 655 .
  • Embodiments of microfluidic devices 640 which may also be referred to as “lab-on-a-chip” systems, biomedical micro-electro-mechanical systems (bioMEMs), or multicomponent integrated systems, are exemplary kits/systems for analyzing polynucleotide regions. Such systems may miniaturize and compartmentalize processes such as probe/target hybridization, nucleic acid amplification, and capillary electrophoresis reactions in a single functional device. Such microfluidic devices 640 typically utilize detection reagents in at least one aspect of the system, and such detection reagents may be used to detect one or more polynucleotide region of the present disclosure.
  • Exemplary microfluidic devices 640 may comprise a pattern of microchannels designed onto a glass, silicon, quartz, or plastic wafer included on a microchip.
  • the movements of the samples may be controlled by electric, electroosmotic or hydrostatic forces applied across different areas of the microchip to create functional microscopic valves and pumps with no moving parts. Varying the voltage can be used as a means to control the liquid flow at intersections between the micro-machined channels and to change the liquid flow rate for pumping across different sections of the microchip.
  • the microarray or microfluidic device may comprise one or more stabilizing agent capable of reducing the degradation of a predefined diagnostic marker.
  • a stabilizing agent may be incorporated onto the surface of the microarray system 620 , and/or on at least one part of the microfluidic device 640 .
  • the method 700 comprises the step 702 of mixing a body fluid with an embodiment of a stabilizing agent.
  • the step of mixing a body fluid with a stabilizing agent may comprise the step 704 of introducing the stabilizing agent into the patient, such as through the oral cavity, so that the stabilizing agent mixes with the bodily fluid, and the step 706 of retrieving a body fluid comprising a stabilized diagnostic marker.
  • the method comprises the step 708 of isolating a body fluid having a diagnostic marker, the step 710 of treating the body fluid with an embodiment of a carrier, such as a rinse, gum, or beverage, as described herein, and the step 712 of analyzing the diagnostic marker.
  • Isolation of the body fluid may occur through any customary mechanism, including, but not limited to, rinsing, swabbing, suction, collection, and lavage.
  • the rinse in an exemplary embodiment of a method 700 of the present disclosure may be ingested to stabilize a diagnostic marker present in the bodily fluid, such as in urine, semen, anal secretions, and vaginal secretions.
  • the step 702 of mixing a body fluid with a carrier may occur prior to, or after isolation step 708 of the body fluid.
  • a rinse may be used to rinse the mouth.
  • the rinse containing the treated body fluid may be collected by spitting, suction, or other means.
  • expirated saliva may be mixed with a rinse to treat the saliva ex vivo. Similar treatment of body fluids may be performed with any type of body fluid.
  • the diagnostic marker may be analyzed through any known means.
  • the analysis of the treated body fluid may include the separation of solid materials from soluble materials through means such as filtration, or by centrifugation.
  • Analysis of the treated body fluid may use any appropriate technique, such as western blot analysis, Enzyme-Linked Immunosorbent Assay (ELISA), protein activity assays, reverse transcription polymerase chain reaction (RT-PCR), microarray, high pressure liquid chromatography, or any comparable assay to determine a characteristic of the diagnostic marker.
  • ELISA Enzyme-Linked Immunosorbent Assay
  • RT-PCR reverse transcription polymerase chain reaction
  • microarray a microarray
  • high pressure liquid chromatography or any comparable assay to determine a characteristic of the diagnostic marker.
  • Such an analysis in an exemplary embodiment, may be of a modified product, a cleavage product, or cleavage pattern, of a diagnostic marker.
  • the treated body fluid may be placed in contact with an embodiment of a test strip, such as a nitrocellulose strip, prior to detection with an appropriate probe specific for the diagnostic marker.
  • a test strip such as a nitrocellulose strip
  • the nitrocellulose strip may be designed to trap or filter particulates in the body fluid. This may reduce or eliminate potential contaminants or other substances in the oral fluid that would otherwise reduce the signal to noise ration during the detection step.
  • a probe (such as an antibody) with affinity to the diagnostic marker may be affixed or immobilized to a specific location on the nitrocellulose membrane for detection of the analyte. Utilizing standard principles of immunoassay detection (or nucleotide detection), the signal generated may be visible to the eye or detectable by an instrument.
  • the method 800 comprises the step 802 of operating a system for the detection of a diagnostic marker in a body fluid, where the system comprises a diagnostic device having a plurality of test wells each capable of containing at least one diagnostic marker binding agent, and a detection device capable of interacting with diagnostic device, wherein the detection device is capable of detecting an interaction between the at least one diagnostic marker binding agent and a diagnostic marker.
  • the sample of bodily fluid may be diluted in step 803 with a reagent which may contain an embodiment of stabilizing agent as described herein.
  • At least one embodiment of method 800 further comprises the steps of: contacting 804 a sample of a bodily fluid with at least one diagnostic marker binding agent, incubating 806 the contacted sample in the test well at a first temperature for a pre-determined period of time.
  • the step 804 of contacting of the sample of bodily fluid with at least one diagnostic marker binding agent may be accomplished through a magnetic field applied to the test well.
  • the first temperature may be at room or at body temperature (such as about 22° C. or about 37° C.).
  • the pre-determined period of time may, for example, be between five and fifteen minutes.
  • the incubated and contacted sample may then be removed in step 808 from the plurality of test wells, and the test wells analyzed in step 810 to detect a binding event with the detection device.
  • a stabilizing agent may be incubated with the diagnostic marker binding agent (also referred to as a detection agent) prior to introduction of a bodily fluid.
  • each diagnostic marker binding agent may have a different monoclonal antibody designed to bind a specific hapten molecule.
  • the characteristic may in step 812 be compared to a standard value to determine the presence or absence of a disease state.
  • the characteristic determined may be an activity level, the concentration of the diagnostic marker, or a particular modification, such as glycosylation or methylation.
  • an embodiment of the method 800 may further comprise the step 814 of customizing a detection method through screening of applicable pre-clinical samples through a database of stabilizing agents/cocktails.
  • an embodiment of method 800 of diagnostic marker detection may further comprise the step 816 of comparing a level or characteristic (such as a predictable degradation products) of a diagnostic agent with a library of known characteristics.
  • an exemplary embodiment of method 800 may additionally comprise the step 818 of determining a diagnosis of a disease state using the compared characteristic.
  • a method of diagnostic marker detection may be automated, and capable of detecting at least about ten thousand samples in a twenty-four hour period.
  • Method 800 in at least one embodiment, may be at least partially completed by a computer processor.
  • the processor may be in communication with at least one more processor and/or a computer database. For instance, in at least one embodiment of method 800 , most of the steps of the method may be completed with or monitored by a computer processor. Moreover, method 800 , in at least one embodiment, may use a computer processor to perform additional step 820 of generating a report by using a comparison of determined characteristics (such as a binding report where a comparison is performed of binding characteristics), and step 822 of delivering the report to a recipient (such as a user, or secondary processor).
  • a computer processor may use a computer processor to perform additional step 820 of generating a report by using a comparison of determined characteristics (such as a binding report where a comparison is performed of binding characteristics), and step 822 of delivering the report to a recipient (such as a user, or secondary processor).
  • a method of biomarker stabilization may, in an exemplary embodiment, may be used to diagnose a particular disease state or condition of a patient.
  • a disease state or condition as used with this method may include one or more of Acid-Base Disorders, Acidosis and Alkalosis, Acidosis/Alkalosis, Acute inflammatory demyelinating polyneuropathy, Acute myocardial infarct, Addison's Disease, Adrenal Insufficiency, Adrenal Insufficiency & Addison's Disease, Alcohol dependence, Alcoholism, Allergies, Alzheimer's Disease, Anemia, Angina Pectoris, Anthrax, Arthritis, Asthma, Atypical Pneumonia, Autoimmune Disorders, Autoimmune thyroiditis, Avian flu, Benign Prostatic Hyperplasia, Benign Prostatic Hypertrophy, Bioterrorism Agents, Bleeding Disorders, Bone Marrow Disorders, Breast Cancer, Cardiovascular Disease, Celiac Disease, Cerv
  • a computer processor and a database may be used in or during the system, method, or device.
  • An exemplary embodiment of a system framework comprising at least one computer processor and at least one database, as may be used in the embodiments of the present disclosure is shown in FIG. 9 .
  • a result from a diagnostic method may be compared using a computer processor to at least one additional result from the diagnostic test, and/or to a result stored on a database.
  • Such a comparison in at least one embodiment of the present disclosure, may reveal the stabilizing agent with a greater effect on the binding characteristic, may allow for the diagnosis of a disease state or health/lifestyle characteristic. Further, such a comparison, in at least one embodiment, may be used to develop, confirm, or modify a therapeutic treatment of a patient having a disease state or health/lifestyle characteristic.
  • one or more user computers 902 may be operably connected to a system server 904 .
  • a user computer 902 may be a computer, computing device, or system of a type known in the art, such as a personal computer, mainframe computer, workstation, notebook computer, laptop computer, hand-held computer, wireless mobile telephone, personal digital assistant device, and the like.
  • One or more administrator computers 906 may also be operably connected to system server 904 including through a network 908 such as the Internet.
  • Administrator computers 906 similar to user computers, may be computers, computing devices, or systems of a type known in the art, such as personal computers, mainframe computers, workstations, notebook computers, laptop computers, hand-held computers, wireless mobile telephones, personal digital assistant devices, and the like.
  • user computers 902 and administrator computers 906 may each comprise such software (operational and application), hardware, and componentry as would occur to one of skill of the art, such as, for example, one or more microprocessors, memory, input/output devices, device controllers, and the like.
  • User computers and administrator computers 906 may also comprise one or more data entry means (not shown in FIG.
  • User computers 902 and administrator computers 906 also may comprise an audio display means (not shown in FIG. 9 ) such as one or more loudspeakers and/or other means known in the art for emitting an audibly perceptible output.
  • the configuration of user computers and administrator computers 906 in a particular implementation of one or more systems of the present disclosure is left to the discretion of the practitioner.
  • System server 904 may comprise one or more server computers, computing devices, or systems of a type known in the art.
  • System server 904 may comprise server memory.
  • System server 904 may comprise one or more components of solid-state electronic memory, such as random access memory.
  • System server 904 may also comprise an electromagnetic memory such as one or more hard disk drives and/or one or more floppy disk drives or magnetic tape drives, and may comprise an optical memory such as a Compact Disk Read Only Memory (CD-ROM) drive.
  • System server 904 may further comprise such software (operational and application), hardware, and componentry, as would occur to one of skill of the art, such as, for example, microprocessors, input/output devices, device controllers, video display means, and the like.
  • System server 904 may comprise one or more host servers, computing devices, or computing systems configured and programmed to carry out the functions allocated to system server 904 .
  • System server 904 may be operated by, or under the control of, a “system operator,” which may be an individual or a business entity.
  • System server 904 is shown in FIG. 9 and referred to herein as a single server.
  • System server 904 need not, however, be a single server.
  • System server 904 may comprise a plurality of servers or other computing devices or systems connected by hardware and software that collectively are operable to perform the functions allocated to the various systems of present disclosure.
  • system server 904 may be operable to be a web server, configured and programmed to carry out the functions allocated to a system server according to the present disclosure.
  • user computers 902 and administrator computers 906 may be connected directly to system server 904 , these computers may be connected to system server 904 through any suitable network, such as network 908 .
  • the users need not be provided access to system server 904 , but instead the content posts from users are made by the user(s) and saved to one or more particular locations and the posts are accessed or harvested by the administrator or system automatically.
  • System server 904 may be operably connected to the various user computers 902 and/or an administrator computers 906 by network 908 , which in an embodiment of the present disclosure comprises the Internet, a global computer network. However, network 908 need not comprise the Internet. Network 908 may comprise any means for electronically interconnecting system server 904 and a user computer 902 and/or an administrator computer 906 . Thus, it will be appreciated by those of ordinary skill in the art that the network 908 may comprise the Internet, the commercial telephone network, one or more local area networks, one or more wide area networks, one or more wireless communications networks, coaxial cable, fiber optic cable, twisted-pair cable, the equivalents of any of the foregoing, or the combination of any two or more of the foregoing.
  • network 908 comprises the hardware and software means interconnecting system server 904 and user computer 902 and/or an administrator computer 906 within the single computing device.
  • Network 908 may comprise packet switched facilities, such as the Internet, circuit switched facilities, such as the public switched telephone network, radio based facilities, such as a wireless network, etc.
  • the various systems, methods, schema, ontologies, and architectures of the present disclosure may be used for purposes outside of the medical transcription field as referenced in the various examples cited herein.
  • the system for analyzing verbal records may comprise various components and relationships suitable for use in any number of areas where various experiences are utilized and processed, with feedback being fed back into system componentry to improve overall system outcomes.
  • various components described herein may share a name (or a portion thereof) but have duplicative reference numbers, and therefore the descriptions for the various components should read in view of one another.
  • such systems may be operable, as desired by a user of such systems, to generate visual, electronic (video, audio, database, transcript, etc.), and/or printed reports, outputs, outcomes, and the like.
  • Such exemplary outputs may be used for any number of purposes, and may be useful generally to “report” results, data, and/or knowledge contained within and generated from such systems.
  • the disclosure of the present application further encompasses uses of the various methods, systems, architectures, etc., to perform various tasks in connection therewith.
  • At least one assay used for the detection of ⁇ -amylases activity includes the use of a chromagenic substrate, 2-chloro-p-nitrophenol linked to maltotriose. Enzymatic activity of ⁇ -amylase on the substrate yields 2-chloro-p-nitrophenol, which can be measured spectrophotometrically at 405 nm. The amount of ⁇ -amylase activity present in the experimental sample is directly proportional to the increase in absorbance at 405 nm.
  • test compounds are mixed with saliva, prior to the addition of ⁇ -amylase substrate.
  • the test compounds such as GRAS materials, are prepared at a concentration of 5,000 ppm in distilled water. Following this preparation, 300 ⁇ l of ⁇ -amylase substrate (Salimetrics ⁇ -amylase Assay Kit) that has been pre-heated to 37° C. is added to 20 ⁇ l of the test compound.
  • ⁇ -amylase substrate saliva a concentration of 5,000 ppm in distilled water.
  • 300 ⁇ l of ⁇ -amylase substrate (Salimetrics ⁇ -amylase Assay Kit) that has been pre-heated to 37° C. is added to 20 ⁇ l of the test compound.
  • 10 ⁇ l of a dilute (3.5%) or full strength (100%) saliva solution is added to the mixture. Following the initiation, each mixture is measured at 1 minute intervals using a spectrophotometer (such as Molecular Devices M5 reader) at 405 nm. Temperatures of the mixture
  • At least one assay used for the detection of ⁇ -amylases activity includes the use of a Micrococcus lysodeikticus labeled with fluorescein.
  • the assay measures lysozyme activity on Micrococcus lysodeikticus cell walls, which are labeled to such a degree that the fluorescence is quenched. Lysozyme activity can relieve this quenching; yielding increased fluorescence that is proportional to lysozyme activity.
  • test compounds are mixed with saliva, prior to the addition of lysozyme substrate.
  • the test compounds such as GRAS materials, are prepared at a concentration of 5,000 ppm in distilled water. Following this preparation, 50 ⁇ l of lysozyme substrate at 1 mg/ml (Molecular Probes EnzChek Lysozyme Assay Kit) that has been pre-heated to 37° C. is added to 50 ⁇ l of the test compound. To initiate the assay, 50 ⁇ l of a dilute (3.5%) or full strength (100%) saliva solution is added to the mixture.
  • each mixture is measured at 1 minute intervals using a spectrophotometer (such as Molecular Devices M5 reader) for absorption at 494 nm and fluorescence emission at 518 nm. Temperatures of the mixtures are kept constant at 37° C. during the assay.
  • a spectrophotometer such as Molecular Devices M5 reader
  • At least one assay used for the detection of galactose oxidase activity includes the use of a chromagenic or fluorogenic substrate, such as with Amplex® Red (Molecular Probes).
  • a chromagenic or fluorogenic substrate such as with Amplex® Red (Molecular Probes).
  • galactose oxidase catalyzes the oxidation of galactose at the C6 position and generates hydrogen peroxide (H2O2).
  • H2O2 then, in the presence of horseradish peroxidase (HRP), reacts with 1:1 stoichiometry with Amplex® Red reagent to generate the red-fluorescent oxidation product, resorufin.
  • HRP horseradish peroxidase
  • Resorufin has absorption and fluorescence emission maxima of approximately 571 nm and 585 nm, respectively, and because the extinction coefficient is high (54,000 cm-1M-1), the assay can be performed either fluorometrically or spectrophotometrically.
  • test compounds are mixed with Amplex Red, reaction buffer, horseradish peroxidase (HRP) at 100 U/ml, and galactose stock solution (according to manufacturers recommended protocol, Molecular Probes A22179), prior to the addition of saliva.
  • the test compounds such as GRAS materials, are prepared under various concentration for testing. Following this preparation, 50 ⁇ l of the test compound solution (with Amplex Red, reaction buffer, and HRP) is added to 50 ⁇ l of saliva. Following the initiation, each mixture is measured at 1 minute intervals using a spectrophotometer (such as Molecular Devices M5 reader) using excitation in the range of 530-560 nm and emission detection at about 590 nm or absorbance at 560 nm. Temperatures of the mixtures are kept constant at 37° C. during the assay.
  • At least one procedure used for the stabilization and detection of glycosylated hemoglobin includes the incubation of HbA1c with a stabilizing agent prior to, or concurrently with, detection.
  • Analysis of samples was conducted on a HPLC 1100 series (Agilent Technologies) with a 2.1 ⁇ 150 (3.5 ⁇ ) C18 reverse phase column. The mobile phase used was acetonitrile with 6.5 mM Ammonium Carbonate.
  • a HbA1c standard was prepared by dissolving HbA1c (Sigma Aldrich, 405 RM) with 1.0 ml of deionized water.
  • FIGS. 14 100 ml HbA1c mixed with 100 ml of fresh saliva
  • FIGS. 15 200 ml HbA1c mixed with 100 ml of fresh saliva and 100 ml of a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1:1:1 ratio)
  • a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1:1:1 ratio)
  • FIGS. 14B , 15 B, and 16 B a chromatogram
  • FIGS. 14C , 15 C, and 16 C a comparison of the analysis HbA1c in the presence of saliva with and without stabilizing agent is shown in FIGS. 17 (chromatography), 18 (UV image), and 19 (three dimensional plot).
  • a series of GRAS compounds were tested, as shown in FIGS. 14-16 , for the ability to protect HbA1c from degradation by components in saliva.
  • the tested compounds are included in Table I.
  • a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1:1:1 ratio) minimized the degradation of HbA1c, and yielded a defined pattern for HbA1c.
  • At least one procedure used for the stabilization and detection of Cancer Antigen 19-9 includes the incubation of CA 19-9 with a stabilizing agent prior to, or concurrently with, detection. Analysis of samples was conducted on a HPLC 1100 series (Agilent Technologies) with a 2.1 ⁇ 150 (3.5 ⁇ ) C18 reverse phase column. The mobile phase used was acetonitrile with 6.5 mM Ammonium Carbonate. A CA 19-9 standard was prepared by dissolving CA 19-9 with 1.0 ml of deionized water. The samples analyzed by HPLC included pure CA 19-9 ( FIG. 20 ), 100 ml CA 19-9 mixed with 100 ml of fresh saliva ( FIG.
  • FIG. 21 200 ml CA 19-9 mixed with 100 ml of fresh saliva and 100 ml of a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1:1:1 ratio)
  • a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1:1:1 ratio)
  • FIGS. 20A , 21 A, and 22 A samples of CA 19-9 with saliva were incubated overnight at room temperature prior to analysis.
  • the data was displayed through a three dimensional plot ( FIGS. 20A , 21 A, and 22 A) and a UV image ( FIGS. 20B , 21 B, and 22 B).
  • FIGS. 23 three dimensional plot
  • 24 UV image
  • a series of GRAS compounds were tested, as shown in FIGS. 20-22 , for the ability to protect CA 19-9 from degradation by components in saliva.
  • the tested compounds are included in Table I.
  • a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1:1:1 ratio) minimized the degradation of CA 19- 9 in at least one embodiment.
  • test compounds such as GRAS materials or cocktails of same, were prepared at a concentration of 500 ppm in distilled water.
  • 50 ⁇ l samples of test compounds were mixed with 20-30 ⁇ l of saliva (either “pooled” or “unpooled”) prior to the addition of Amplex® Red.
  • 50 ⁇ l of Amplex® Red/HRP working solution See Protocol for Invitrogen Assay #A22188, which is incorporated herein in its entirety, which has a concentration of 100 ⁇ M of Amplex® Red, is added to the test compound/saliva mixture.
  • each mixture is measured at 1 minute intervals using a spectrophotmoter (such as a Molecular Devices M5 reader) for excitation in the range of 530-560 nm and fluorescence emission detection at approximately 590 nl, or for absorption at approximately 560 nm. Reactions were allowed to run for 30 minutes during these assays.
  • a spectrophotmoter such as a Molecular Devices M5 reader
  • FIGS. 25-29 A series of GRAS compounds and cocktails of GRAS compounds were tested, as shown in FIGS. 25-29 , for the ability to inhibit degradation of a peroxidase-sensitive marker by components in saliva.
  • Various embodiments of the tested compounds are included in Table I.
  • inhibition assays were also conducted with caffeine ( FIG.
  • a test slide with 28 test wells are spotted with 12 to 18 sets of diagnostic markers. These reagents may include diagnostic markers, or controls for the diagnosis.
  • the diagnostic device in this example, has four diagnostic markers spotted in duplicate, along with two positive and two negative control marker. Each marker is spotted with a competitor, such as anti-haptin, which binds the capture antigen particle to the marker.
  • the device may have four different hatpin/anti-haptin systems to serve as the generic capture system.
  • a unique haptin may be conjugated to a fully paramagnetic particle (PmMp).
  • the PmMp is also coated with a capture monoclonal antibody (MAb) specific for the marker.
  • MAb capture monoclonal antibody
  • a second particle (polystyrene microparticle with Europium) is conjugated with the second or detection MAb.
  • the hash marks on the side of the diagnostic device are indentations molded into the plastic, which may be used as cog grooves. Additionally the slide is designed to fit into a processor that may have more than one processing station, as shown in FIG. 32 .
  • the user In performing a method of detecting a diagnostic marker using an embodiment of the diagnostic device, such as that in FIGS. 30 and 32 , the user adds a pre-determined amount of specimen (such as 120 ⁇ l) to a specimen diluent (such as 40 to 120 ⁇ l), and adds the mixture to each test well. Following addition of the diluted specimen, the test slide is incubated at 37° C. for about 5 to about 15 minutes with agitation. Following the incubation period, the test wells move over a magnetic source that brings the PmMp to the bottom of the well, so that there is maximal binding of the PmMp with the anti-haptin bound in the test well. Next, specimen and assay reagents are removed, wash buffer is added and aspirated off. Lastly, each well is scanned using a visualization device, such as a CCD camera, and then the fluroescent signal (or other signal) of each marker is determined.
  • a visualization device such as a CCD camera
  • FIG. 31 An example of a collection device for the collection of an bodily fluids is depicted in FIG. 31 .
  • the collection device is capable of collecting at least 500 ⁇ l of bodily fluid, such as saliva.
  • the cap is first removed from the tube. Squeezing the bulb provides suction adequate to pull an amount of saliva into the bulb.
  • a filter may be included in the tube to remove unwanted materials (such as particulate and mucous).
  • the filter may be of any known filter material, such as rayon, that is appropriate for the purposes described herein.
  • the dispensing tube is in fluid communication with the bulb and is sized and shaped to allow for drops of a predetermined size to be formed when the bulb is squeezed.
  • the dispensing tube may allow drops of 30 ⁇ l to be formed when the bulb is squeezed.
  • the disclosure may have presented a method and/or process as a particular sequence of steps.
  • the method or process should not be limited to the particular sequence of steps described.
  • Other sequences of steps may be possible. Therefore, the particular order of the steps disclosed herein should not be construed as limitations of the present disclosure.
  • disclosure directed to a method and/or process should not be limited to the performance of their steps in the order written. Such sequences may be varied and still remain within the scope of the present disclosure.

Abstract

Devices and systems for biomarker detection. In at least one embodiment of a device for analyzing the presence or absence of a diagnostic marker to be detected in a body fluid, the device comprises a container, wherein the container houses a sample reservoir having a sample input port in fluid connection with at least one detector array, wherein the detector array comprises at least one detector comprising a reaction chamber comprising an immobilized capture molecule, a stabilizing agent contained within the sample reservoir, the stabilizing agent useful to completely or substantially prevent degradation or inactivation of a diagnostic marker, and a reagent contained within the detector array, the reagent capable of undergoing a colorimetric reaction or displaying an optically detectable signal and capable of reacting with the diagnostic marker.

Description

    PRIORITY
  • This U.S. utility patent application claims the priority benefit of U.S. Provisional Patent Application Ser. No. 61/390,002, filed Oct. 5, 2010; U.S. Provisional Patent Application Ser. No. 61/385,347, filed Sep. 22, 2010; U.S. Provisional Patent Application Ser. No. 61/383,401, filed Sep. 16, 2010; U.S. Provisional Patent Application Ser. No. 61/379,598, filed Sep. 2, 2010; U.S. Provisional Patent Application Ser. No. 61/378,960, filed Sep. 1, 2010; U.S. Provisional Patent Application Ser. No. 61/373,619, filed Aug. 13, 2010; U.S. Provisional Patent Application Ser. No. 61/364,964, filed Jul. 16, 2010; U.S. Provisional Patent Application Ser. No. 61/364,969, filed Jul. 16, 2010; U.S. Provisional Patent Application Ser. No. 61/364,975, filed Jul. 16, 2010; U.S. Provisional Patent Application Ser. No. 61/364,978, filed Jul. 16, 2010; U.S. Provisional Patent Application Ser. No. 61/364,982, filed Jul. 16, 2010; U.S. Provisional Patent Application Ser. No. 61/365,179, filed Jul. 16, 2010; U.S. Provisional Patent Application Ser. No. 61/322,768, filed Apr. 9, 2010; and U.S. Provisional Patent Application Ser. No. 61/303,165, filed Feb. 10, 2010. The contents of each of these applications are hereby incorporated by reference in their entirety into this disclosure.
  • BACKGROUND
  • Biomarkers are molecular indicators of a specific biological property, a biochemical feature or facet that can be used to measure the progress of a disease or the effects of a treatment. For example, serum low-density lipoprotein (LDL) is a biomarker of cholesterol and blood pressure, while the P53 gene is a biomarker for cancer. For chronic diseases and conditions, such as diabetes and allergies, accurate diagnosis is particularly important, especially where the side effects of a treatment are severe.
  • Diagnostic tests using biomarkers as molecular indicators not only detect the presence or absence of the biomarker, but often must measure the exact concentration of a biomarker to determine whether an abnormal condition exists. Because of the requirement for accuracy, the process of sample collection, preparation, and analysis are often complicated and time consuming. Currently, blood-based assays for biomarker presence or activity are considered to be the “gold standard” for biomarker-type assays.
  • Despite the desire for accurate results, rapid, point of care analysis of biological samples using biomarkers is becoming increasingly important in the present medical environment due to the need for quick results.
  • SUMMARY
  • The disclosure of the present application provides various devices for analyzing the presence or absence of a diagnostic marker to be detected in a body fluid.
  • In at least one embodiment of a device for analyzing the presence or absence of a diagnostic marker to be detected in a body fluid, the device comprises a container, wherein the container houses a sample reservoir having a sample input port in fluid connection with at least one detector array, wherein the detector array comprises at least one detector comprising a reaction chamber comprising an immobilized capture molecule, a stabilizing agent contained within the sample reservoir, the stabilizing agent useful to completely or substantially prevent degradation or inactivation of a diagnostic marker, and a reagent contained within the detector array, the reagent capable of undergoing a colorimetric reaction or displaying an optically detectable signal and capable of reacting with the diagnostic marker.
  • In at least one embodiment of a device for analyzing the presence or absence of a diagnostic marker to be detected in a body fluid, the device further comprising filter capable of separating at least one unwanted component from a body fluid. Additionally, an embodiment of the device of the present disclosure, may further comprise a results display, wherein a result from the detector array is visually detectable. The results display, of an embodiment of a device of the present disclosure, may comprise a plurality of displays capable to visualizing a plurality of detected diagnostic markers.
  • In at least one embodiment of a device for analyzing the presence or absence of a diagnostic marker to be detected in a body fluid, the device further comprises a control display, wherein the detection of at least one control reagent by the at least one detector is visualized.
  • In at least one embodiment of a device for analyzing the presence or absence of a diagnostic marker to be detected in a body fluid, the stabilizing agent is selected from the group consisting of a protease inhibitor, a DNase inhibitor, and a RNase inhibitor.
  • In at least one embodiment of a device for analyzing the presence or absence of a diagnostic marker to be detected in a body fluid, the stabilizing agent is useful to completely or substantially prevent degradation or inactivation of a diagnostic marker selected from the group consisting of a protein, a glycoprotein, a nucleic acid, an enzyme, an enzyme inhibitor, and a metabolite.
  • In at least one embodiment of a device for analyzing the presence or absence of a diagnostic marker to be detected in a body fluid, the stabilizing agent is useful to completely or substantially prevent degradation or inactivation of a diagnostic marker indicative of a disease.
  • In at least one embodiment of a device for analyzing the presence or absence of a diagnostic marker to be detected in a body fluid, the disease state is selected from the group consisting of a cancer, a metabolic disease, a cardiac disease, diabetes, a bacterial infection, a viral infection, a fungal infection, and a parasitic infection.
  • In at least one embodiment of a device for analyzing the presence or absence of a diagnostic marker to be detected in a body fluid, the stabilizing agent is selected from the group consisting of Fixanal® Buffer 6.0, acetic acid, aluminum hydroxide bentonite, aluminum sulfate hydrate, benzoic acid, aluminum potassium sulfate dodecahydrate, caffeine, and 3-tert-butyl-hydroxyanisole, or a combination thereof.
  • 1. In at least one embodiment of a device for analyzing the presence or absence of a diagnostic marker to be detected in a body fluid, the device comprises a sample reservoir having a sample input port in fluid connection with at least one detector array, wherein said detector array comprises at least one detector comprising a reaction chamber having an immobilized capture molecule, a stabilizing agent useful to completely or substantially prevent degradation or inactivation of a diagnostic marker, a reagent capable of undergoing a colorimetric reaction or displaying an optically detectable signal and capable of reacting with the diagnostic marker, a filter capable of separating at least one unwanted component from the body fluid, and a results display, wherein a result from the detector array is visualized.
  • DESCRIPTION OF THE DRAWINGS
  • The features and advantages of the present disclosure, and the manner of attaining them, will be more apparent and better understood by reference to the following descriptions taken in conjunction with the accompanying figures, wherein:
  • FIGS. 1A and B show a diagram of a detection system, according to at least one embodiment of the present disclosure;
  • FIGS. 2-4 shows a diagram of a method of producing a bead system, according to at least one embodiment of the present disclosure;
  • FIG. 5 shows a flowchart of a method for creating a bead system, according to at least one embodiment of the present disclosure;
  • FIGS. 6A and B show a diagram of a test strip, according to at least one embodiment of the present disclosure;
  • FIG. 6C show a diagram of a microarray, according to at least one embodiment of the present disclosure;
  • FIG. 6D shows a diagram of a microfluidic system, according to at least one embodiment of the present disclosure;
  • FIG. 7 shows a flowchart of a method of stabilizing diagnostic markers, according to at least one embodiment of the present disclosure;
  • FIG. 8 shows a flowchart of a method analyzing a diagnostic markers, according to at least one embodiment of the present disclosure;
  • FIG. 9 shows a an exemplary system framework, according to at least one embodiment of the present disclosure;
  • FIG. 10 shows a graphical depiction of α-amylase inhibition according to at least one embodiment of the present disclosure;
  • FIG. 11 shows a graphical depiction of α-amylase inhibition according to at least one embodiment of the present disclosure;
  • FIG. 12 shows a graphical depiction of lysozyme inhibition according to at least one embodiment of the present disclosure;
  • FIG. 13 shows a graphical depiction of a galactose oxidase screen according to at least one embodiment of the present disclosure;
  • FIG. 14A shows a three dimensional plot of glycated Hemoglobin A1c (HbA1c) analyzed by high pressure liquid chromatography (HPLC) according to at least one embodiment of the present disclosure;
  • FIG. 14B shows a ultraviolet (UV) image of HbA1c analyzed by HPLC according to at least one embodiment of the present disclosure;
  • FIG. 14C shows a chromatogram of HbA1c analyzed by HPLC according to at least one embodiment of the present disclosure;
  • FIG. 15A shows a three dimensional plot of HbA1c analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure;
  • FIG. 15B shows a UV image of HbA1c analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure;
  • FIG. 15C shows a chromatogram of HbA1c analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure;
  • FIG. 16A shows a three dimensional plot of HbA1c analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure;
  • FIG. 16B shows a UV image of HbA1c analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure;
  • FIG. 16C shows a chromatogram of HbA1c analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure;
  • FIG. 17 shows a comparison of chromatograms from HPLC analyzed HbA1c (FIG. 17A), HbA1c+Saliva (FIG. 17B), and HbA1c+Saliva+Stabilizing mixture (FIG. 17C) according to at least one embodiment of the present disclosure;
  • FIG. 18 shows a comparison of UV images from HPLC analyzed HbA1c (FIG. 18A), HbA1c+Saliva (FIG. 18B), and HbA1c+Saliva+Stabilizing mixture (FIG. 18C) according to at least one embodiment of the present disclosure;
  • FIG. 19 shows a comparison of three dimensional plots from HPLC analyzed HbA1c (FIG. 19A), HbA1c+Saliva (FIG. 19B), and HbA1c+Saliva+Stabilizing mixture (FIG. 19C) according to at least one embodiment of the present disclosure;
  • FIG. 20A shows a three dimensional plot of Cancer Antigen 19-9 (CA 19-9) analyzed by high pressure liquid chromatography (HPLC) according to at least one embodiment of the present disclosure;
  • FIG. 20B shows a ultraviolet (UV) image of CA 19-9 analyzed by HPLC according to at least one embodiment of the present disclosure;
  • FIG. 21A shows a three dimensional plot of CA 19-9 analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure;
  • FIG. 21B shows a UV image of CA 19-9 analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure;
  • FIG. 22A shows a three dimensional plot of CA 19-9 analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure;
  • FIG. 22B shows a UV image of CA 19-9 analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure;
  • FIG. 23 shows a comparison of three dimensional plots from HPLC analyzed CA 19-9 (FIG. 23A), CA 19-9+Saliva (FIG. 23B), and CA 19-9+Saliva+Stabilizing mixture (FIG. 23C) according to at least one embodiment of the present disclosure;
  • FIG. 24 shows a comparison of UV images from HPLC analyzed CA 19-9 (FIG. 24A), CA 19-9+Saliva (FIG. 24B), and CA 19-9+Saliva+Stabilizing mixture (FIG. 24C) according to at least one embodiment of the present disclosure;
  • FIG. 25 shows a graphical depiction of Peroxidase activity inhibition by an inhibitor cocktail according to at least one embodiment of the present disclosure;
  • FIG. 26 shows a graphical depiction of Peroxidase activity inhibition by caffeine according to at least one embodiment of the present disclosure;
  • FIG. 27 shows a graphical depiction of Peroxidase activity inhibition by 3-tert-butyl-hydroxyanisole according to at least one embodiment of the present disclosure;
  • FIG. 28 shows a graphical depiction of Peroxidase activity inhibition by benzoic acid according to at least one embodiment of the present disclosure;
  • FIG. 29 shows a graphical depiction of Peroxidase activity inhibition by aluminum potassium sulfate dodecahydrate according to at least one embodiment of the present disclosure;
  • FIG. 30 shows a schematic of a diagnostic system, according to at least one embodiment of the present disclosure;
  • FIG. 31 shows a graphical representation of an assay component, according to at least one embodiment of the present disclosure; and
  • FIG. 32 shows a graphical depiction of a fluid collection device, according to at least one embodiment of the present disclosure.
  • DETAILED DESCRIPTION
  • For the purposes of promoting an understanding of the principles of the present disclosure, reference will now be made to the embodiments illustrated in the drawings, and specific language will be used to describe the same. It will nevertheless be understood that no limitation of the scope of this disclosure is thereby intended.
  • The disclosure of the present application provides various compositions, systems, and methods for biomarker stabilization and analysis. Specifically, formulations are disclosed herein, which function to preserve the level of diagnostic markers, such as biomarkers, in body fluids, from enzymatic alteration, or degradation. Additionally, methods for the collection and analysis of body fluids are disclosed. The methods and formulations disclosed herein can be used to improve the sampling and testing of a body fluid by conditioning the body fluids for the stabilization of specific biomolecules and/or drug metabolites. As used herein, the term “body fluids” includes fluids produced by the body, such as saliva, or fractions thereof, mucous secretions, tears, sweat, bile, semen, urine, vaginal secretions, exhalations, anal secretions, blood, plasma, serum and mixtures of thereof. Body fluids may also comprise cancer cells, peripheral blood mononuclear cells, lymphocytes, lymph fluid, and other tissue secretions or fluid.
  • Saliva
  • Saliva is clear, viscous fluid with a slightly alkaline pH and a pI range from 11.5-3.0. It is hypotonic, composed of about 99.5% water, and also contains ions (e.g., K+, Na+, Ca2+, Mg2+, H+, Cl, HCO3 , I, F, HPO4 2−), and small organic molecules (e.g., ureas, hormones, lipids, DNA, and RNA). There are multiple contributors to the composition of saliva. Saliva has a complex “proteome”—106 D glycoproteins to 1000D peptides. It contains secretory products of salivary glands, products of B cells, PMNs, epithelial cells, and bacteria. Major (e.g., parotid, submandibular, and sublingual) and minor (e.g., palatine and retromolar) glands contribute to the composition of saliva, along with extraneous contributors such as gingival crevicular fluid, serum proteins, white blood cells and their byproducts, oral epithelial cells, oral bacteria, food debris and dissolved food components.
  • Saliva from different glands may differ in composition. For example, saliva from the parotid gland is dominated by serous secretory cells, whereas saliva from the submandibular and sublingual glands and minor glands are mixed serous or mostly mucous. There can also be qualitative and quantitative differences in saliva output that affect its composition. Glandular contribution to saliva is affected by level of gland activity. The amount of saliva secreted per minute or the salivary flow rate influences the concentration of the constituents as well as the proportion of the constituents from each of the three pairs of major salivary glands and the minor salivary glands.
  • I. Stabilizing Agents
  • According to at least one embodiment of the present disclosure, conditioning of body fluids for the preservation of diagnostic markers may be accomplished through the use of stabilizing agents, which may be incorporated into one or more carrier vehicles, such as rinses, gums, beverages, and confectionaries. For example, specific rinses and/or pre-rinses specially formulated according to the specific molecule or molecules (the diagnostic marker) to be detected in the body fluid, may contain a stabilization agent. The use of rinses and pre-rinses of the present disclosure to condition the body fluid may enhance the sensitivity for detection of specific diagnostic markers for clinical diagnosis. Additionally, this process may improve the signal-to-noise ratio for a better diagnostic yield. Further, each rinse/pre-rinse can be specifically formulated to reduce or prevent false positive and false negative results.
  • In at least one embodiment of a stabilizing agent of the present disclosure, the stabilizing agent may act to prevent/decrease the degradation, or reduction of activity, of diagnostic markers. In at least one embodiment, the stabilizing agent may be comprised of one or more Generally Regarded as Safe (GRAS) compound, as determined by the Food and Drug Administration of the United States of America, such as those listed in Table I. An exemplary embodiment of a stabilization agent may act to stabilize a diagnostic marker, and/or to bind to a destructive component to prevent the destructive component from degrading or decreasing the activity of the diagnostic marker.
  • In additional exemplary embodiments of the stabilization agent, the stabilizing agent may comprise at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, or at least 25 compounds which acts to stabilize the diagnostic marker, and/or to bind to a destructive component to prevent the destructive component from degrading or decreasing the activity of the diagnostic marker. Further, in various embodiments the stabilizing agent of the present disclosure may be present in at a level of between about 200ppm to about 2,000 ppm, about 400ppm to about 1600 ppm, about 600ppm to about 1400 ppm, about 800 ppm to about 1200 ppm. Moreover, in at least one embodiment, the stabilizing agent may be present at a level of approximately 500 ppm.
  • TABLE I
    GRAS Compounds as Stabilizing Agents
    1 Agar Sigma-Aldrich
    2 Alginic acid sodium salt Sigma-Aldrich
    3 L-Ascorbic acid Sigma-Aldrich
    4 (+)-Sodium ascorbate Sigma-Aldrich
    5 Buffer solution Fluka
    6 Sodium citrate Sigma-Aldrich
    7 Tragacanth Sigma-Aldrich
    8 Gum arabic, from acacia tree Sigma-Aldrich
    9 Sodium tripolyphosphate Sigma-Aldrich
    10 Potassium citrate monobasic Sigma-Aldrich
    11 Sodium pyrophosphate tetrabasic Sigma-Aldrich
    12 Calcium citrate Sigma-Aldrich
    13 Citric acid Sigma-Aldrich
    14 Potassium sodium tartrate tetrahydrate Sigma-Aldrich
    15 Sodium benzoate Sigma-Aldrich
    16 Benzoic acid Sigma-Aldrich
    17 D-aspartic acid Sigma-Aldrich
    18 Sodium phosphate dibasic Sigma-Aldrich
    19 Malic acid Sigma-Aldrich
    20 Sodium propionate Sigma-Aldrich
    21 Sodium L-ascorbate Sigma-Aldrich
    22 DL-Phenylalanine Sigma-Aldrich
    23 Calcium L-ascorbate dihydrate Sigma-Aldrich
    24 Choline chloride Sigma-Aldrich
    25 L-Ascorbic acid Sigma-Aldrich
    26 Sodium diacetate SAFC
    27 PH buffer 4.01 Orion
    28 PH Buffer 7.00 Orion
    29 PH buffer 10.0 Orion
    30 Fixanal Buffer 6.0 Sigma-Aldrich
    31 Ammonium Chloride Sigma-Aldrich
    32 Ammonium Citrate tribasic, anhydrous Sigma-Aldrich
    33 Caffeine Sigma-Aldrich
    34 Aluminum sulfate hydrate Sigma-Aldrich
    35 Beeswax refined Sigma-Aldrich
    36 Aluminum Hydroxide Sigma-Aldrich
    37 Bentonite Sigma-Aldrich
    38 Ammonium bicarbonate Sigma-Aldrich
    39 3-tert--butyl-hydroxyanisole Sigma-Aldrich
    40 Benzoic acid Sigma-Aldrich
    41 trans-aconitic acid Sigma-Aldrich
    42 Aluminum potassium sulfate Sigma-Aldrich
    dodecahydrate
    43 Adpic acid Sigma-Aldrich
    44 Ammonium hydrogen phosphate Sigma-Aldrich
    45 Ammonium dihydrogen phosphate Sigma-Aldrich
    46 Ammonium carbonate Sigma-Aldrich
    47 Acetic acid Sigma-Aldrich
  • According to at least one exemplary embodiment, the stabilizing agent may block at least one enzymatic activity of a destructive component. The destructive component may be one or more component of a bodily fluid which acts to degrade, or decrease the activity of a diagnostic marker. Further, the destructive component may be one or more Amylases, Lysozymes, Peroxidases, Glycosidases, Esterases, Proteases, and/or Peptidases. In an exemplary embodiment, the stabilizing agent may be a naturally occurring or artificial protease inhibitor, DNase inhibitor, or RNase inhibitor. Specifically, a natural stabilizing agent may include any component found from food products (including, but not limited to, the Solanaceae family), herbs, or spices that acts to decrease the effect of a destructive component. For example, exemplary embodiments of the natural stabilizing agents may be any of the inhibitors found in lima beans, soybeans, or avian eggs (such as ovomucoid glycoprotein protease inhibitors) (See Table II). Moreover, an embodiment of the stabilizing agent may also comprise a molecule which specifically inhibits the activity of a molecule which targets the diagnostic marker for degradation or inactivation. Further, the stabilizing agent may act to alter the composition of the body fluid (such as pH, or ion concentration) to decrease the degradation or inactivation of the diagnostic marker.
  • TABLE II
    Molecular Inhibitory
    Material weight power Details
    Lima beans 8-10 kDa 2.2 times There are six different lima bean inhibitors.
    weight
    Ovomucoid 8-10 kDa 1.2 times Ovomucoids are the glycoprotein protease inhibitors
    weight found in raw avian egg white. There are other protease
    inhibitors in ovomucoids as well.
    Soybeans 20.7-22.3 kDa 1.2 times Soybeans contain several inhibitors; the one in the chart is
    weight considered the primary one. All of them bind
    chymotrypsin to a lesser degree.
  • As used herein, “diagnostic marker” may be any biological molecule whose presence, specific concentration, or specific activity may be indicative of a disease state or heath/lifestyle characteristic. As used herein, “indicative of a disease state” refers to the presence, progression, prevention, remission, amelioration, or prognosis of a disease. This also includes, but is not limited to, the effects of a therapeutic on a disease. The disease state analyzed through use of the diagnostic marker may in an exemplary embodiment be directed towards any cancer, metabolic disease (such as diabetes I or II), cardiovascular disease, or the predisposition of any of the same. Additionally, a disease state may also include the infection of the patient by a bacterial, virus, yeast, fungus, or parasite.
  • A. Disease States
  • Bacterial infections, as referenced herein, may include a mammalian infection by any bacterial species. Embodiments of the bacterial species, as used herein, may include, but are not limited to, Bacillus anthracis, Bacillus cereus, Staphylococcus aureus, Listeria monocytogenes, Streptococcus pneumoniae, Streptococcus pyogenes, Clostridium botulinum, Clostridium difficile, Clostridium perfringens, Clostridium tetani, Borrelia burgdorferi, Treponema pallidum, Chlamydia trachomatis, Chlamydophila psittaci, Corynebacterium diphtherias, Mycobacterium tuberculosis, and Mycobacterium avium, Rickettsia prowazekii, Rickettsia rickettsii, Rickettsia typhi, Anaplasma phagocytophilum, Ehrlichia chaffeensis, Brucella melitensis, Bordetella pertussis, Burkholderia mallei, B. pseudomallei, Neisseria gonorrhoeae, Neisseria meningitides, Campylobacter jejuni, Helicobacter pylori, Legionella pneumophila, Acinetobacter baumannii, Moraxella catarrhalis, Pseudomonas aeruginosa, Aeromonas sp., Vibrio cholerae, Vibrio parahaemolyticus, Thiotrichales sp., Haemophilus influenzae, Klebsiella pneumoniae, Proteus mirabilis, Yersinia pestis, Yersinia enterocolitica, Shigella flexneri, Salmonella enterica, and Escherichia coli.
  • Viral infections, as referenced herein, may include a mammalian infection by any viral species. Embodiments of the viral species, as used herein, may include, but are not limited to, single-stranded DNA viruses, including the genus Parvoviridae, double-stranded DNA viruses, including the genus Papillomaviridae, Polyomaviridae, Poxviridae, Herpesviridae (ex. Herpes Simplex Virus 1, 2, 6, Varicella-zoster virus, Epstein-Barr virus EBV, Human cytomegalovirus, and human herpesvirus 7), single-stranded RNA viruses, Reo- and Retroviruses (ex. Hepatitis B virus, Human immunodeficiency virus 1 and 2, and Human T-lymphotropic virus 1).
  • Fungal infections, as referenced herein, may include a mammalian infection by any bacterial species. Embodiments of the fungal species, as used herein, may include, but are not limited to, Fusarium oxysporum, Pneumocystis jirovecii, Aspergillus spp., Coccidioides immitis/posadasii, Candida albicans, Filobasidiella neoformans, Trichosporon, Encephalitozoon cuniculi, Enterocytozoon bieneusi, Mucor circinelloides, Rhizopus oryzae, and Lichtheimia corymbifera.
  • Parasite infections, as referenced herein, may include a mammalian infection by any parasite. Embodiments of the parasite may be, but are not limited to, Entamoeba histolytica, Babesia microti, Babesia sp. WA1, Trypanosoma cruzi, Taenia solium, Echinococcus granulosus, Leishmania braziliensis, L. donovani, L. tropica, Plasmodium falciparum, Plasmodium vivax, Plasmodium ovale and Plasmodium malariae, Plasmodium knowlesi, Paragonimus westermani, chistosoma sp., S. mansoni, S. haematobium, S. japonicum, Strongyloides stercoralis, Toxocara canis, Toxoplasma gondii, Trichinella spiralis African trypanosomiasis, Ancylostoma, Angiostrongylus, Anisakis, Baylisascaris procyonis, Clonorchis (Opisthorchis) sinensis, Echinococcus multilocularis, Fasciola hepatica, Filariasis, Gnathostoma
  • Exemplary embodiments of diagnostic markers include those listed in Table III. Further exemplary embodiments for the diagnosis of type-II diabetes include, but are not limited to, Albumin, Alpha-1 antitrypsin (A1AT)—G Protein, Cystatin C, Cystatin A, alpha-2-macroglobulin (A2MG), Uteroglobin, Transthyretin (TTR), Annexin A1, Annexin A2, Annexin A3 and Calnexin.
  • Diagnostic markers directed towards cardiovascular disease, in an exemplary embodiment, may include, but are not limited to, any isoform of creatine kinase, troponin I and T, LD, Myoglobin, ALT/AST, H-FABP, and Glycogen phosphorylase B.
  • Diagnostic markers directed towards cancer, in addition to those described in Table III, may include, but are not limited to, Aldose reductase, Angiogenin, Annexin A1, B-cell activating factor (BAFF), B-cell lymphoma 2 (BCL2)-like 2, Beta Human chorionic gonadotropin, Ca15-3, Calcyclin, Calvasculin, Cathepsin D, Caveolin-1, Chromogranin A, Alpha-crystallin B chain (CRYAB), Endostatin, Eotaxin-2, Epithelial cell adhesion molecule (EpCAM), Ezrin, fatty acid binding protein 4 (FABP4), Galectin-3, γ-glutamylcysteine ligase regulatory chain (GCLR), Gelsolin, Glucose 6-phosphate (G6P), Glycoprotein 130 (gp130), Glutathione S-transferase Mu 1 (GSTM1), Hepsin, High-mobility group protein B1 (HMGB-1), Insulin-like growth factor binding protein 1 (IGFBP-1), Insulin-like growth factor binding protein 4 (IGFBP-4), Insulin-like growth factor binding protein 5 (IGFBP-5), Insulin-like growth factor binding protein 6 (IGFBP-6), LGL, latency associated peptide (LAP), macrophage stimulating protein (MSP), MHC class I polypeptide-related sequence A (MICA), Nucleoside diphosphate kinase B (NME2), Neuron-specific Enolase (NSE), Osteopontin, Osteoprotegerin, Pepsinogen, Peroxiredoxin, Phosphoserine aminotransferase (PSAT1), Prostate Specific Antigen, Receptor tyrosine-protein kinase erbB-3 (ErbB3), Serpin B3, Vascular smooth muscle cell growth factor R2 (VSGF R2/KDR), Vascular endothelial growth factor R3 (VEGF R3/Flt-4), Thyroglobulin, Tyrosine kinase with immunoglobulin-like and EGF-like domains 2 (TIE-2), Tissue plasminogen activator (tPA), Transforming growth factor beta (TGF-β1), Tumor necrosis factor receptor 1 (TNF-R1), urokinase-type Plasminogen Activator (uPA), urokinase-type Plasminogen Activator Receptor (uPAR), Brcal, BrcaII, kallikreins, e-cadherin, Hox peptide, and Engrailed-2.
  • Diagnostic markers directed towards a bacterial or virus may include any surface or secreted antigen from the bacteria or virus, or a conserved nucleotide sequence.
  • TABLE III
    Present in Present
    Relevant Serum/ in Class of
    Tumor Marker Cancer Plasma Saliva Molecules
    α-fetoprotein Heptacellular Yes Yes Protein
    carcinoma
    Cancer Antigen Breast cancer Yes Yes Protein
    (CA15-3)
    Cancer Antigen Ovarian cancer Yes Yes Protein
    (CA 125)
    Carcinoembryonic Many epithelial Yes Yes
    antigen cancers
    Prostate specific Prostate cancer Yes Yes Protein
    antigen
    c-erb-2 Breast cancer Yes Nucleic acid
    P16, p53 Oral SCC
    BRACA1, Breast cancer Yes Yes Nucleic acid
    BRACA2
    Hemoglobin-A1c Yes Protein
  • B. Diagnostic Markers
  • Further, diagnostic markers according to an embodiment of the present disclosure may include one or more of: 1,25 dihydroxy-vitamin D, 17-Hydroxyprogesterone, 25-hydroxy-vitamin D, Antineutrophil Cytoplasmic Antibodies, 5-Hydroxy Tryptamine, 5-hydroxyindoleacetic acid, Acetoacetate, Activated Partial Thromboplastin Time, Adrenocorticotropic Hormone, Alanine aminotransferase, Alanine transaminase, Albumin, Albumin-to-Creatinine ratio, Albumin/Globulin ratio, Alcohol, Aldolase, Aldosterone, Aldosterone and plasma renin activity, Aldosterone and Renin, Alkaline Phosphatase, Allergen-specific IgE, Alpha tryptase, Alpha-1 Antitrypsin, Alpha-fetoprotein, Alpha1-antitrypsin, Alzheimer biomarkers (including Tau protein and Amyloid Beta 42 peptides), Amylase, ANCA Antibodies, ANCA/MPO/PR3 Antibodies, Angiotensin-Converting Enzyme, Anti-citrulline antibody, anti-cyclic citrullinated peptide antibody, Anti-retroviral drug resistance testing, anti-ribonucleoprotein, anti-Sjögren's Syndrome A, anti-Sjögren's Syndrome B, Anti-Smooth Muscle Antibody, anti-topoisomerase, Anticardiolipin Antibodies, Antidiuretic Hormone, Antifactor Xa heparin, Antiglobulin, Antihistidyl Transfer RNA, Synthase Antibodies, Antimicrosomal antibody, Antimitochondrial Antibody and Antimitochondrial M2 Antibody, Antineutrophil Cytoplasmic Antibodies, Antinuclear Antibody test, Antiphospholipid antibodies, Antiphospholipids, Antistreptolysin O titer, Antithrombin, Antithyroglobulin antibody, Antithyroid antibodies, Apolipoprotein A-I, Apolipoprotein B-100, Arginine Vasopressin, Aspartate aminotransferase, Aspartate transaminase, B-type natriuretic peptide, Beta hCG, Beta tryptase, Beta-2 Microglobulin, Beta-hydroxybutyrate, Beta-hydroxybutyric acid, Beta2 Microglobulin, Bicarbonate, Bilirubin, cholesterol, Blood clotting factors, Bordetella pertussis Antibodies, Borrelia burgdorferi antibodies, IgM/IgG, Brain natriuretic peptide, Breast Cancer Gene 1 and Breast Cancer Gene 2, c-ANCA, c-erbB-2, C-peptide, C-Reactive Protein, Caffeine, Calcidiol, Calcifidiol, Calcitonin, Calcitriol, Calcium, Calcofluor white stain, Cancer Antigen 125, Cancer Antigen 15-3, Cancer Antigen 19-9, Cancer antigen-breast, Cancer antigen-GI, Carbamazepine, Carcinoembryonic Antigen, Cardiac-specific Troponin I and Troponin T, Cardiolipin Antibodies, Catecholamines, Celiac Disease Tests, Ceruloplasmin, Chickenpox, Chickenpox and Shingles Tests, Chlamydia, Chloride, Cholesterol, Chromogranin A, Chymotrypsin, Citrulline antibody, Coagulation Factors, Cobalamin, Complement Component C3, Complement Component C4, Complexed PSA, Conjugated bilirubin, Copper, Corticotropin, Cortisol, Cotinine, Creatine Kinase, Creatine Kinase-MB, Creatinine, Cryoglobulin, Cryoprotein, Cyclic Citrullinated Peptide Antibody, Cyclosporine, Cystatin C, Cystic Fibrosis Gene Mutation Panel, Cystic fibrosis genotyping, Cytomegalovirus, Cytotoxic T-cells, Dehydroepiandrosterone Sulfate, Delta-aminolevulinic acid, Depakene, Depakote, Des-gamma-carboxy prothrombin, DHEA Sulfate, Diabetes mellitus autoantibody panel, Digoxin, Dilantin, Direct Anti-human Globulin test, Direct Antiglobulin Test, Direct bilirubin, Direct Low-density lipoprotein cholesterol, Dopamine, Drug screen, eGFR, Epidermal Growth Factor Receptor, Epinephrine, Epstein-Barr Virus Antibodies, Erythropoietin, Estradiol, Estriol, Estrogens, Estrone, Ethanol, F-Actin Antibody, Factor I, Factor V Leiden, Factor V Leiden Mutation and PT 20210 Mutation, Factor V Leiden mutation: Activated protein C resistance, Factor V R506Q, Ferritin, Fetal fibronectin, Fibrin degradation fragment, Fibrinogen, Fluorescent Antinuclear Antibody, Fluorescent treponemal antibody absorption, Folic Acid, Follicle-stimulating hormone, Fragment D-dimer, Fructosamine, Gamma-glutamyl transferase, Gamma-glutamyl transpeptidase, Gastrin, Genital Human Papillomavirus, Glucose-6-Phosphate Dehydrogenase, Glutamic Acid Decarboxylase Autoantibodies, Gluten-Sensitive Enteropathy Tests, Glycated Albumin, Glycated hemoglobin, Heavy Metals (such as lead, mercury, iron, copper, and zinc), Hemoglobin, Hemoglobin-binding Protein, Hemogram, Heparin Anti-Xa, Hepatitis A, Hepatitis B, Hepatitis C, Herpes Simplex Virus, Type 1 and Type 2, Herpes Zoster, Heterophile Antibodies, High-density lipoprotein cholesterol, High-sensitivity C-reactive protein, Homocysteine, Human calcitonin, Human chorionic gonadotropin, Human epidermal growth factor receptor, Human Growth Hormone, Human immunodeficiency virus antibody test, Human Immunodeficiency Virus Genotypic Resistance Testing, Human Leukocyte Antigen, Immunoreactive trypsinogen, Insulin Autoantibodies, Insulin C-peptide, Insulin-like Growth Factor-1, Insulinoma-Associated-2 Autoantibodies, Intact PTH, Interstitial Cell Stimulating Hormone, Iron, Ischemia-Modified Albumin, Islet autoantibodies Islet Cell Cytoplasmic Autoantibodies, Janus Kinase 2, Ketone bodies Ketones, blood, Lactate, Lactate dehydrogenase, Lactic Acid, Lactic dehydrogenase, Lead, Lipoprotein, Lithium, Low-density lipoprotein cholesterol, Lupus Antibody, Luteinizing hormone, Lyme antibodies IgM/IgG by Western blot, Magnesium, Mast cell tryptase, Measles or Mumps IgM and IgG Antibodies, Mercury, Metanephrine and Normetanephrine, Methotrexate, Methylmalonic Acid, Microalbumin, Mitochondrial Antibody, Mononuclear heterophile test, Mycophenolic acid, Mycoplasma, Mycoplasma pneumoniae IgG and IgM antibodies, Myeloperoxidase Antibodies, Myoglobin, N-terminal pro b-type natriuretic peptide, Neonatal bilirubin, Nicotine, Norepinephrine, p24 antigen, Parathyroid Hormone, Parvovirus, Pertussis, Phenobarbital, Phenytoin, Phosphate, Phosphorus, Platelet-activating factor acetylhydrolase, porphobilinogen, Potassium, Prealbumin, Presenilin 1 gene, Procalcitonin, Progesterone, Proinsulin C-peptide, Prolactin, Prostate Specific Antigen, Protein C, Protein C and Protein S, Protein Tyrosine Phosphatase-like Autoantibodies, Proteinase 3 Antibodies, Prothrombin 20210 mutation, Prothrombin 20210 mutation: PT G20210A, factor II 20210, Red Blood Cell Antibody Identification, Renin, Respiratory Syncytial Virus, Rheumatoid Factor, Ristocetin Cofactor, Rubella, Scleroderma antibodies, Serine Protease 3, Serotonin, Siderophilin, Sirolimus, Smith antibody, Smooth Muscle Antibody, Sodium, Soluble Mesothelin-Related Peptides, Somatomedin C, Somatotropin, Syphilis, Tacrolimus, Tegretol®, Testosterone, Testosterone-estrogen Binding Globulin, Theophylline, Theophylline and Caffeine, Thiopurine methyltransferase, Thiopurine S-methyltransferase, Thymotaxin, Thyrocalcitonin, Thyroglobulin, Thyroid peroxidase antibody, Thyroid stimulating hormone receptor antibody, Thyroid stimulating immunoglobulin, Thyroid-stimulating hormone, Thyroperoxidase antibody, Thyrotropin, Thyroxine, Thyroxine-binding prealbumin, Transferrin, Transthyretin test, Treponema pallidum particle agglutination assay, Trichomonas, Trichomoniasis, Triglycerides, Triiodothyronine, Troponins, Trypsin, Trypsin and Chymotrypsin, Trypsinogen, Tryptase, Valproic acid, Vancomycin, Vanillylmandelic acid, Zoster Virus, Vasopressin, Very Low-Density Lipoprotein Cholesterol, Viral Hepatitis A Antibody, Viral Hepatitis C. Viral Load (HIV), Vitamin B12, Vitamin B12 & Folate, Vitamin D, Vitamin D2, Vitamin D3, Vitamin K, von Willebrand Factor, West Nile Virus, Westergren sedimentation rate, and Zinc Protoporphyrin.
  • C. Exemplary Targets for Stabilizing Agent(s)
  • 1. Amylase
  • Amylase is an enzyme that breaks down starch into sugar, and is found in such places as human saliva, where it begins the chemical process of digestion. The α-amylases are calciummetalloenzymes that are completely unable to function in the absence of calcium. Through acting at random locations on the starch chain, α-amylase breaks down carbohydrates into maltotriose and maltose from amylase. In animals, α-amylase is a major digestive enzyme that has an optimum pH of 6.7 to 7.0.
  • In at least one embodiment of a stabilizing agent of the present disclosure, the stabilizing agent may be an inhibitor of α-amylase. Further, the stabilizing agent may be active in a biological fluid, such as in saliva. In an exemplary embodiment, a stabilizing agent may comprise one or more of Fixanal® Buffer 6.0 (Sigma-Aldrich Co.), aluminum sulfate hydrate, aluminum hydroxide, bentonite, and aluminum potassium sulfate dodecahydrate.
  • 2. Lysozyme
  • Lysozyme, also known as muramidase or N-acetylmuramide glycanhydrolase, is a glycoside hydrolase, that damage bacterial cell walls by catalyzing hydrolysis of 1,4-beta-linkages between N-acetylmuramic acid and N-acetyl-D-glucosamine residues in a peptidoglycan and between N-acetyl-D-glucosamine residues in chitodextrins. Lysozyme is abundant in a number of secretions, such as tears, saliva, human milk, and mucus. It is also present in cytoplasmic granules of the polymorphonuclear neutrophils (PMN).
  • In at least one embodiment, a stabilizing agent of the present disclosure may be an inhibitor of lysozyme. Further, the stabilizing agent may be active in a biological fluid, such as saliva. In an exemplary embodiment, a stabilizing agent may comprise one or more of Fixanal® Buffer 6.0 (Sigma-Aldrich Co.), bentonite, benzoic acid, and acetic acid.
  • 3. Peroxidase
  • Peroxidase are a class of oxidoreductase enzymes that catalyze the oxidation of a compound by the decomposition of hydrogen peroxide or an organic peroxide. These peroxidases may be found in many different bodily fluids. For example, saliva contains both salivary peroxidase (SPX) and myeloperoxidase (MPO). These peroxides may act on a number of different diagnostic markers found in bodily fluids to mask the detection or analysis of the markers.
  • In at least one embodiment of a stabilizing agent of the present disclosure, the stabilizing agent may be an inhibitor peroxidase activity. Further, the stabilizing agent may be active in a biological fluid, such as saliva, blood, serum, or cancer cells. In an exemplary embodiment, a stabilizing agent may comprise one or more of aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate.
  • 4. Insulin Resistance
  • In at least one embodiment of a stabilizing agent of the present disclosure, the stabilizing agent may preserve a diagnostic marker for insulin resistance and/or glucose intolerance. Further, the stabilizing agent may be active in a biological fluid, such as saliva. In an exemplary embodiment, a diagnostic marker may be one or more of α-hydroxybutyrate, 1-linoleoyl-GPC, palmitate, Glycine, 3-methyl-2-oxybutyrate. Moreover, a diagnostic marker may be any marker of early stage insulin resistance and glucose intolerance in a non-diabetic individual.
  • 5. Galactose Oxidase
  • Galactose oxidase is a member of the family of oxidoreductaases, and has been shown to participate in galactose metabolism. Because of this activity, galactose oxidase may be used to detect mucin-like glycoproteins. These glycoproteins are a major component of mucus secreted by epithelial and glandular cells and are primarily responsible for the protective properties of the viscoeleastic mucous barrier. Mucins have been implicated in the process of cholesterol gallstone formation, and has been identified as having abnormal expression in some cancers.
  • In at least one embodiment of a stabilizing agent of the present disclosure, the stabilizing agent may be an inhibitor of galactose oxidase. Further, the stabilizing agent may be active in a biological fluid, such as saliva. In an exemplary embodiment, a stabilizing agent may comprise one or more of aluminum potassium sulfate dodecahydrate, 3-tert-butyl-hydroxyanisole, benzoic acid, and acetic acid.
  • 6. Glycated Hemoglobin
  • Glycated hemoglobin (HbA1c) is a form of hemoglobin that is indicative of the average plasma glucose concentration over a period of time. Due to the transport of components of the plasma into bodily fluids, such as saliva, the level of HbA1c may be determined in bodily fluids in addition to plasma.
  • In at least one embodiment of a stabilizing agent of the present disclosure, the stabilizing agent may be an inhibitor of HbA1c degradation. Further, the stabilizing agent may be active in a biological fluid, such as saliva. In an exemplary embodiment, a stabilizing agent may comprise one or more of acetic acid, aluminum hydroxide bentonite, aluminum sulfate hydrate, aluminum potassium sulfate dodecahydrate, benzoic acid, caffeine, and 3-tert-butyl-hydroxyanisole, or a combination thereof.
  • 7. Cancer Antigen 19-9 (CA 19-9)
  • Cancer Antigen 19-9 (CA 19-9), which may in some instances be referred to as Cancer Angigen 19.9, Cancer antigen-GI or CA-GI, is an antigen associated with various cancers, such as prostate and colon cancer. At least one use of CA 19-9 may be see whether a pancreatic tumor is secreting antigen. If that is the case, then the levels may decrease when the tumor is treated, and they may rise again if the disease recurs.
  • In at least one embodiment of a stabilizing agent of the present disclosure, the stabilizing agent may be an inhibitor of CA 19-9 degradation. Further, the stabilizing agent may be active in a biological fluid, such as saliva, blood, serum, or cancer cells. In an exemplary embodiment, a stabilizing agent may comprise one or more of aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate.
  • Further, in an additional exemplary embodiment, the components of a stabilizing agent may be present at or about equal amounts, such as for example 1:1:1 in a stabilizing agent with three active components. Additionally, the stabilizing agents may in at least one embodiment inhibit degradation or inactivation of a diagnostic marker by at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 99%. Moreover, the stabilizing agent may, in at least one embodiment, inhibit degradation of a diagnostic marker for at least about 1 minute, at least about 5 minutes, at least about 10 minutes, at least about 15 minutes, at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 4 hours, or at least about 8 hours.
  • Additional Components for Stabilizing Agent Carriers:
  • According to at least one embodiment of the present disclosure, stabilizing agents may be incorporated into various carriers for their use with mammals. Such carriers may include various rinses, gums, lozenge, mouthwash, beverages, confectionary, washes, or other applicable vehicles to deliver a stabilizing agent to the site of a diagnostic marker.
  • Embodiments of a carrier, such as a rinse, according to the present disclosure may also comprise one or more additional component which may include an anti-caking agent, a chemical preservative, an emulsifying agent, a nutrient and dietary supplement, a sequestrant, a stabilizer, an additive, a synthetic and flavoring substance. Exemplary embodiments of these components are listed herein (See section entitled “Additional Components for Rinse”). Further, the one or more additional component may be a substance which has been labeled as Generally Recognized As Safe (GRAS) by the Food and Drug Administration of the United States of America.
  • A exemplary embodiment of a carrier of the present disclosure may be a mouthwash composition which may comprise water and pharmaceutically acceptable excipients or additives such as: one or more oils, such as an oil selected from the group comprising anethole, anisole, camphor, methyl salicylate, vanillin, eugenol, furaneol, linalool, menthol, thymol, cinnamaldehyde, citral, methyl butanoate, pentylbutanoate, pentylpentanoate, tea tree oil, peppermint oil, spearmint oil, pineapplemint oil and eucalyptus oil; sweetening agents, for example sorbitol; thickening agents, such as xanthan gum, carrageenan, carbomer, or HPMC (hydroxypropyl methyl cellulose); preservative agents, such as sodium benzoate, methyl paraben, or propyl paraben; emulsifiers, such as polysorbate 80 (or Tween™ 80); and/or at least one antacid such as aluminium or magnesium hydroxide.
  • Gum compositions may be provided in a variety of different forms, such as, for example, slab, pellet, sticks, balls, cubes, center-fill gums, candy gum, multi-region gum, multi-layer gum, bubble gum, deposited gums and compressed gums. The gum compositions also may include at least one flavor and a variety of optional additives. In some embodiments, the gum compositions described herein may be formed in a rope or sheet form and then processed into individual forms as described above.
  • A gum composition also may include a gum base. The gum base may include any component known in the chewing gum art. Such components may be water soluble, water-insoluble or a combination thereof. For example, the gum base may include elastomers, bulking agents, waxes, elastomer solvents, emulsifiers, plasticizers, fillers and mixtures thereof.
  • A ingestible beverage composition of the present disclosure may comprise any formulation commonly known in the art.
  • At least one embodiment of a carrier of the present application may comprise a liquid. The liquid, in at least one embodiment, is a non-toxic liquid. Further, the liquid may comprise water, a beverage containing water, or glycerin. Moreover, in at least one embodiment, a stabilizing agent may be at least partially dissolved in the liquid.
  • It will be appreciated that the above list of excipients and/or additives is provided merely by way of example and that various other such components may be used in the formulation of the present invention.
  • The following components, in at least one embodiment of the rinse of the present disclosure, may include:
  • 1. ANTI-CAKING AGENTS: aluminum calcium silicate, calcium silicate, magnesium silicate, sodium calcium aluminosilicate, and tricalcium silicate.
  • 2. CHEMICAL PRESERVATIVES: ascorbic acid, ascorbyl palmitate, benzoic acid, butylated hydroxyanisole, butylated hydroxytoluene, calcium ascorbate, calcium propionate, calcium sorbate, caprylic acid, dilauryl thiodipropionate, erythorbic acid, gum guaiac, methylparaben, potassium bisulfite, potassium metabisulfite, potassium sorbate, propionic acid, propyl gallate, propylparaben, sodium ascorbate, sodium benzoate, sodium bisulfite, sodium metahisulfite, sodium propionate, sodium sorbate, sodium sulfite, sorbic acid, stannous chloride, sulfur dioxide, thiodipropionic acid, tocopherols.
  • 3. EMULSIFYING AGENTS: cholic acid, desoxycholic acid, diacetyl tartaric acid esters of (M)mono- and diglycerides, glycocholic acid, mono- and diglycerides, monosodium phosphate derivatives of above, propylene glycol, ox bile extract, taurocholic acid.
  • 4. NUTRIENTS AND DIETARY SUPPLEMENTS: alanine, arginine, ascorbic acid, aspartic acid, biotin, calcium carbonate, calcium citrate, calcium glycerophosphate, calcium oxide, calcium pantothenate, calcium phosphate, calcium pyrophosphate, calcium sulfate, carotene, choline bitartrate, choline chloride, copper gluconate, cuprous iodide, cysteine, cystine, ferric phosphate, ferric pyrophosphate, ferric sodium pyrophosphate, ferrous gluconate, ferrous lactate, ferrous sulfate, glycine, histidine, inositol, iron (reduced), isoleucine, leucine, linoleic acid, lysine, magnesium oxide, magnesium phosphate, magnesium sulfate, manganese chloride, manganese citrate, manganese gluconate, manganese glycerophosphate, manganese hypophosphite, manganese sulfate, manganous oxide, mannitol, methionine, methionine hydroxy analogue, niacin, niacinamide D-pantothenyl alcohol, phenylalanine, potassium chloride, potassium glycerophosphate, potassium iodide, proline, pyridoxine hydrochloride, riboflavin, riboflavin-5-phosphate, serine, sodium pantothenate, sodium phosphate, sorbitol, thiamine hydrochloride, thiamine mononitrate, threonine, tocopherols, tocopherol acetate, tyrosine, valine, vitamin A, vitamin A acetate, vitamin A palmitate, vitamin B12, vitamin D2, vitamin D3, zinc sulfate, zinc gluconate, zinc chloride, zinc oxide, zinc stearate.
  • 5. SEQUESTRANTS: calcium acetate, calcium chloride, calcium citrate, calcium diacetate, calcium gluconate, calcium hexametaphosphate, calcium phosphate. monobasic, calcium phytate, citric acid, dipotassium phosphate, disodium phosphate, isopropyl citrate, monoisopropyl citrate, potassium citrate, sodium acid phosphate, sodium citrate, sodium diacetate, sodium gluconate, sodium hexametaphosphate, sodium metaphosphate, sodium phosphate, sodium potassium tartrate, sodium pyrophosphate, sodium pyrophosphate, tetra, sodium tartrate, sodium thiosulfate, sodium tripolyphosphate, stecaryl citrate, tartaric acid.
  • 6. STABILIZERS: acacia (gum arabic) , agar-agar, ammonium alginate, calcium alginate, carob bean gum, chondrus extract, ghatti gum, guar gum, potassium alginate, sodium alginate, sterculia (or karava) gum, tragacanth.
  • 7. ADDITIVES: acetic acid, adipic acid, aluminum ammonium sulfate, aluminum potassium sulfate aluminum sodium sulfate, aluminum sulfate, ammonium bicarbonate, ammonium carbonate, ammonium hydroxide, ammonium phosphate, ammonium sulfate, bees wax, bentonite, butane, caffeine, calcium carbonate, calcium chloride, calcium citrate, calcium gluconate, calcium hydroxide, calcium lactate, calcium oxide, calcium phosphate, caramel, carbon dioxide, carnauba wax, citric acid, dextrans, ethyl formate, glutamic acid, glutamic acid hydrochloride, glycerin, glyceryl monostearate, helium, hydrochloric acid, hydrogen peroxide, lactic acid, lecithin, magnesium carbonate, magnesium hydroxide, magnesium oxide, magnesium stearate, malic acid, methylcellulose, monoammonium glutamate, monopotassium glutamate, nitrogen, nitrous oxide, papain, phosphoric acid, potassium acid tartrate, potassium bicarbonate, potassium carbonate , potassium citrate, potassium hydroxide, potassium sulfate, propane, propylene glycol, rennet, silica aerogel, sodium acetate, sodium acid pyrophosphate, sodium aluminum phosphate, sodium bicarbonate, sodium carbonate, sodium citrate, sodium carboxy-methylcellulose, sodium caseinate, sodium citrate, sodium hydroxide, sodium pectinate, sodium phosphate, sodium potassium tartrate, sodium sesquicarbonate, sodium tripolyphosphate, succinic acid, sulfuric acid, tartaric acid, triacetin, triethyl citrate.
  • 8. SYNTHETIC FLAVORING SUBSTANCES: acetaldehyde, acetoin, aconitic acid, anethole, benzaldehyde, N-butyric acid, d- or l-carvone cinnamaldehyde, citral, decanal, diacetyl, ethyl acetate, ethyl butyrate, ethyl vanillin, eugenol, geraniol, geranyl acetate, glycerol tributyrate limonene, linalool, linalyl acetate, l-malic acid, methyl anthranilate, 3-methyl-3-phenyl glycidic acid, ethyl ester, piperonal, vanillin.
  • II. Indicator Carriers
  • In at least one additional embodiment of a carrier, of the present disclosure, the carrier comprises an indicator compound capable of binding to and/or reacting with a target to produce an indicator signal. The target in an exemplary embodiment may be a diagnostic marker, such as the presence of glucose or glucose in excess of a defined threshold, the presence of one or more glycosylated protein related to diabetes or pre-diabetes, the presence of cancer (or pre-cancer) markers, and the presence of cardiac (or pre-cardiac) markers.
  • A. Indicators
  • The indicator compound in an exemplary embodiment of a carrier according to the present disclosure may be any compound, chemical, or biological component which may interact with a target or byproduct of a target. For example, an indicator compound may comprise an antibody, a reactive chemical compound, a labeled molecule, or any combination thereof. Antibodies used in an embodiment of the present disclosure may be monoclonal or polyclonal and derived from any species (e.g. human, rat, mouse, rabbit, pig). Further, indicator molecules may be aptamers, proteins, peptides, small organic molecules, natural compounds (e.g. steroids), non-peptide polymers, WIC multimers (including MHC-dextramers, MHC-tetramers, MHC-pentamers and other MHC-multimers), or any other molecules that specifically and efficiently bind to other molecules are also marker molecules.
  • Labeled molecules, for use as indicator compounds, may be any molecule that absorbs, excites, or modifies radiation, such as the absorption of light (e.g. dyes and chromophores) and the emission of light after excitation (fluorescence from flurochromes). Additionally, labeled molecules may have an enzymatic activity, by which it catalyzes a reaction between chemicals in the near environment of the labeling molecules, producing a signal which include production of light (chemi-luminescence) or precipitation of chromophors, dyes, or a precipitate that can be detected by an additional layer of detection molecules.
  • Fluorescence labels may produce the presence of light at a single wavelength, or a shift in wavelengths. Exemplary fluorescent labels may include:
      • Fluor dyes, Pacific Blue™, Pacific Orange™, Cascade Yellow™;
      • AlexaFluor®(AF); o AF405, AF488.AF500, AF514, AF532, AF546, AF555, AF568, AF594, AF610, AF633, AF635, AF647, AF680, AF700, AF710, AF750, AF800;
      • Fluorescein (Flu) or any derivate of that, ex. FITC (fluorescein isothiocyanate) Cy-Dyes o Cy2, Cy3, Cy3.5, Cy5, Cy5.5, Cy7 Fluorescent Proteins; o RPE(R-phycoerythrin), PerCp, APC(Allophycocyanin); other of phycobillin containing proteins, e.g. phycobiliprotein o Green fluorescent proteins (GFP);
      • GFP and GFP derivated mutant proteins; BFP1CFP, YFP, DsRed, T1, Dimer2, mRFP1, MBanana, mOrange, dTomato, tdTomato, mTangerine, mStrawberry, mCherry Tandem dyes: o RPE-Cy5, RPE-Cy5.5, RPE-Cy7, RPE-AlexaFluor® tandem conjugates;
      • RPE-Alexa610, RPE-TxRed o APC-Aleca600, APC-Alexa610, APC-Alexa750, APC-Cy5, APC-Cy5.5 Multi fluorochrome assemblies o Multiple fluorochromes attached to a polymer molecule, such as a peptide/protein, Dex, or poly-sacceride. o Any combination of the fluorescent dyes involving in generation of FRET;
      • (Fluorescence resonance energy transfer) based techniques, Multiple fluorochromes associated or coupled to a polymeric molecule, or consisting of polymeric residues, lonophors; ion chelating fluorescent probes or Probes that change wavelength when binding a specific ion, such as calcium;
      • Probes that change intensity when binding to a specific ion, such as calcium; and
      • Combinations of fluorochromes on the same marker. The marker is not identified by a single fluorochrome but by a code of identification being a specific combination of fluorochromes, as well as inter related ratio's of intensities.
  • B. Target
  • The target of an exemplary indicator compound of the present disclosure may be any diagnostic marker or diagnostic condition for a disease state (or pre-disease state) of an individual. For example, the disease state may be diabetes, pre-diabetes, cardiac disease, pre-cardiac conditions, cancers of any stage and type, or pre-cancerous conditions. Specifically, the target may be the presence, or level of, glucose found in saliva for diabetes. Alternately, the target may be the presence, or level of, glycosylated proteins, such as advanced glycosylation end products. Further, the target may be the presence or level of cardiac markers, such as Creatine kinase-MB (CK-MB), myoglobin, homocysteine, C-reactive protein (CRP), troponin T (cTnT), and troponin I (cTnI). Moreover, the target may be the presence of cancer markers, such as Cancer Antigen 125, Cancer Antigen 15.3, Cancer Antigen 19.9, Prostate specific antigen, Carcinoembryonic Antigen, Alpha Feto-Protein, Epidermal growth factor receptor, Kallikrein 3, Vascular endothelial growth factor A (VEGF), Calcitonin, Chromogranin A, Gastrin, S100 alpha chain, Somatostatin, Thyroglobulin, V-erb-b2, cyckub-dependent kinase inhibitor 1 (p21), Breast Cancer Antigen 1 and 2 (BRCA1 and 2), MutL homolog 1 (MLH1), MutS homolog 2 (MSH2), MutS homolog 6 (MSH6), and postmeiotic segregation increased 1 and 2 (PMS1 and 2). Moreover, the target may be the presence or level of any diagnostic marker included herein.
  • C. Indicator Signal
  • The indictor signal in at least one embodiment of the present disclosure may comprise any detectable signal, including but not limited to, color change, fluorescence, and chemical/structural change of the target (and/or indicator compound) so as to be amenable to reacting with a secondary detection marker. Further, the detection of a signal may involve a secondary reactive molecule.
  • D. Glucose
  • In an embodiment of the present disclosure, glucose may be detected through an indicator rinse by a chemical or enzymatic method. Exemplary embodiments of this method may include alkaline copper reduction, alkaline ferricyanide reduction, glucose oxidase, and/or hexokinase.
  • In an exemplary embodiment of an indicator rinse of the present disclosure, the rinse may further comprise one or more additional component as described herein. These additional components may include stabilizing agents, as well as GRAS components (such as those listed herein).
  • E. Glycosylated Hemoglobin
  • In an embodiment of the present disclosure, glycated Hemoglobin A1c (HbA1c) may be detected through any known method for the detection of HbA1c or fragments thereof, such as high pressure liquid chromatography, immunoassays, and an indicator rinse by a chemical or enzymatic method. In an exemplary embodiment of an indicator rinse of the present disclosure, the rinse may further comprise one or more additional component as described herein. These additional components may include stabilizing agents, as well as GRAS components (such as those listed herein).
  • F. Cancer Antigen 19-9
  • In an embodiment of the present disclosure, cancer antigen 19-9 (CA 19-9) may be detected through any known method for the detection of CA 19-9 or fragments thereof, such as high pressure liquid chromatography, immunoassays, and an indicator rinse by a chemical or enzymatic method. In an exemplary embodiment of an indicator rinse of the present disclosure, the rinse may further comprise one or more additional component as described herein. These additional components may include stabilizing agents, as well as GRAS components (such as those listed herein).
  • III. Diagnostic Marker Detection
  • In an embodiment of the present disclosure, a diagnostic marker susceptible to degradation or alteration by a destructive component may be detected through any known method, including, but not limited to, high pressure liquid chromatography, immunoassays, and an indicator rinse by a chemical or enzymatic method. Further, the detection may be conducted in a micro well, deep well, microarray, microfluidic device, high throughput screen, or any applicable diagnostic device.
  • A. Rapid Diagnostic
  • Detection of a diagnostic marker may be conducted according to a rapid detection system of the present disclosure. Turning to FIG. 1A, a detection platform 100 may comprise a platform 102 having a plurality of test wells 104, where each well is capable of containing a plurality of test spots 106. The plurality of test wells 104 may include at least 2, at least 4, at least 8, at least 16, at least 32, at least 64, or at least 256 wells. The plurality of tests spots 106 may include at least 2, at least 4, at least 8, at least 16, at least 32, or at least 64 test spots 106. Further, each of the plurality of test spots 106 may comprise one or more diagnostic marker or control marker.
  • An embodiment of detection platform 100 may further be sized and shaped to allow for automated detection and analysis of a diagnostic or control marker. In at least one embodiment, the detection system 100 may comprise an identifier 108, such as a barcode or Radio-frequency identification (RFID) tag, to allow for the identification of the detection system 100 and data collected from the analysis of test spots 106.
  • Further, detection platform 100 may be comprised of any applicable material sufficient to house or define test wells 104. Exemplary embodiments of applicable materials for detection platform 100 may be a polymer, glass, metal, quartz, nylon, silica-based material, resin, or combination thereof. Further, embodiments of test wells 104 may include not only a depression defined or housed by platform 102, but may also include a defined area or region of the platform 102 (such as may be found on a microarray based platform).
  • Turning to FIG. 1B, at least one embodiment of detection system 150 of the present disclosure is shown. An exemplary embodiment of detection system 150 comprises an embodiment of detection platform 100, a detection device 120 capable of determining a binding characteristic between a detection agent and a diagnostic marker on the detection platform 100, and a computer processor 130 coupled to a computer database 135 and to the detection device 120. In at least one embodiment, the computer processor 130 controlling the detection device 120 is able to (1) determine the binding characteristic of a detection agent to a diagnostic agent, (2) compare the binding characteristic among each of a plurality of samples tested, (3) generate a binding report using the compared binding characteristics, and deliver the binding report to a recipient or external computer (not shown) in communication with the computer processor 130.
  • A binding characteristic as used herein may include any measurement of the attraction (or repulsion) of two molecules (such as a detection agent and a diagnostic marker).
  • Detection system 100 may also comprise an embodiment of a bead-based system of the present disclosure having at least one bead type. For example, a dual bead system may be used wherein at least one bead type is magnetic or paramagnetic. These beads may also be bound to a carrier protein with a conjugated hapten. Further, the beads may have an analyte capture monoclonal antibody reagent bound to it. Additionally, the beads, in at least one embodiment, do not retain their magnetic properties when removed from a magnetic field. In at least one embodiment, the beads may be Europium micro-particles. The sizes of the Europium micro-particles may be varied as needed to accommodate the method of detection used.
  • At least one embodiment of a bead based system may comprise a latex particle (See FIGS. 2-4). The latex particle according to at least one embodiment may be a Carboxylate modified latex (CML) bead. Further, an exemplary embodiment of a bead may include bovine serum albumin (BSA) and/or human serum albumin (HSA). Additionally, an embodiment of a bead based system of the present disclosure may include a linker coupled to HSA and/or BAS. The linker according to an embodiment, may comprise a maleimide compound. At least one exemplary embodiment of a maleimide compound may be Succinimidyl 4-[N-Maleimidomethyl] Cyclohexane-1-Carboxylate (SMCC) or N-Hydroxysuccinimide-activated hexa(ethylene glycol) undecane Thiol (NHS). Moreover, at least one exemplary embodiment of a bead based system may further comprise an antibody linked to maleimide on the bead. Accordingly, at least one embodiment of the bead based system comprises a latex particle coupled to BAS or HSA, maleimide coupled to BAS or HSA, and an antibody coupled to maleimide.
  • The latex particle, according to at least one embodiment may be in a size range from about 0.02 μm to about 7.0 μm.
  • Turning to FIG. 5, at least one at least one method 500 of coupling an antibody to a bead comprises the step 502 of modifying an embodiment of a bead (such as a latex bead/particle) with BSA or HAS, wherein the BSA or HSA may further comprise a maleimide containing molecule, such as SMCC or NHS. Additionally, an embodiment of the method 500 may further comprise the step 504 of attaching a diagnostic marker binding agent (such as an antibody) to the bead. Step 504 of attaching the diagnostic marker binding agent to the bead may be accomplished through binding the diagnostic marker binding agent to maleimide. Further, method 500 may also comprise the step 506 of attaching one or more stabilizing agent to an embodiment of a bead. Additionally, while an embodiment of method 500 may use latex beads for attachment of a maleimide containing molecule and/or a stabilizing agent, alternate embodiments of the bead may comprise silicon, a polymer, or other applicable materials.
  • According to at least one embodiment of the present disclosure, one or more stabilizing agent may be attached or adsorbed to any one of an embodiment of a detection platform 100, test strip, microchip, or microfluidic device.
  • B. Test Strip
  • Turning to FIGS. 6A and B, exemplary embodiments of a diagnostic testing device 600 are described. An exemplary embodiment of diagnostic testing device 600 comprises housing structure 602 which comprises a collection chamber 604 capable of receiving body fluids, at least one membrane test strip 606 in fluid communication with collection chamber 604, an immunoassay-based fingerprint acquisition pad 608 in fluid communication with the collection chamber, and a plurality of reaction zones 610 which may allow the visual display of the presence of a predetermined diagnostic marker. For example, reaction zone 610 may in an exemplary embodiment be capable of displaying a characteristic of the predetermined diagnostic marker, such as the presence of, the concentration of, or a concentration above or below a predetermined value for the predetermined diagnostic agent.
  • In an exemplary embodiment of diagnostic testing device 600, diagnostic testing device 600 further comprises a fluid collector 612 which is operable to collect body fluids from a subject, and wherein the fluid collector is capable of supply collected body fluid to collection chamber 604.
  • Exemplary embodiments of test strip 606 may comprise any material capable of adsorbing or attaching a stabilizing agent and may bind a diagnostic marker. Further, exemplary embodiments of test strip 606 may comprise compositions comprising a polyvinyl chloride-silica combination, nitrocellulose, or any suitable synthetic, resinous material. At least one embodiment, housing 602 is capable of reducing the exposure of the test strip 606 at sites other than the collection chamber 604 to foreign material. Test strip 606 in at least one embodiment may be capable of interfacing with an analysis device 615. The analysis device 615 in an exemplary embodiment may be capable of measuring the level of the predetermined diagnostic marker in the sample.
  • Turning to FIG. 6C, an exemplary embodiment of a microarray system 620 is described. An exemplary embodiment of microarray system 620 may comprise a microarray product 622 having a microarray identifier 624, and a plurality of microarray sites 626, a control microarray product 628 located on at least one microarray site 626 and bound to microarray product 622, and operably coupled to a computer processor 130 for providing information regarding the identification and concentration of markers on the microarray product 622 based on the microarray identifier 624. Additionally, microarray product 622 may further comprise, in at least one embodiment, an embodiment of a stabilization agent 630 located on at least one microarray site 626 and bound to microarray product 622. Further, microarray product 622 may comprise one or more diagnostic marker 632 located on at least one microarray site 626 and bound to microarray product 622. Moreover, microarray system 620, in at least one embodiment, may further comprise a microarray detector 634 operable to detect a signal from at least one of the control microarray product 628 and/or diagnostic marker 632.
  • A microchip or microarrays may refer to an array of distinct polynucleotides affixed to a substrate, such as glass, plastic, paper, nylon or other type of membrane, filter, chip, or any other suitable solid support. The polynucleotides can be synthesized directly on the substrate, or synthesized separate from the substrate and then affixed to the substrate. In an embodiment, the microarray may be prepared and used at least according to the methods described in U.S. Pat. No. 5,837,832, Chee et al., PCT application WO95/11995 (Chee et al.), Lockhart, D. J. et al. (1996; Nat. Biotech. 14: 1675-1680) and Schena, M. et al. (1996; Proc. Natl. Acad. Sci. 93: 10614-10619), all of which are incorporated herein in their entirety by reference.
  • Turning to FIG. 6D, an exemplary embodiment of microfluidic device 640 is described. An embodiment of microfluidic device 640 comprises a sample reservoir 642 for receiving a body fluid (or other fluid sample) through a sample input 644, where the sample reservoir 642 is fluidly connected to a detector array 646. Additionally, in at least one embodiment of microfluidic device 640, detector array 646 comprises a reaction site 647 and a results display 648. Moreover, an exemplary embodiment of microfluidic device 640 may further comprise a filter device 650 capable of separating at least one unwanted component from a fluid sample and fluidly coupled between sample reservoir 642 and detector array 646. Detector array 646 may also, in at least one embodiment comprise a control reagent display 652, where at least one control reagent may be visually detected.
  • Further, microfluidic device 640 may also, in at least one embodiment, be able to couple to a processor 655. Processor 655 may be able to compare at least one reading, such as from results display 648 or control reagent display 652 from an embodiment of microfluidic device 640 to at least one additional stored reading on a computer database 657 of the processor 655. An embodiment of microfluidic system 660 of the present disclosure, may comprise one or more microfluidic device 640 operationally coupled to a processor 655.
  • Embodiments of microfluidic devices 640, which may also be referred to as “lab-on-a-chip” systems, biomedical micro-electro-mechanical systems (bioMEMs), or multicomponent integrated systems, are exemplary kits/systems for analyzing polynucleotide regions. Such systems may miniaturize and compartmentalize processes such as probe/target hybridization, nucleic acid amplification, and capillary electrophoresis reactions in a single functional device. Such microfluidic devices 640 typically utilize detection reagents in at least one aspect of the system, and such detection reagents may be used to detect one or more polynucleotide region of the present disclosure. Exemplary microfluidic devices 640 may comprise a pattern of microchannels designed onto a glass, silicon, quartz, or plastic wafer included on a microchip. The movements of the samples may be controlled by electric, electroosmotic or hydrostatic forces applied across different areas of the microchip to create functional microscopic valves and pumps with no moving parts. Varying the voltage can be used as a means to control the liquid flow at intersections between the micro-machined channels and to change the liquid flow rate for pumping across different sections of the microchip.
  • In at least one embodiment of a microarray or microfludic device 640, the microarray or microfluidic device may comprise one or more stabilizing agent capable of reducing the degradation of a predefined diagnostic marker. For example, a stabilizing agent may be incorporated onto the surface of the microarray system 620, and/or on at least one part of the microfluidic device 640.
  • IV. Methods of Biomarker Stabilization
  • In at least one embodiment of a method for stabilizing biomarkers, as described in FIG. 7, the method 700 comprises the step 702 of mixing a body fluid with an embodiment of a stabilizing agent. Optionally, the step of mixing a body fluid with a stabilizing agent may comprise the step 704 of introducing the stabilizing agent into the patient, such as through the oral cavity, so that the stabilizing agent mixes with the bodily fluid, and the step 706 of retrieving a body fluid comprising a stabilized diagnostic marker. In an additional exemplary embodiment of a method of stabilizing a diagnostic marker, the method comprises the step 708 of isolating a body fluid having a diagnostic marker, the step 710 of treating the body fluid with an embodiment of a carrier, such as a rinse, gum, or beverage, as described herein, and the step 712 of analyzing the diagnostic marker. Isolation of the body fluid may occur through any customary mechanism, including, but not limited to, rinsing, swabbing, suction, collection, and lavage. Further, the rinse in an exemplary embodiment of a method 700 of the present disclosure, may be ingested to stabilize a diagnostic marker present in the bodily fluid, such as in urine, semen, anal secretions, and vaginal secretions.
  • The step 702 of mixing a body fluid with a carrier may occur prior to, or after isolation step 708 of the body fluid. As an exemplary embodiment, a rinse may be used to rinse the mouth. Afterwards, the rinse containing the treated body fluid may be collected by spitting, suction, or other means. Alternately, expirated saliva may be mixed with a rinse to treat the saliva ex vivo. Similar treatment of body fluids may be performed with any type of body fluid.
  • In analyzing the treated body fluid, the diagnostic marker may be analyzed through any known means. Optionally, the analysis of the treated body fluid may include the separation of solid materials from soluble materials through means such as filtration, or by centrifugation. Analysis of the treated body fluid may use any appropriate technique, such as western blot analysis, Enzyme-Linked Immunosorbent Assay (ELISA), protein activity assays, reverse transcription polymerase chain reaction (RT-PCR), microarray, high pressure liquid chromatography, or any comparable assay to determine a characteristic of the diagnostic marker. Such an analysis, in an exemplary embodiment, may be of a modified product, a cleavage product, or cleavage pattern, of a diagnostic marker.
  • In an exemplary embodiment of the analyzing step 712, the treated body fluid may be placed in contact with an embodiment of a test strip, such as a nitrocellulose strip, prior to detection with an appropriate probe specific for the diagnostic marker. The nitrocellulose strip may be designed to trap or filter particulates in the body fluid. This may reduce or eliminate potential contaminants or other substances in the oral fluid that would otherwise reduce the signal to noise ration during the detection step. A probe (such as an antibody) with affinity to the diagnostic marker may be affixed or immobilized to a specific location on the nitrocellulose membrane for detection of the analyte. Utilizing standard principles of immunoassay detection (or nucleotide detection), the signal generated may be visible to the eye or detectable by an instrument.
  • In an exemplary embodiment of a method of analyzing a stabilized diagnostic marker of the present disclosure as depicted in FIG. 8, the method 800 comprises the step 802 of operating a system for the detection of a diagnostic marker in a body fluid, where the system comprises a diagnostic device having a plurality of test wells each capable of containing at least one diagnostic marker binding agent, and a detection device capable of interacting with diagnostic device, wherein the detection device is capable of detecting an interaction between the at least one diagnostic marker binding agent and a diagnostic marker. Following step 802, the sample of bodily fluid may be diluted in step 803 with a reagent which may contain an embodiment of stabilizing agent as described herein. At least one embodiment of method 800 further comprises the steps of: contacting 804 a sample of a bodily fluid with at least one diagnostic marker binding agent, incubating 806 the contacted sample in the test well at a first temperature for a pre-determined period of time. The step 804 of contacting of the sample of bodily fluid with at least one diagnostic marker binding agent may be accomplished through a magnetic field applied to the test well.
  • In at least one embodiment of method 800, the first temperature may be at room or at body temperature (such as about 22° C. or about 37° C.). The pre-determined period of time may, for example, be between five and fifteen minutes. The incubated and contacted sample may then be removed in step 808 from the plurality of test wells, and the test wells analyzed in step 810 to detect a binding event with the detection device. Alternately, a stabilizing agent may be incubated with the diagnostic marker binding agent (also referred to as a detection agent) prior to introduction of a bodily fluid. In at least one embodiment of the method, each diagnostic marker binding agent may have a different monoclonal antibody designed to bind a specific hapten molecule.
  • Following the detection of a characteristic of a treated diagnostic marker (the results of which may be termed a “profile” such as a “cancer profile”), the characteristic may in step 812 be compared to a standard value to determine the presence or absence of a disease state. The characteristic determined may be an activity level, the concentration of the diagnostic marker, or a particular modification, such as glycosylation or methylation.
  • Optionally, an embodiment of the method 800 may further comprise the step 814 of customizing a detection method through screening of applicable pre-clinical samples through a database of stabilizing agents/cocktails. Further, an embodiment of method 800 of diagnostic marker detection may further comprise the step 816 of comparing a level or characteristic (such as a predictable degradation products) of a diagnostic agent with a library of known characteristics. Moreover, an exemplary embodiment of method 800 may additionally comprise the step 818 of determining a diagnosis of a disease state using the compared characteristic. In at least one embodiment, a method of diagnostic marker detection may be automated, and capable of detecting at least about ten thousand samples in a twenty-four hour period. Method 800 in at least one embodiment, may be at least partially completed by a computer processor. Additionally, the processor may be in communication with at least one more processor and/or a computer database. For instance, in at least one embodiment of method 800, most of the steps of the method may be completed with or monitored by a computer processor. Moreover, method 800, in at least one embodiment, may use a computer processor to perform additional step 820 of generating a report by using a comparison of determined characteristics (such as a binding report where a comparison is performed of binding characteristics), and step 822 of delivering the report to a recipient (such as a user, or secondary processor).
  • A method of biomarker stabilization may, in an exemplary embodiment, may be used to diagnose a particular disease state or condition of a patient. For example, a disease state or condition as used with this method may include one or more of Acid-Base Disorders, Acidosis and Alkalosis, Acidosis/Alkalosis, Acute inflammatory demyelinating polyneuropathy, Acute myocardial infarct, Addison's Disease, Adrenal Insufficiency, Adrenal Insufficiency & Addison's Disease, Alcohol dependence, Alcoholism, Allergies, Alzheimer's Disease, Anemia, Angina Pectoris, Anthrax, Arthritis, Asthma, Atypical Pneumonia, Autoimmune Disorders, Autoimmune thyroiditis, Avian flu, Benign Prostatic Hyperplasia, Benign Prostatic Hypertrophy, Bioterrorism Agents, Bleeding Disorders, Bone Marrow Disorders, Breast Cancer, Cardiovascular Disease, Celiac Disease, Cervical Cancer, Chlamydia, Chronic Fatigue and Immune Dysfunction Syndrome, Chronic Fatigue Syndrome, Chronic thyroiditis, Colon Cancer, Community-Acquired Pneumonia, Congestive Heart Failure, Conn's Syndrome, Cushing's Syndrome, Cystic Fibrosis, Degenerative Joint Disease, Diabetes, Diarrhea, Diffuse thyrotoxic goiter, Diseases of the Pancreas, Disseminated lupus erythematosus, Double pneumonia, Down Syndrome, Encephalitis, Endocrine Syndromes, Endocrine System and Syndromes, Epilepsy, Fibromyalgia, Flu, Folate Deficiency, Fungal Infections, Genital Herpes, Gonorrhea, Gout, Gouty arthritis, Graves' Disease, Guillain-Barre Syndrome, Influenza H1N1, Hashimoto's Thyroiditis, Healthcare-Associated Pneumonia, Heart Attack, Heart Attack and Acute Coronary Syndrome, Heart Disease, Hemochromatosis, Hepatitis, Herpes, Herpes Zoster, High blood pressure, Hospital-Acquired Pneumonia, Human Immunodeficiency Virus, Human Papillomavirus, Hypercoagulable Disorders, Hypersensitivity, Hypertension, Hyperthyroidism, Hypothyroidism, Infectious Arthritis, Infectious polyneuritis, Infertility, Inflammatory Bowel Disease, Influenza, Influenza A, Influenza B, Insulin Resistance, Jaundice, Juvenile Rheumatoid Arthritis, Kidney and Urinary Tract Function, Disorders, and Diseases, Kidney Disease, Landry's ascending paralysis, Lead Poisoning, Leukemia, Liver Disease, Lobar pneumonia, Lower Respiratory Tract Infection, Lung Diseases, Lupus, Lupus erythematosus, Lyme Disease, Lymphoma, Malnutrition, Meningitis, Meningitis and Encephalitis, Menopause, Metabolic Syndrome, Metabolic Syndrome/Syndrome X, Multiple Myeloma, Multiple Sclerosis, Myeloproliferative Disorders, Myocardial infarct, Neural Tube Defects, Nontuberculous Mycobacteria, Osteoarthritis, Osteoporosis, Ovarian Cancer, Pancreatic Cancer, Pancreatic Diseases, Pancreatic Insufficiency, Pancreatitis, Pelvic Inflammatory Disease, Peptic Ulcer, Pituitary Disorders, Pneumonia, Polycystic ovarian syndrome, Pregnancy, Primary hyperaldosteronism, Prostate Cancer, Proteinuria, Rheumatoid Arthritis, Septic Arthritis, Sexually Transmitted Diseases, Sexually transmitted infections, Shingles, Sickle Cell Anemia, Sickle Cell Disease, Sjögren's Syndrome, Staph Wound Infections, Staph Wound Infections and Methicillin Resistant Staphylococcus aureus, Stein-Leventhal syndrome, Stroke, Swine flu, Syphilis, Systemic Lupus Erythematosus, Testicular Cancer, Thalassemia, Thyroid Diseases, Travelers' Diseases, Trichomonas, Tuberculosis, Types of Liver Disease, Urinary Tract Infection, Venereal diseases, Vitamin B12 and Folate Deficiency, Vitamin B12 Deficiency, Vitamin K Deficiency, Walking pneumonia, West Nile Virus, Wilson's Disease, Wound and Skin Infections.
  • V. Computational Analysis
  • In at least one embodiment of the systems, methods, and devices of the present disclosure, a computer processor and a database may be used in or during the system, method, or device. An exemplary embodiment of a system framework comprising at least one computer processor and at least one database, as may be used in the embodiments of the present disclosure is shown in FIG. 9.
  • In at least one embodiment, a result from a diagnostic method may be compared using a computer processor to at least one additional result from the diagnostic test, and/or to a result stored on a database. Such a comparison, in at least one embodiment of the present disclosure, may reveal the stabilizing agent with a greater effect on the binding characteristic, may allow for the diagnosis of a disease state or health/lifestyle characteristic. Further, such a comparison, in at least one embodiment, may be used to develop, confirm, or modify a therapeutic treatment of a patient having a disease state or health/lifestyle characteristic.
  • As shown in exemplary system framework 900 shown in FIG. 9, one or more user computers 902 may be operably connected to a system server 904. A user computer 902 may be a computer, computing device, or system of a type known in the art, such as a personal computer, mainframe computer, workstation, notebook computer, laptop computer, hand-held computer, wireless mobile telephone, personal digital assistant device, and the like.
  • One or more administrator computers 906 may also be operably connected to system server 904 including through a network 908 such as the Internet. Administrator computers 906, similar to user computers, may be computers, computing devices, or systems of a type known in the art, such as personal computers, mainframe computers, workstations, notebook computers, laptop computers, hand-held computers, wireless mobile telephones, personal digital assistant devices, and the like. In addition, user computers 902 and administrator computers 906 may each comprise such software (operational and application), hardware, and componentry as would occur to one of skill of the art, such as, for example, one or more microprocessors, memory, input/output devices, device controllers, and the like. User computers and administrator computers 906 may also comprise one or more data entry means (not shown in FIG. 9) operable by a user of client computer and/or an administrator computer, such as, for example, a keyboard, keypad, pointing device, mouse, touchpad, touchscreen, microphone, and/or other data entry means known in the art. User computers 902 and administrator computers 906 also may comprise an audio display means (not shown in FIG. 9) such as one or more loudspeakers and/or other means known in the art for emitting an audibly perceptible output. The configuration of user computers and administrator computers 906 in a particular implementation of one or more systems of the present disclosure is left to the discretion of the practitioner.
  • System server 904 may comprise one or more server computers, computing devices, or systems of a type known in the art. System server 904 may comprise server memory. System server 904 may comprise one or more components of solid-state electronic memory, such as random access memory. System server 904 may also comprise an electromagnetic memory such as one or more hard disk drives and/or one or more floppy disk drives or magnetic tape drives, and may comprise an optical memory such as a Compact Disk Read Only Memory (CD-ROM) drive. System server 904 may further comprise such software (operational and application), hardware, and componentry, as would occur to one of skill of the art, such as, for example, microprocessors, input/output devices, device controllers, video display means, and the like.
  • System server 904 may comprise one or more host servers, computing devices, or computing systems configured and programmed to carry out the functions allocated to system server 904. System server 904 may be operated by, or under the control of, a “system operator,” which may be an individual or a business entity. For purposes of clarity, System server 904 is shown in FIG. 9 and referred to herein as a single server. System server 904 need not, however, be a single server. System server 904 may comprise a plurality of servers or other computing devices or systems connected by hardware and software that collectively are operable to perform the functions allocated to the various systems of present disclosure. Specifically, system server 904 may be operable to be a web server, configured and programmed to carry out the functions allocated to a system server according to the present disclosure. Further, although user computers 902 and administrator computers 906 may be connected directly to system server 904, these computers may be connected to system server 904 through any suitable network, such as network 908. Further, in one embodiment, the users need not be provided access to system server 904, but instead the content posts from users are made by the user(s) and saved to one or more particular locations and the posts are accessed or harvested by the administrator or system automatically.
  • System server 904 may be operably connected to the various user computers 902 and/or an administrator computers 906 by network 908, which in an embodiment of the present disclosure comprises the Internet, a global computer network. However, network 908 need not comprise the Internet. Network 908 may comprise any means for electronically interconnecting system server 904 and a user computer 902 and/or an administrator computer 906. Thus, it will be appreciated by those of ordinary skill in the art that the network 908 may comprise the Internet, the commercial telephone network, one or more local area networks, one or more wide area networks, one or more wireless communications networks, coaxial cable, fiber optic cable, twisted-pair cable, the equivalents of any of the foregoing, or the combination of any two or more of the foregoing. In an embodiment where system server 904 and user computer 902 and/or an administrator computer 906 comprise a single computing device operable to perform the functions delegated to both system server 904 and user computer 902 and/or an administrator computer 906 according to the present disclosure, network 908 comprises the hardware and software means interconnecting system server 904 and user computer 902 and/or an administrator computer 906 within the single computing device. Network 908 may comprise packet switched facilities, such as the Internet, circuit switched facilities, such as the public switched telephone network, radio based facilities, such as a wireless network, etc.
  • The various systems, methods, schema, ontologies, and architectures of the present disclosure may be used for purposes outside of the medical transcription field as referenced in the various examples cited herein. For example, the system for analyzing verbal records may comprise various components and relationships suitable for use in any number of areas where various experiences are utilized and processed, with feedback being fed back into system componentry to improve overall system outcomes. In addition, various components described herein may share a name (or a portion thereof) but have duplicative reference numbers, and therefore the descriptions for the various components should read in view of one another.
  • In addition, and regarding the various systems of the present disclosure, such systems may be operable, as desired by a user of such systems, to generate visual, electronic (video, audio, database, transcript, etc.), and/or printed reports, outputs, outcomes, and the like. Such exemplary outputs may be used for any number of purposes, and may be useful generally to “report” results, data, and/or knowledge contained within and generated from such systems. Furthermore, the disclosure of the present application further encompasses uses of the various methods, systems, architectures, etc., to perform various tasks in connection therewith.
  • EXAMPLES Examples
  • 1. α-Amylase Inhibition Saliva Screening Method.
  • At least one assay used for the detection of α-amylases activity, as used herein includes the use of a chromagenic substrate, 2-chloro-p-nitrophenol linked to maltotriose. Enzymatic activity of α-amylase on the substrate yields 2-chloro-p-nitrophenol, which can be measured spectrophotometrically at 405 nm. The amount of α-amylase activity present in the experimental sample is directly proportional to the increase in absorbance at 405 nm.
  • For the assay, test compounds are mixed with saliva, prior to the addition of α-amylase substrate. The test compounds, such as GRAS materials, are prepared at a concentration of 5,000 ppm in distilled water. Following this preparation, 300 μl of α-amylase substrate (Salimetrics α-amylase Assay Kit) that has been pre-heated to 37° C. is added to 20 μl of the test compound. To initiate the assay, 10 μl of a dilute (3.5%) or full strength (100%) saliva solution is added to the mixture. Following the initiation, each mixture is measured at 1 minute intervals using a spectrophotometer (such as Molecular Devices M5 reader) at 405 nm. Temperatures of the mixtures are kept constant at 37° C. during the assay.
  • 2. α-Amylase Inhibitor Assays.
  • A series of GRAS compounds were tested using the α-amylase assay protocol described here. The tested compounds are included in Table I. From the screen of GRAS compounds, five compounds (CDI-030, 034, 036, 037 and 042) were shown to have α-Amylase Inhibitor Activity in dilute (3.5%) saliva (See FIG. 10). Further details of these five compounds may also be seen in Table 4.
  • TABLE 4
    100% 0%
    Saliva Amylase
    Time CDI-30 CDI-34 CDI-36 CDI-37 CDI-42 Control Control
    0 min 1.60 0.40 3.63 4.00 0.80 4.00 0.09
    2.03 0.36 3.20 3.85 1.13 4.00 0.09
    2.02 0.40 3.46 4.00 0.90 4.00 0.09
    1.87 0.45 4.00 4.00 0.84 4.00 0.10
    % CV 10.67 9.13 9.42 1.85 16.35 0.00 1.84
    Ave 1.88 0.40 3.57 3.96 0.92 4.00 0.09
    % Std 46.96 10.04 89.29 99.08 22.93 100.00 2.34
    % Inhibition 53.04 89.96 10.71 0.92 77.07 0.00 97.66
    5 min 4.00 0.49 4.00 4.00 1.65 4.00 0.09
    4.00 0.50 4.00 4.00 2.15 4.00 0.09
    4.00 0.50 4.00 4.00 1.80 4.00 0.09
    4.00 0.52 4.00 4.00 1.81 4.00 0.10
    % CV 0.00 2.32 0.00 0.00 11.31 0.00 2.79
    Ave 4.00 0.50 4.00 4.00 1.85 4.00 0.09
    % Std 100.00 12.61 100.00 100.00 46.24 100.00 2.36
    % Inhibition 0.00 87.39 0.00 0.00 53.76 0.00 97.64
    15 min  4.00 0.53 4.00 4.00 2.37 4.00 0.09
    4.00 0.61 4.00 4.00 2.38 4.00 0.10
    4.00 0.54 4.00 4.00 2.25 4.00 0.09
    4.00 0.56 4.00 4.00 2.17 4.00 0.10
    % CV 0.00 6.67 0.00 0.00 4.33 0.00 1.88
    Ave 4.00 0.56 4.00 4.00 2.29 4.00 0.09
    % Std 100.00 14.02 100.00 100.00 57.35 100.00 2.37
    % Inhibition 0.00 85.98 0.00 0.00 42.65 0.00 97.64

    Among these five compounds, CDI-034 and CDI-42 were shown to inhibit α-Amylase in 100% saliva samples (See FIG. 11).
  • 3. Lysozyme Inhibition Saliva Screening Method.
  • At least one assay used for the detection of α-amylases activity, as used herein includes the use of a Micrococcus lysodeikticus labeled with fluorescein. The assay measures lysozyme activity on Micrococcus lysodeikticus cell walls, which are labeled to such a degree that the fluorescence is quenched. Lysozyme activity can relieve this quenching; yielding increased fluorescence that is proportional to lysozyme activity.
  • For the assay, test compounds are mixed with saliva, prior to the addition of lysozyme substrate. The test compounds, such as GRAS materials, are prepared at a concentration of 5,000 ppm in distilled water. Following this preparation, 50 μl of lysozyme substrate at 1 mg/ml (Molecular Probes EnzChek Lysozyme Assay Kit) that has been pre-heated to 37° C. is added to 50 μl of the test compound. To initiate the assay, 50 μl of a dilute (3.5%) or full strength (100%) saliva solution is added to the mixture. Following the initiation, each mixture is measured at 1 minute intervals using a spectrophotometer (such as Molecular Devices M5 reader) for absorption at 494 nm and fluorescence emission at 518 nm. Temperatures of the mixtures are kept constant at 37° C. during the assay.
  • 4. Lysozyme Inhibitor Assays.
  • A series of GRAS compounds were tested using the Lysozyme assay protocol described above. The tested compounds are included in Table I. From the screen of GRAS compounds, four compounds including Fixanal® Buffer 6.0 (Sigma-Aldrich Co., CDI-030), bentonite (CDI-37), benzoic acid (CDI-40), and acetic acid (CDI-047) were shown to have Lysozyme Inhibitor Activity in dilute (3.5%) saliva (See FIG. 12). Further details of these five compounds may also be seen in Table 5.
  • TABLE 5
    Cell RFU Reading
    Location (60 mins)
    CDI
    Candidate #
    30 F 4 51.13
    37 E 5 92.06
    40 H 5 35.82
    47 G 6 33.53
    CONTROLS
    Pooled Saliva E 12 127.33
    Fresh Saliva F 12 182.02
    Lysozyme G 12 233.43

    Among these four compounds, benzoic acid (CDI-40), and acetic acid (CDI-047) were shown to inhibit Lysozyme in 100% saliva samples (data not shown).
  • 5. Galactose Oxidase Inhibition Saliva Screening Method.
  • At least one assay used for the detection of galactose oxidase activity, as used herein, includes the use of a chromagenic or fluorogenic substrate, such as with Amplex® Red (Molecular Probes). In an embodiment of the assay used herein, galactose oxidase catalyzes the oxidation of galactose at the C6 position and generates hydrogen peroxide (H2O2). The H2O2 then, in the presence of horseradish peroxidase (HRP), reacts with 1:1 stoichiometry with Amplex® Red reagent to generate the red-fluorescent oxidation product, resorufin. Resorufin has absorption and fluorescence emission maxima of approximately 571 nm and 585 nm, respectively, and because the extinction coefficient is high (54,000 cm-1M-1), the assay can be performed either fluorometrically or spectrophotometrically.
  • For the assay, test compounds are mixed with Amplex Red, reaction buffer, horseradish peroxidase (HRP) at 100 U/ml, and galactose stock solution (according to manufacturers recommended protocol, Molecular Probes A22179), prior to the addition of saliva. The test compounds, such as GRAS materials, are prepared under various concentration for testing. Following this preparation, 50 μl of the test compound solution (with Amplex Red, reaction buffer, and HRP) is added to 50 μl of saliva. Following the initiation, each mixture is measured at 1 minute intervals using a spectrophotometer (such as Molecular Devices M5 reader) using excitation in the range of 530-560 nm and emission detection at about 590 nm or absorbance at 560 nm. Temperatures of the mixtures are kept constant at 37° C. during the assay.
  • 6. Galactose Oxidase Inhibitor Assays.
  • A series of GRAS compounds were tested using the Galactose Oxidase assay protocol described herein. The tested compounds are included in Table I. From the screen of GRAS compounds, four compounds (CDI-039, 040, 042, and 047) were shown to have Galactose Oxidase Inhibitor Activity in dilute (3.5%) saliva (See FIG. 13). FIG. 13 includes the following symbols which are: open circle=Aluminum potassium sulfate dodecahydrate, open square=3-tert-butyl-hydroxyanisole, open triangle=benzoic acid, open diamond=acetic acid, filled circle=fresh saliva, and filled square=water (control blank).
  • 7. Stabilization of Glycosylated Hemoglobin.
  • At least one procedure used for the stabilization and detection of glycosylated hemoglobin (HbA1c), as described herein, includes the incubation of HbA1c with a stabilizing agent prior to, or concurrently with, detection. Analysis of samples was conducted on a HPLC 1100 series (Agilent Technologies) with a 2.1×150 (3.5μ) C18 reverse phase column. The mobile phase used was acetonitrile with 6.5 mM Ammonium Carbonate. A HbA1c standard was prepared by dissolving HbA1c (Sigma Aldrich, 405 RM) with 1.0 ml of deionized water. The samples analyzed by HPLC included pure HbA1c (FIG. 14), 100 ml HbA1c mixed with 100 ml of fresh saliva (FIGS. 15), and 200 ml HbA1c mixed with 100 ml of fresh saliva and 100 ml of a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1:1:1 ratio) (FIG. 16). While the pure HbA1c sample was analyzed fresh, samples of HbA1c with saliva were incubated overnight at room temperature prior to analysis. For each sample analyzed, the data was displayed through a three dimensional plot (FIGS. 14A, 15A, and 16A), a UV image (FIGS. 14B, 15B, and 16B), and a chromatogram (FIGS. 14C, 15C, and 16C). Additionally, a comparison of the analysis HbA1c in the presence of saliva with and without stabilizing agent is shown in FIGS. 17 (chromatography), 18 (UV image), and 19 (three dimensional plot).
  • 8. HbA1c Protection Assays.
  • A series of GRAS compounds were tested, as shown in FIGS. 14-16, for the ability to protect HbA1c from degradation by components in saliva. The tested compounds are included in Table I. As described above, a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1:1:1 ratio) minimized the degradation of HbA1c, and yielded a defined pattern for HbA1c.
  • 9. Stabilization of Cancer Antigen 19-9.
  • At least one procedure used for the stabilization and detection of Cancer Antigen 19-9 (CA 19-9) as described herein, includes the incubation of CA 19-9 with a stabilizing agent prior to, or concurrently with, detection. Analysis of samples was conducted on a HPLC 1100 series (Agilent Technologies) with a 2.1×150 (3.5μ) C18 reverse phase column. The mobile phase used was acetonitrile with 6.5 mM Ammonium Carbonate. A CA 19-9 standard was prepared by dissolving CA 19-9 with 1.0 ml of deionized water. The samples analyzed by HPLC included pure CA 19-9 (FIG. 20), 100 ml CA 19-9 mixed with 100 ml of fresh saliva (FIG. 21), and 200 ml CA 19-9 mixed with 100 ml of fresh saliva and 100 ml of a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1:1:1 ratio) (FIG. 22). While the pure CA 19-9 sample was analyzed fresh, samples of CA 19-9 with saliva were incubated overnight at room temperature prior to analysis. For each sample analyzed, the data was displayed through a three dimensional plot (FIGS. 20A, 21A, and 22A) and a UV image (FIGS. 20B, 21B, and 22B). Additionally, a comparison of the analysis CA 19-9 in the presence of saliva with and without stabilizing agent is shown in FIGS. 23 (three dimensional plot) and 24 (UV image).
  • 10. CA 19-9 Protection Assays.
  • A series of GRAS compounds were tested, as shown in FIGS. 20-22, for the ability to protect CA 19-9 from degradation by components in saliva. The tested compounds are included in Table I. As described above, a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1:1:1 ratio) minimized the degradation of CA 19-9 in at least one embodiment.
  • 11. Peroxidase Assay.
  • To determine the effect of various GRAS compounds as inhibitors of peroxidase activity, a standard assay using Amplex® Red (Life Technologies), which releases a fluorescent oxidation product. This oxidation product, named resorufin, has an excitation and emission maxima of approximately 571 nm and 585 nm, respectively, and because the extinction coefficient is high (58,000±5,000 cm-1M-1), it allows for the use with fluorometric or spectrophotometric assays.
  • The test compounds, such as GRAS materials or cocktails of same, were prepared at a concentration of 500 ppm in distilled water. For the assay, 50 μl samples of test compounds were mixed with 20-30 μl of saliva (either “pooled” or “unpooled”) prior to the addition of Amplex® Red. Following the mixture of the test compounds with saliva, 50 μl of Amplex® Red/HRP working solution (See Protocol for Invitrogen Assay #A22188, which is incorporated herein in its entirety), which has a concentration of 100 μM of Amplex® Red, is added to the test compound/saliva mixture. Following the initiation of the reaction, each mixture is measured at 1 minute intervals using a spectrophotmoter (such as a Molecular Devices M5 reader) for excitation in the range of 530-560 nm and fluorescence emission detection at approximately 590 nl, or for absorption at approximately 560 nm. Reactions were allowed to run for 30 minutes during these assays.
  • 12. Peroxidase Protection Assays.
  • A series of GRAS compounds and cocktails of GRAS compounds were tested, as shown in FIGS. 25-29, for the ability to inhibit degradation of a peroxidase-sensitive marker by components in saliva. Various embodiments of the tested compounds are included in Table I. As described above, a stabilizing compound/cocktail comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1:1:1 ratio) minimized the degradation of a peroxidase sensitive marker in at least one embodiment (FIG. 25, open circle=blank control, open square=stabilizing cocktail). Further, inhibition assays were also conducted with caffeine (FIG. 26, CDI #33; Key: open circle=pooled saliva, open square=unpooled saliva, open triangle=caffeine, open triangle=caffeine (repeat)), 3-tert-butyl-hydroxyanisole (FIG. 27, CDI #39; Key: open circle=pooled saliva, open square=unpooled saliva, open triangle=3-tert-butyl-hydroxyanisole, open triangle=3-tert-butyl-hydroxyanisole (repeat)), benzoic acid (FIG. 28, CDI #16; Key: open circle=pooled saliva, open square=unpooled saliva, open triangle=benzoic acid, open triangle=benzoic acid (repeat)), and aluminum potassium sulfate dodecahydrate (FIG. 29, CDI #42; Key: open circle=pooled saliva, open square=unpooled saliva, open triangle=aluminum potassium sulfate dodecahydrate, open triangle=aluminum potassium sulfate dodecahydrate (repeat)), each of which showed significant peroxidase inhibition capabilities.
  • 13. Diagnostic Devices.
  • In an example of a diagnostic device, as shown in FIG. 30, a test slide with 28 test wells are spotted with 12 to 18 sets of diagnostic markers. These reagents may include diagnostic markers, or controls for the diagnosis. The diagnostic device, in this example, has four diagnostic markers spotted in duplicate, along with two positive and two negative control marker. Each marker is spotted with a competitor, such as anti-haptin, which binds the capture antigen particle to the marker. The device may have four different hatpin/anti-haptin systems to serve as the generic capture system. A unique haptin may be conjugated to a fully paramagnetic particle (PmMp). The PmMp is also coated with a capture monoclonal antibody (MAb) specific for the marker. A second particle (polystyrene microparticle with Europium) is conjugated with the second or detection MAb. In the diagnostic device shown in FIG. 30, the hash marks on the side of the diagnostic device (slide) are indentations molded into the plastic, which may be used as cog grooves. Additionally the slide is designed to fit into a processor that may have more than one processing station, as shown in FIG. 32.
  • In performing a method of detecting a diagnostic marker using an embodiment of the diagnostic device, such as that in FIGS. 30 and 32, the user adds a pre-determined amount of specimen (such as 120 μl) to a specimen diluent (such as 40 to 120 μl), and adds the mixture to each test well. Following addition of the diluted specimen, the test slide is incubated at 37° C. for about 5 to about 15 minutes with agitation. Following the incubation period, the test wells move over a magnetic source that brings the PmMp to the bottom of the well, so that there is maximal binding of the PmMp with the anti-haptin bound in the test well. Next, specimen and assay reagents are removed, wash buffer is added and aspirated off. Lastly, each well is scanned using a visualization device, such as a CCD camera, and then the fluroescent signal (or other signal) of each marker is determined.
  • 14. Fluid Collection Device.
  • An example of a collection device for the collection of an bodily fluids is depicted in FIG. 31. In this example, the collection device is capable of collecting at least 500 μl of bodily fluid, such as saliva. The cap is first removed from the tube. Squeezing the bulb provides suction adequate to pull an amount of saliva into the bulb. In some cases, a filter may be included in the tube to remove unwanted materials (such as particulate and mucous). The filter may be of any known filter material, such as rayon, that is appropriate for the purposes described herein. Following the obtaining of the bodily fluid in the collection device, a cap is placed on the tube, and a second cap is removed from a dispensing tube. The dispensing tube is in fluid communication with the bulb and is sized and shaped to allow for drops of a predetermined size to be formed when the bulb is squeezed. For example, the dispensing tube may allow drops of 30 μl to be formed when the bulb is squeezed.
  • While various embodiments of the systems, methods, and compositions of the present disclosure have been described in considerable detail herein, the embodiments are merely offered by way of non-limiting examples of the disclosure described herein. It will therefore be understood that various changes and modifications may be made, and equivalents may be substituted for elements thereof, without departing from the scope of the disclosure. Indeed, this disclosure is not intended to be exhaustive or to limit the scope of the present disclosure.
  • Further, in describing representative embodiments, the disclosure may have presented a method and/or process as a particular sequence of steps. However, to the extent that the method or process does not rely on the particular order of steps set forth herein, the method or process should not be limited to the particular sequence of steps described. Other sequences of steps may be possible. Therefore, the particular order of the steps disclosed herein should not be construed as limitations of the present disclosure. In addition, disclosure directed to a method and/or process should not be limited to the performance of their steps in the order written. Such sequences may be varied and still remain within the scope of the present disclosure.

Claims (13)

1. A microfluidic device for analyzing the presence or absence of a diagnostic marker to be detected in a body fluid, the microfluidic device comprising:
a container, the container housing:
a sample reservoir having a sample input port in fluid connection with at least one detector array, wherein the detector array comprises at least one detector comprising a reaction chamber comprising an immobilized capture molecule;
a stabilizing agent contained within the sample reservoir, the stabilizing agent useful to completely or substantially prevent degradation or inactivation of a diagnostic marker; and
a reagent contained within the detector array, the reagent capable of undergoing a colorimetric reaction or displaying an optically detectable signal and capable of reacting with the diagnostic marker.
2. The microfluidic device of claim 1, further comprising filter capable of separating at least one unwanted component from a body fluid.
3. The microfluidic device of claim 1, further comprising a results display, wherein a result from the detector array is visually detectable.
4. The microfluidic device of claim 1, wherein the results display comprises a plurality of displays capable to visualizing a plurality of detected diagnostic markers.
5. The microfluidic device of claim 1, further comprising a control display, wherein the detection of at least one control reagent by the at least one detector is visualized.
6. The microfluidic device of claim 1, wherein the stabilizing agent is selected from the group consisting of a protease inhibitor, a DNase inhibitor, and a RNase inhibitor.
7. The microfluidic device of claim 1, wherein the stabilizing agent is useful to completely or substantially prevent degradation or inactivation of a diagnostic marker selected from the group consisting of a protein, a glycoprotein, a nucleic acid, an enzyme, an enzyme inhibitor, and a metabolite.
8. The microfluidic device of claim 1, wherein the stabilizing agent is useful to completely or substantially prevent degradation or inactivation of a diagnostic marker indicative of a disease.
9. The microfluidic device of claim 8, wherein the disease state is selected from the group consisting of a cancer, a metabolic disease, a cardiac disease, diabetes, a bacterial infection, a viral infection, a fungal infection, and a parasitic infection.
10. The microfluidic device of claim 1, wherein the stabilizing agent is selected from the group consisting of Fixanal® Buffer 6.0, acetic acid, aluminum hydroxide bentonite, aluminum sulfate hydrate, benzoic acid, aluminum potassium sulfate dodecahydrate, caffeine, and 3-tert-butyl-hydroxyanisole, or a combination thereof.
11. A microfluidic device for analyzing the presence or absence of a diagnostic marker to be detected in a body fluid, the microfluidic device comprising:
a sample reservoir having a sample input port in fluid connection with at least one detector array, wherein said detector array comprises at least one detector comprising a reaction chamber having an immobilized capture molecule;
a stabilizing agent useful to completely or substantially prevent degradation or inactivation of a diagnostic marker;
a reagent capable of undergoing a colorimetric reaction or displaying an optically detectable signal and capable of reacting with the diagnostic marker;
a filter capable of separating at least one unwanted component from the body fluid; and
a results display, wherein a result from the detector array is visualized;
wherein the stabilizing agent is selected from the group consisting of Fixanal® Buffer 6.0, acetic acid, aluminum hydroxide bentonite, aluminum sulfate hydrate, benzoic acid, aluminum potassium sulfate dodecahydrate, caffeine, and 3-tert-butyl-hydroxyanisole, or a combination thereof.
12. The microfluidic device of claim 11, further comprising a control display, wherein the detection of at least one control reagent by the at least one detector is visually detectable.
13. The microfluidic device of claim 11, wherein the diagnostic marker selected from the group consisting of a protein, a glycoprotein, a nucleic acid, an enzyme, an enzyme inhibitor, and a metabolite.
US12/906,168 2010-02-10 2010-10-18 Devices and systems for biomarker detection Abandoned US20110195491A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US12/906,168 US20110195491A1 (en) 2010-02-10 2010-10-18 Devices and systems for biomarker detection
PCT/US2011/031935 WO2011127467A1 (en) 2010-04-09 2011-04-11 Devices, systems, and methods for biomarker stabilization
US13/911,784 US20140155282A1 (en) 2010-02-10 2013-06-06 Methods of determining stabilization compounds for predictive biomarkers

Applications Claiming Priority (15)

Application Number Priority Date Filing Date Title
US30316510P 2010-02-10 2010-02-10
US32276810P 2010-04-09 2010-04-09
US36498210P 2010-07-16 2010-07-16
US36496910P 2010-07-16 2010-07-16
US36496410P 2010-07-16 2010-07-16
US36497510P 2010-07-16 2010-07-16
US36517910P 2010-07-16 2010-07-16
US36497810P 2010-07-16 2010-07-16
US37361910P 2010-08-13 2010-08-13
US37896010P 2010-09-01 2010-09-01
US37959810P 2010-09-02 2010-09-02
US38340110P 2010-09-16 2010-09-16
US38534710P 2010-09-22 2010-09-22
US39000210P 2010-10-05 2010-10-05
US12/906,168 US20110195491A1 (en) 2010-02-10 2010-10-18 Devices and systems for biomarker detection

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US12/906,169 Continuation US20110194996A1 (en) 2010-02-10 2010-10-18 Transport container for stabilized liquid samples

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/906,167 Continuation US20110195487A1 (en) 2010-02-10 2010-10-18 Sample container for biomarker maintenance

Publications (1)

Publication Number Publication Date
US20110195491A1 true US20110195491A1 (en) 2011-08-11

Family

ID=44352629

Family Applications (40)

Application Number Title Priority Date Filing Date
US12/906,225 Abandoned US20110195401A1 (en) 2010-02-10 2010-10-18 Systems and methods for determining food allergies
US12/906,263 Abandoned US20110195403A1 (en) 2010-02-10 2010-10-18 Systems and methods for fertility analysis
US12/906,196 Abandoned US20110195395A1 (en) 2010-02-10 2010-10-18 Rinse composition for the stabilization of diagnostic biomarkers
US12/906,273 Abandoned US20110195404A1 (en) 2010-02-10 2010-10-18 Systems and methods for determining calcium levels
US12/906,221 Abandoned US20110195858A1 (en) 2010-02-10 2010-10-18 Systems and methods of analyzing biomarker levels
US12/906,167 Abandoned US20110195487A1 (en) 2010-02-10 2010-10-18 Sample container for biomarker maintenance
US12/906,199 Abandoned US20110195396A1 (en) 2010-02-10 2010-10-18 Chewable compositions for the stabilization of diagnostic biomarkers
US12/906,223 Expired - Fee Related US8293537B2 (en) 2010-02-10 2010-10-18 Systems and methods for hemoglobin analysis
US12/906,164 Abandoned US20110195851A1 (en) 2010-02-10 2010-10-18 Systems and methods for determining therapeutic potential
US12/906,202 Abandoned US20110195398A1 (en) 2010-02-10 2010-10-18 Systems and methods for detecting insulin resistance biomarkers
US12/906,227 Expired - Fee Related US8426151B2 (en) 2010-02-10 2010-10-18 Systems and methods for the detection of biomarkers
US12/906,204 Expired - Fee Related US8318105B2 (en) 2010-02-10 2010-10-18 Systems and methods for biomarker analysis
US12/906,231 Abandoned US20110195439A1 (en) 2010-02-10 2010-10-18 Systems and methods for determining cardiac conditions
US12/906,255 Abandoned US20110195402A1 (en) 2010-02-10 2010-10-18 Systems and methods for detecting drug use
US12/906,253 Abandoned US20110195424A1 (en) 2010-02-10 2010-10-18 Systems and methods for diagnosing fungal infections
US12/906,168 Abandoned US20110195491A1 (en) 2010-02-10 2010-10-18 Devices and systems for biomarker detection
US12/906,235 Expired - Fee Related US8318641B2 (en) 2010-02-10 2010-10-18 Systems and methods for the detection of biomarkers
US12/906,162 Expired - Fee Related US8574856B2 (en) 2010-02-10 2010-10-18 Methods for determining the efficiency of a therapeutic
US12/906,201 Abandoned US20110195397A1 (en) 2010-02-10 2010-10-18 Drink mixture for the stabilization of diagnostic biomarkers
US12/906,224 Abandoned US20110195400A1 (en) 2010-02-10 2010-10-18 Systems and methods for diagnosing cancer
US12/906,245 Abandoned US20110195394A1 (en) 2010-02-10 2010-10-18 Systems and methods for diagnosing viral infections
US12/906,236 Abandoned US20110195440A1 (en) 2010-02-10 2010-10-18 Systems and methods for identifying allergies
US12/906,211 Abandoned US20110195857A1 (en) 2010-02-10 2010-10-18 Systems for analyzing stabilized biomarkers
US12/906,157 Abandoned US20110195855A1 (en) 2010-02-10 2010-10-18 Systems and methods for improving biomarker availability
US12/906,166 Abandoned US20110196085A1 (en) 2010-02-10 2010-10-18 Biomarker stabilizing agents
US12/906,169 Abandoned US20110194996A1 (en) 2010-02-10 2010-10-18 Transport container for stabilized liquid samples
US12/906,268 Abandoned US20110195427A1 (en) 2010-02-10 2010-10-18 Systems and methods for detecting pregnancy
US12/906,161 Abandoned US20110195856A1 (en) 2010-02-10 2010-10-18 Methods of determining stabilization compounds for predictive biomarkers
US12/906,260 Abandoned US20110195425A1 (en) 2010-02-10 2010-10-18 Systems and methods for diagnosing parasite infections
US12/906,208 Abandoned US20110195399A1 (en) 2010-02-10 2010-10-18 Systems and methods for the inhibition of galactose oxidase
US12/906,173 Abandoned US20110192239A1 (en) 2010-02-10 2010-10-18 Devices for collection and stabilization of biomarkers in liquid samples
US12/906,239 Abandoned US20110195423A1 (en) 2010-02-10 2010-10-18 Systems and methods for diagnosing bacterial infections
US12/906,159 Expired - Fee Related US8501669B2 (en) 2010-02-10 2010-10-18 Systems and methods of screening biomarkers in bodily fluids
US12/906,158 Abandoned US20110195421A1 (en) 2010-02-10 2010-10-18 Methods of identifying biomarkers for cancer
US12/906,171 Abandoned US20110195488A1 (en) 2010-02-10 2010-10-18 Devices for collection and stabilization of biomarkers in liquid samples
US12/906,170 Abandoned US20110195872A1 (en) 2010-02-10 2010-10-18 Devices and systems for biomarker detection
US13/658,554 Abandoned US20130157373A1 (en) 2010-02-10 2012-10-23 Systems and methods for hemoglobin analysis
US13/686,345 Abandoned US20130184186A1 (en) 2010-02-10 2012-11-27 Systems and methods for the detection of biomarkers
US13/686,314 Abandoned US20130184187A1 (en) 2010-02-10 2012-11-27 Systems and methods for biomarker analysis
US13/911,784 Abandoned US20140155282A1 (en) 2010-02-10 2013-06-06 Methods of determining stabilization compounds for predictive biomarkers

Family Applications Before (15)

Application Number Title Priority Date Filing Date
US12/906,225 Abandoned US20110195401A1 (en) 2010-02-10 2010-10-18 Systems and methods for determining food allergies
US12/906,263 Abandoned US20110195403A1 (en) 2010-02-10 2010-10-18 Systems and methods for fertility analysis
US12/906,196 Abandoned US20110195395A1 (en) 2010-02-10 2010-10-18 Rinse composition for the stabilization of diagnostic biomarkers
US12/906,273 Abandoned US20110195404A1 (en) 2010-02-10 2010-10-18 Systems and methods for determining calcium levels
US12/906,221 Abandoned US20110195858A1 (en) 2010-02-10 2010-10-18 Systems and methods of analyzing biomarker levels
US12/906,167 Abandoned US20110195487A1 (en) 2010-02-10 2010-10-18 Sample container for biomarker maintenance
US12/906,199 Abandoned US20110195396A1 (en) 2010-02-10 2010-10-18 Chewable compositions for the stabilization of diagnostic biomarkers
US12/906,223 Expired - Fee Related US8293537B2 (en) 2010-02-10 2010-10-18 Systems and methods for hemoglobin analysis
US12/906,164 Abandoned US20110195851A1 (en) 2010-02-10 2010-10-18 Systems and methods for determining therapeutic potential
US12/906,202 Abandoned US20110195398A1 (en) 2010-02-10 2010-10-18 Systems and methods for detecting insulin resistance biomarkers
US12/906,227 Expired - Fee Related US8426151B2 (en) 2010-02-10 2010-10-18 Systems and methods for the detection of biomarkers
US12/906,204 Expired - Fee Related US8318105B2 (en) 2010-02-10 2010-10-18 Systems and methods for biomarker analysis
US12/906,231 Abandoned US20110195439A1 (en) 2010-02-10 2010-10-18 Systems and methods for determining cardiac conditions
US12/906,255 Abandoned US20110195402A1 (en) 2010-02-10 2010-10-18 Systems and methods for detecting drug use
US12/906,253 Abandoned US20110195424A1 (en) 2010-02-10 2010-10-18 Systems and methods for diagnosing fungal infections

Family Applications After (24)

Application Number Title Priority Date Filing Date
US12/906,235 Expired - Fee Related US8318641B2 (en) 2010-02-10 2010-10-18 Systems and methods for the detection of biomarkers
US12/906,162 Expired - Fee Related US8574856B2 (en) 2010-02-10 2010-10-18 Methods for determining the efficiency of a therapeutic
US12/906,201 Abandoned US20110195397A1 (en) 2010-02-10 2010-10-18 Drink mixture for the stabilization of diagnostic biomarkers
US12/906,224 Abandoned US20110195400A1 (en) 2010-02-10 2010-10-18 Systems and methods for diagnosing cancer
US12/906,245 Abandoned US20110195394A1 (en) 2010-02-10 2010-10-18 Systems and methods for diagnosing viral infections
US12/906,236 Abandoned US20110195440A1 (en) 2010-02-10 2010-10-18 Systems and methods for identifying allergies
US12/906,211 Abandoned US20110195857A1 (en) 2010-02-10 2010-10-18 Systems for analyzing stabilized biomarkers
US12/906,157 Abandoned US20110195855A1 (en) 2010-02-10 2010-10-18 Systems and methods for improving biomarker availability
US12/906,166 Abandoned US20110196085A1 (en) 2010-02-10 2010-10-18 Biomarker stabilizing agents
US12/906,169 Abandoned US20110194996A1 (en) 2010-02-10 2010-10-18 Transport container for stabilized liquid samples
US12/906,268 Abandoned US20110195427A1 (en) 2010-02-10 2010-10-18 Systems and methods for detecting pregnancy
US12/906,161 Abandoned US20110195856A1 (en) 2010-02-10 2010-10-18 Methods of determining stabilization compounds for predictive biomarkers
US12/906,260 Abandoned US20110195425A1 (en) 2010-02-10 2010-10-18 Systems and methods for diagnosing parasite infections
US12/906,208 Abandoned US20110195399A1 (en) 2010-02-10 2010-10-18 Systems and methods for the inhibition of galactose oxidase
US12/906,173 Abandoned US20110192239A1 (en) 2010-02-10 2010-10-18 Devices for collection and stabilization of biomarkers in liquid samples
US12/906,239 Abandoned US20110195423A1 (en) 2010-02-10 2010-10-18 Systems and methods for diagnosing bacterial infections
US12/906,159 Expired - Fee Related US8501669B2 (en) 2010-02-10 2010-10-18 Systems and methods of screening biomarkers in bodily fluids
US12/906,158 Abandoned US20110195421A1 (en) 2010-02-10 2010-10-18 Methods of identifying biomarkers for cancer
US12/906,171 Abandoned US20110195488A1 (en) 2010-02-10 2010-10-18 Devices for collection and stabilization of biomarkers in liquid samples
US12/906,170 Abandoned US20110195872A1 (en) 2010-02-10 2010-10-18 Devices and systems for biomarker detection
US13/658,554 Abandoned US20130157373A1 (en) 2010-02-10 2012-10-23 Systems and methods for hemoglobin analysis
US13/686,345 Abandoned US20130184186A1 (en) 2010-02-10 2012-11-27 Systems and methods for the detection of biomarkers
US13/686,314 Abandoned US20130184187A1 (en) 2010-02-10 2012-11-27 Systems and methods for biomarker analysis
US13/911,784 Abandoned US20140155282A1 (en) 2010-02-10 2013-06-06 Methods of determining stabilization compounds for predictive biomarkers

Country Status (1)

Country Link
US (40) US20110195401A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013184830A1 (en) * 2012-06-07 2013-12-12 Pioma Inc. 5-ala for detection of brain tumors
US8815508B2 (en) 2008-08-12 2014-08-26 Zinfandel Pharmaceuticals, Inc. Method of identifying disease risk factors
US8846315B2 (en) 2008-08-12 2014-09-30 Zinfandel Pharmaceuticals, Inc. Disease risk factors and methods of use
US9102666B2 (en) 2011-01-10 2015-08-11 Zinfandel Pharmaceuticals, Inc. Methods and drug products for treating Alzheimer's disease
US10175254B2 (en) 2013-07-16 2019-01-08 Palo Alto Health Sciences, Inc. Methods and systems for quantitative colorimetric capnometry

Families Citing this family (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080124355A1 (en) 2006-09-22 2008-05-29 David Gordon Bermudes Live bacterial vaccines for viral infection prophylaxis or treatment
US8241623B1 (en) 2009-02-09 2012-08-14 David Bermudes Protease sensitivity expression system
US9597379B1 (en) 2010-02-09 2017-03-21 David Gordon Bermudes Protease inhibitor combination with therapeutic proteins including antibodies
US8771669B1 (en) 2010-02-09 2014-07-08 David Gordon Bermudes Immunization and/or treatment of parasites and infectious agents by live bacteria
US8524220B1 (en) 2010-02-09 2013-09-03 David Gordon Bermudes Protease inhibitor: protease sensitivity expression system composition and methods improving the therapeutic activity and specificity of proteins delivered by bacteria
US20110195401A1 (en) * 2010-02-10 2011-08-11 Selinfreund Richard H Systems and methods for determining food allergies
US20120164674A1 (en) * 2010-10-28 2012-06-28 Selinfreund Richard H Devices and washes for biomarker stabilization
WO2012116074A1 (en) * 2011-02-22 2012-08-30 Mayo Foundation For Medical Education And Research Biomarkers of insulin sensitivity
US9488613B2 (en) * 2011-03-02 2016-11-08 Massachusetts Institute Of Technology Systems, devices and methods for multiplexed diagnostics
EP2748593B1 (en) * 2011-08-22 2017-05-24 Smiths Heimann GmbH Liquid mixture used to test and validate test devices
AU2012310328A1 (en) 2011-09-23 2014-04-10 Technophage, Investigação E Desenvolvimento Em Biotecnologia, S.A. Anti-Tumor Necrosis Factor-alpha agents and uses thereof
CA2849705A1 (en) 2011-09-23 2013-03-28 Technophage, Investigacao E Desenvolvimento Em Biotecnologia, Sa Modified albumin-binding domains and uses thereof to improve pharmacokinetics
EP2650314A1 (en) 2012-04-13 2013-10-16 Clariant International Ltd. Process for inhibition of sulphide scales
RU2493566C1 (en) * 2012-05-25 2013-09-20 Открытое акционерное общество "Медицина" (ОАО "Медицина") Method for screening diagnosis of insulin resistance
US9804149B2 (en) * 2012-10-10 2017-10-31 Bio-Rad Laboratories, Inc. Patient-based results display
WO2014062819A1 (en) * 2012-10-16 2014-04-24 Companion Diagnostics Inc. Compositions and methods for improving diagnosis
JP6116268B2 (en) * 2013-01-31 2017-04-19 デンカ生研株式会社 Methods for suppressing false negatives in biological mucosa-derived specimen measurement immunoassays
AU2014215330B2 (en) * 2013-02-06 2017-02-02 Fujirebio Inc. Vitamin D measurement method and measurement kit
US9593339B1 (en) 2013-02-14 2017-03-14 David Gordon Bermudes Bacteria carrying bacteriophage and protease inhibitors for the treatment of disorders and methods of treatment
WO2014164472A1 (en) 2013-03-10 2014-10-09 The Board Of Regents Of The University Of Texas System Biomarkers for chagas disease related cardiomyopathy
CN103513038B (en) * 2013-05-16 2015-06-10 武汉生之源生物科技有限公司 Detection kit for galectin-3 and preparation method thereof
JP6539274B2 (en) 2013-08-12 2019-07-03 ファーマシューティカル マニュファクチュアリング リサーチ サービシズ,インコーポレーテッド Extruded immediate release abuse deterrent pills
CN104422768A (en) 2013-08-30 2015-03-18 广州瑞博奥生物科技有限公司 Antibody chip kit for joint detection of early ovarian cancer markers
US10597696B2 (en) * 2013-09-18 2020-03-24 University Of Notre Dame Du Lac Detection of Niemann-Pick disease comprising detection of lysozyme and cathepsins
US10172797B2 (en) 2013-12-17 2019-01-08 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9492444B2 (en) 2013-12-17 2016-11-15 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
GB201402036D0 (en) * 2014-02-06 2014-03-26 Nobel Biocare Services Ag Method for determination of the status of a disease
JP6632999B2 (en) 2014-06-12 2020-01-22 ユニバーシティ オブ ノートルダム デュ ラック Compositions and methods for treating neurological diseases and cerebral injury
ES2809458T3 (en) 2014-07-17 2021-03-04 Pharmaceutical Manufacturing Res Services Inc Liquid filled, abuse deterrent and immediate release dosage form
CN106796212A (en) 2014-08-12 2017-05-31 新生代吉恩公司 System and method for monitoring health based on the body fluid collected
AU2015336065A1 (en) 2014-10-20 2017-05-04 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form
JP6914192B2 (en) 2014-10-31 2021-08-04 マサチューセッツ インスティテュート オブ テクノロジー Composition and method for emulsion formation
KR20160081669A (en) 2014-12-31 2016-07-08 삼성전자주식회사 Reaction apparatus, test device and control method for the test device
US9662276B2 (en) * 2015-06-16 2017-05-30 President And Fellows Of Harvard College Methodology of dental caries detection
US10753952B2 (en) * 2015-06-19 2020-08-25 Amy J. Reisinger Rapid and sensitive method of forensic toxicology in post-mortem subjects and in live and post-mortem animals using oral fluid testing
GB2547062B (en) 2015-09-09 2020-03-04 Drawbridge Health Inc Systems, methods, and devices for sample collection, stabilization and preservation
KR101949380B1 (en) * 2016-03-31 2019-02-19 (주) 인텍바이오 Ultrasensitive biomarker quantification method using photooxidation-induced amplification
EP3442706A4 (en) 2016-04-13 2020-02-19 NextGen Jane, Inc. Sample collection and preservation devices, systems and methods
US11670409B2 (en) 2016-04-15 2023-06-06 Takeda Pharmaceutical Company Limited Method and apparatus for providing a pharmacokinetic drug dosing regiment
BR112018072432A2 (en) * 2016-05-19 2019-02-19 Unilever Nv oral care composition and method for reducing tooth sensitivity and / or remineralization
CA3018289A1 (en) * 2016-05-24 2017-11-30 Excision Biotherapeutics, Inc. Metabalomics and viral diagnostics suite
RU2630974C1 (en) * 2016-08-05 2017-09-15 Автономная некоммерческая организация "Институт молекулярной диагностики" (АНО "ИнМоДи") Conjugate of antitumour drugs with recombinant alpha-fetoprotein and its functional fragments and method of their producing
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria
US11612888B2 (en) 2017-01-04 2023-03-28 The Research Foundation For The State University Of New York Biomarker detection device
WO2018132515A1 (en) 2017-01-10 2018-07-19 Drawbridge Health, Inc. Devices, systems, and methods for sample collection
US10896749B2 (en) 2017-01-27 2021-01-19 Shire Human Genetic Therapies, Inc. Drug monitoring tool
US10509043B2 (en) * 2017-04-18 2019-12-17 Amir Mor Methods for identification of pregnancy failure
US11105816B2 (en) 2017-04-18 2021-08-31 Amir Mor Methods for identification of pregnancy failure
CN106932572A (en) * 2017-04-26 2017-07-07 深圳市博卡生物技术有限公司 For the kit and its detection method of infertile antibody test
WO2019094950A1 (en) * 2017-11-13 2019-05-16 Cornell University A multiplexed diagnostic assay for iron and vitamin a deficiency and methods of use thereof
WO2019119087A1 (en) * 2017-12-20 2019-06-27 Fundação Universidade Estadual Do Ceará - Funece Kit and method for detecting immunoglobulin e and immunoglobulin g1
CN108344866B (en) * 2018-01-12 2020-07-28 天津大学 Micro-fluidic chip detection system and method for detecting sample based on same
WO2019190787A1 (en) * 2018-03-27 2019-10-03 Exact Sciences Corporation Method for stabilizing hemoglobin and reagents for performing the same
CN109297924B (en) * 2018-11-16 2020-06-05 禚元华 Multi-functional intelligent gynecological secretion inspection device
CN110051936B (en) * 2019-04-28 2021-05-07 王燕 Paediatrics clinician is with paediatrics jaundice treatment device
CN111530273B (en) * 2020-05-07 2022-05-31 广东绿羽环保科技有限公司 Hydrolysis catalytic adsorption ball for high-temperature waste gas treatment
JP2023541044A (en) * 2020-09-11 2023-09-27 ベクトン・ディキンソン・アンド・カンパニー Environmental sample test device
WO2022096486A1 (en) * 2020-11-03 2022-05-12 Ideogen Ag Methods of diagnosing and/or prognosing a disease in tears
EP3992631A1 (en) * 2020-11-03 2022-05-04 Ideogen AG Methods of diagnosing and/or prognosing a disease in tear sample
US20220364973A1 (en) * 2021-05-13 2022-11-17 Honeywell International Inc. In situ fluid sampling device and method of using the same
US20220403372A1 (en) * 2021-06-18 2022-12-22 Luminex Corporation Method for extraction of cell-free dna
CN113933131B (en) * 2021-09-24 2024-01-26 合肥天一生物技术研究所有限责任公司 Vaginal microorganism fluorescent staining solution
WO2023172575A2 (en) * 2022-03-08 2023-09-14 Aeena Dx, Inc. Methods for disease detection

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5532170A (en) * 1991-03-09 1996-07-02 Fisons Plc Processing analytical reagents
US6586259B1 (en) * 1999-11-15 2003-07-01 Pharmanetics Incorporated Platelet/leukocyte interaction assay and reagent therefor
US6632656B1 (en) * 1998-04-27 2003-10-14 Gyros Ab Microfabricated apparatus for cell based assays
US20070065893A1 (en) * 2005-09-19 2007-03-22 Carol Carte Liquid-phase galactose oxidase-schiff's assay
US20070248533A1 (en) * 2004-06-17 2007-10-25 Kuperus John H Stabilized and Lyophilized Radiopharmaceutical Agents For Destroying Tumors
US20080248493A1 (en) * 2007-04-09 2008-10-09 Mattingly Phillip G Acridinium phenyl esters useful in the analysis of biological samples
US20090098533A1 (en) * 2003-10-06 2009-04-16 Marc Munnes Methods and kits for investigating cancer
US20090111121A1 (en) * 2005-10-14 2009-04-30 Christine Des Rosiers Method for detecting a biomarker of oxidative stress in a biological sample
US20100092978A1 (en) * 2008-10-09 2010-04-15 The University Of Hong Kong Cadherin-17 as diagnostic marker and therapeutic target for liver cancer
US7732490B2 (en) * 2002-03-04 2010-06-08 Merck Hdac Research, Llc Methods of treating cancer

Family Cites Families (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL56342A (en) * 1978-12-29 1982-05-31 Zer Tamar Method and means for determining human chorionic gonadotropin in urine
US4562043A (en) * 1983-09-09 1985-12-31 Mennen Frederick C Self-contained swab cartridge apparatus for detecting occult blood
DE3603053A1 (en) * 1986-01-30 1987-08-06 Schering Ag PREGNANCY DETECTION WITH EPF
JPS6336143A (en) * 1986-07-30 1988-02-16 Tosoh Corp Method and instrument for measuring stable type saccharogenic hemoglobin
US5169789A (en) * 1986-12-03 1992-12-08 New Horizons Diagnostics Corporation Device and method for self contained solid phase immunodiffusion assay
US5162229A (en) * 1988-03-15 1992-11-10 Akzo N.V. Device and method for enhanced recovery and detection of microbial growth in the presence of antimicrobial substances
US5022409A (en) * 1989-09-21 1991-06-11 Epitope, Inc. Oral rinse immunoglobulin collection kit for immunoassay and method thereof
CA2102526A1 (en) * 1991-06-19 1992-12-20 Michael D. Fiechtner Rapid determination of glycated hemoglobin
US5998216A (en) * 1996-10-01 1999-12-07 Beth Israel Deaconess Medical Center Stabilizing formulation for preserving the integrity of proteins present in a body fluid sampled ex-vivo for evaluation as a specimen
US20030008816A1 (en) * 1997-05-28 2003-01-09 Pilon Aprile L. Methods and compositions for the treatment of fibrotic conditions & impaired lung function & to enhance lymphocyte production
US6043221A (en) * 1997-07-30 2000-03-28 Amgen Inc. Method for preventing and treating hearing loss using a neuturin protein product
US6194222B1 (en) * 1998-01-05 2001-02-27 Biosite Diagnostics, Inc. Methods for monitoring the status of assays and immunoassays
WO1999050442A1 (en) * 1998-03-30 1999-10-07 Friedrich Srienc A flow injection flow cytometry system for on-line monitoring of bioreactors and method for monitoring
GB2339615B (en) * 1998-07-14 2001-02-07 Cozart Bioscience Ltd Screening device and method of screening an immunoassay test
AUPP915799A0 (en) * 1999-03-11 1999-04-15 Enterix Inc. Sample collection and testing system
US6556923B2 (en) * 2000-01-26 2003-04-29 Caliper Technologies Corp. Software for high throughput microfluidic systems
US6255047B1 (en) * 2000-02-28 2001-07-03 Bio-Rad Laboratories, Inc. Biosynthetic carbohydrate-deficient transferrin references
US7465540B2 (en) * 2000-09-21 2008-12-16 Luminex Corporation Multiple reporter read-out for bioassays
CA2428864C (en) * 2000-11-08 2011-04-12 Becton, Dickinson And Company Method and device for collecting and stabilizing a biological sample
EP1339318B1 (en) * 2000-11-13 2009-10-14 Ensisheim Partners LLC Device for collecting, handling and processing mammary fluid samples for evaluating breast diseases, including cancer
JP4651868B2 (en) * 2001-03-28 2011-03-16 株式会社ジーシー Saliva pretreatment kit and saliva pretreatment method using the same
US20040176476A1 (en) * 2001-06-22 2004-09-09 Gyurik Robert J. Pharmaceutical composition
WO2003007814A1 (en) * 2001-07-18 2003-01-30 Agilex Biosciences, Inc. Device and method for collecting, transporting and recovering low molecular weight analytes in saliva
EP1438583B1 (en) * 2001-09-20 2009-09-16 Board of Regents, The University of Texas System Measuring circulating therapeutic antibody, antigen and antigen/antibody complexes using elisa assays
AU2003225254A1 (en) * 2002-04-30 2003-11-17 Biowhittaker Technologies, Inc. Automated sequential injection analysis systems for the determination of trace endotoxin levels
CA2483958A1 (en) * 2002-05-02 2003-11-13 Aspenbio, Inc. Pregnancy detection
CN102353569A (en) * 2002-05-13 2012-02-15 贝克顿·迪金森公司 Protease inhibitor sample collection system
EP1551376A4 (en) * 2002-08-12 2010-10-06 Dynavax Tech Corp Immunomodulatory compositions, methods of making, and methods of use thereof
US10966421B2 (en) * 2002-10-16 2021-04-06 Streck, Inc. Method and device for collecting and preserving cells for analysis
US20040081970A1 (en) * 2002-10-28 2004-04-29 Athersys, Inc. Calcium-sensing receptor 2 (CaR2) and methods for using
JP4462964B2 (en) * 2003-03-10 2010-05-12 第一三共株式会社 Antibodies targeting cancer-specific antigens
US20050158303A1 (en) * 2003-04-04 2005-07-21 Genentech, Inc. Methods of treating IgE-mediated disorders comprising the administration of high concentration anti-IgE antibody formulations
JP3910156B2 (en) * 2003-05-28 2007-04-25 アークレイ株式会社 Sample stabilization method
US6991898B2 (en) * 2003-10-20 2006-01-31 Kimberly-Clark Worldwide, Inc. Diagnostic test device and method of using same
WO2005055812A2 (en) * 2003-12-05 2005-06-23 Ciphergen Biosystems, Inc. Serum biomarkers for chagas disease
US20050175977A1 (en) * 2004-02-10 2005-08-11 Posner Alan H. Hemoglobin isolation and preparation of glycosylated hemoglobin
US20060099567A1 (en) * 2004-04-08 2006-05-11 Biomatrica, Inc. Integration of sample storage and sample management for life science
EP1742616B1 (en) * 2004-04-30 2014-10-01 Abraxis BioScience, LLC Sustained-release microspheres and methods of making and using same
US7678578B2 (en) * 2005-02-07 2010-03-16 Beckman Coulter, Inc. Cell permeabilization and stabilization reagent and method of use
TW200700727A (en) * 2005-03-23 2007-01-01 Astrazeneca Ab Screen
US20060281143A1 (en) * 2005-04-01 2006-12-14 Msp Corporation Method and apparatus for automatic cell and biological sample preparation and detection
US20060246513A1 (en) * 2005-05-02 2006-11-02 Bohannon Robert C Method and device to detect the presence of analytes in a sample
US7521577B2 (en) * 2006-01-12 2009-04-21 Molecular Probes, Inc. Heavy metal binding compounds and their method of use
US7634844B2 (en) * 2006-04-19 2009-12-22 Newfrey Llc Adjustable prewinder assembly for wire insert installation tool
US9101161B2 (en) * 2006-11-02 2015-08-11 The Coca-Cola Company High-potency sweetener composition with phytoestrogen and compositions sweetened therewith
EP2074422A4 (en) * 2006-11-13 2010-02-17 Life Technologies Corp Methods and kits for detecting prostate cancer biomarkers
WO2008147847A1 (en) * 2007-05-22 2008-12-04 Baylor College Of Medicine Immune complex vaccination as a strategy to enhance immunity in the elderly and other immune compromised populations
WO2009041577A1 (en) * 2007-09-27 2009-04-02 Niigata University Urine pretreatment agent for urinary protein determination, urine pretreatment method, and urinary protein determination method
JP5422108B2 (en) * 2007-10-16 2014-02-19 栄研化学株式会社 Heme protein stabilization method and storage solution
US20110195401A1 (en) * 2010-02-10 2011-08-11 Selinfreund Richard H Systems and methods for determining food allergies

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5532170A (en) * 1991-03-09 1996-07-02 Fisons Plc Processing analytical reagents
US6632656B1 (en) * 1998-04-27 2003-10-14 Gyros Ab Microfabricated apparatus for cell based assays
US6586259B1 (en) * 1999-11-15 2003-07-01 Pharmanetics Incorporated Platelet/leukocyte interaction assay and reagent therefor
US7732490B2 (en) * 2002-03-04 2010-06-08 Merck Hdac Research, Llc Methods of treating cancer
US20090098533A1 (en) * 2003-10-06 2009-04-16 Marc Munnes Methods and kits for investigating cancer
US20070248533A1 (en) * 2004-06-17 2007-10-25 Kuperus John H Stabilized and Lyophilized Radiopharmaceutical Agents For Destroying Tumors
US20070065893A1 (en) * 2005-09-19 2007-03-22 Carol Carte Liquid-phase galactose oxidase-schiff's assay
US20090111121A1 (en) * 2005-10-14 2009-04-30 Christine Des Rosiers Method for detecting a biomarker of oxidative stress in a biological sample
US20080248493A1 (en) * 2007-04-09 2008-10-09 Mattingly Phillip G Acridinium phenyl esters useful in the analysis of biological samples
US20100092978A1 (en) * 2008-10-09 2010-04-15 The University Of Hong Kong Cadherin-17 as diagnostic marker and therapeutic target for liver cancer

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8815508B2 (en) 2008-08-12 2014-08-26 Zinfandel Pharmaceuticals, Inc. Method of identifying disease risk factors
US8846315B2 (en) 2008-08-12 2014-09-30 Zinfandel Pharmaceuticals, Inc. Disease risk factors and methods of use
US10865449B2 (en) 2008-08-12 2020-12-15 Zinfandel Pharmaceuticals, Inc. Method of identifying disease risk factors
US11021751B2 (en) 2008-08-12 2021-06-01 Zinfandel Pharmaceuticals, Inc. Disease risk factors and methods of use
US9102666B2 (en) 2011-01-10 2015-08-11 Zinfandel Pharmaceuticals, Inc. Methods and drug products for treating Alzheimer's disease
US9724339B2 (en) 2011-01-10 2017-08-08 Zinfandel Pharmaceuticals, Inc. Methods and drug products for treating alzheimer's disease
US11179375B2 (en) 2011-01-10 2021-11-23 Zinfandel Pharmaceuticals, Inc. Methods and drug products for treating Alzheimer's disease
WO2013184830A1 (en) * 2012-06-07 2013-12-12 Pioma Inc. 5-ala for detection of brain tumors
US9493810B2 (en) 2012-06-07 2016-11-15 Pioma, Inc. 5-ALA for detection of brain tumors
US11307204B2 (en) 2012-06-07 2022-04-19 Pioma, Inc. 5-ALA for detection of brain tumors
US10175254B2 (en) 2013-07-16 2019-01-08 Palo Alto Health Sciences, Inc. Methods and systems for quantitative colorimetric capnometry
US11538569B2 (en) 2013-07-16 2022-12-27 Freespira. Inc. Methods and systems for quantitative colorimetric capnometry

Also Published As

Publication number Publication date
US8426151B2 (en) 2013-04-23
US8318105B2 (en) 2012-11-27
US20110195857A1 (en) 2011-08-11
US20110195394A1 (en) 2011-08-11
US20110195404A1 (en) 2011-08-11
US20110195872A1 (en) 2011-08-11
US20110195874A1 (en) 2011-08-11
US20130184187A1 (en) 2013-07-18
US20110195402A1 (en) 2011-08-11
US20110195488A1 (en) 2011-08-11
US20110195421A1 (en) 2011-08-11
US20110195873A1 (en) 2011-08-11
US8574856B2 (en) 2013-11-05
US20110195396A1 (en) 2011-08-11
US20110195403A1 (en) 2011-08-11
US8293537B2 (en) 2012-10-23
US20110195397A1 (en) 2011-08-11
US20110195487A1 (en) 2011-08-11
US20130157373A1 (en) 2013-06-20
US20140155282A1 (en) 2014-06-05
US20110195423A1 (en) 2011-08-11
US20110195508A1 (en) 2011-08-11
US20110196085A1 (en) 2011-08-11
US20110195422A1 (en) 2011-08-11
US20110195424A1 (en) 2011-08-11
US20110195401A1 (en) 2011-08-11
US20110195395A1 (en) 2011-08-11
US20110195871A1 (en) 2011-08-11
US20110195398A1 (en) 2011-08-11
US20110195439A1 (en) 2011-08-11
US20110195856A1 (en) 2011-08-11
US20110195400A1 (en) 2011-08-11
US20110195425A1 (en) 2011-08-11
US20110192239A1 (en) 2011-08-11
US20130184186A1 (en) 2013-07-18
US20110195440A1 (en) 2011-08-11
US20110194996A1 (en) 2011-08-11
US20110195859A1 (en) 2011-08-11
US20110195399A1 (en) 2011-08-11
US20110195855A1 (en) 2011-08-11
US20110195851A1 (en) 2011-08-11
US8501669B2 (en) 2013-08-06
US20110195858A1 (en) 2011-08-11
US8318641B2 (en) 2012-11-27
US20110195427A1 (en) 2011-08-11

Similar Documents

Publication Publication Date Title
US8318105B2 (en) Systems and methods for biomarker analysis
WO2011127467A1 (en) Devices, systems, and methods for biomarker stabilization
US20120164674A1 (en) Devices and washes for biomarker stabilization
WO2012088330A2 (en) Devices, systems, and methods for fecal analysis

Legal Events

Date Code Title Description
AS Assignment

Owner name: COMPANION DIAGNOSTICS INC., INDIANA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SELINFREUND, RICHARD H.;VIG, RAKESH;GILL, RICHARD P.;SIGNING DATES FROM 20121023 TO 20121110;REEL/FRAME:029890/0026

AS Assignment

Owner name: BMO HARRIS BANK N.A., ILLINOIS

Free format text: SECURITY INTEREST;ASSIGNOR:COMPANION DIAGNOSTICS, INC.;REEL/FRAME:032597/0400

Effective date: 20120723

STCB Information on status: application discontinuation

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