WO2012088330A2 - Devices, systems, and methods for fecal analysis - Google Patents

Devices, systems, and methods for fecal analysis Download PDF

Info

Publication number
WO2012088330A2
WO2012088330A2 PCT/US2011/066616 US2011066616W WO2012088330A2 WO 2012088330 A2 WO2012088330 A2 WO 2012088330A2 US 2011066616 W US2011066616 W US 2011066616W WO 2012088330 A2 WO2012088330 A2 WO 2012088330A2
Authority
WO
WIPO (PCT)
Prior art keywords
stabilizing agent
diagnostic marker
diagnostic
cancer
acid
Prior art date
Application number
PCT/US2011/066616
Other languages
French (fr)
Other versions
WO2012088330A3 (en
Inventor
Rakesh Vig
Richard H. Selinfreund
Richard P. Gill
Original Assignee
Companion Diagnostics Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Companion Diagnostics Inc. filed Critical Companion Diagnostics Inc.
Publication of WO2012088330A2 publication Critical patent/WO2012088330A2/en
Publication of WO2012088330A3 publication Critical patent/WO2012088330A3/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57419Specifically defined cancers of colon

Definitions

  • Colorectal cancer also called colon cancer or large bowel cancer, includes cancerous growths in the colon, rectum and appendix. With 655,000 deaths worldwide per year, it is the fourth most common form of cancer in the United States and the third leading cause of cancer- related death in the Western world (National Cancer Institute, 2009. http://www.cancer.gov/cancertopics/commoncancers.). Colorectal cancers arise from adenomatous polyps in the colon. These mushroom-shaped growths are usually benign, but some develop into cancer over time. Localized colon cancer is usually diagnosed through colonoscopy.
  • a colonoscopy a long, flexible tube (colonoscope) is inserted into the rectum.
  • a tiny video camera at the tip of the lube allows the doctor to view the inside of the entire colon.
  • polyps or other types of abnormal tissue can be removed through the scope during a colonoscopy.
  • Tissue samples can be taken during a colonoscopy as well. Preparation of the colon to allow for the visualization of abnormal conditions involves the use of laxitives or other processes to cleanse the colon. Because of this invasive nature of the procedure and the costs associated with the procedure, there is a need for an improved method of diagnosis.
  • Fig. 1 shows a perspective view of a device for fecal analysis, according to at least one embodiment of the present disclosure
  • Fig. 2 shows a perspective view of a device for fecal analysis, according to at least one embodiment of the present d isclosure:
  • Fig. 3 shows a flowchart indicating the steps of a method for fecal analysis, according to at least one embodiment of the present disclosure.
  • the disclosure of the present application provides various devices, sy stems, 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 or a device used to test 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, scrum and mixtures of thereof.
  • Body fluids may also comprise cancer cells, peripheral blood mononuclear cells, lymphocytes, lymph fluid, and other tissue secretions or fluid.
  • the device 100 is comprised of a chamber 102 sized and shaped to define an inlet 104, an outlet 106, and a passage 108 fluidly connecting the in let 104 and outlet 1 06.
  • Device 100 may further comprise a diagnostic filter 1 10 positioned in the passage so as to define a introduction lumen 1 12 between the inlet 104 and the diagnostic filter 1 1 0, and an outlet lumen 1 14 between the outlet 1 06 and the diagnostic fi lter 1 10.
  • the device 1 00 may also comprise a stabilizing bar 1 16 coupled to chamber 1 02, where stabilizing bar 1 16 is operable to affix to a stationary apparatus, such as the side of a toilet.
  • device 100 may also comprise a inlet cap 1 18 capable of coupling to the inlet 1 04, and an outlet cap 120 capable of coupling to outlet 1 06.
  • inlet cap 1 1 8 and outlet cap 120 may be coupled to inlet 104 and/or outlet 1 06 respectively to form a fluid- tight seal.
  • Diagnostic filter 1 1 0 in an exemplary embodiment comprises a filter which serves to prevent passage of particulate material below a predetermined size threshold from passing therethrough. Additionally, diagnostic filter 1 10 may also comprise at least one indicator capable of binding or recognizing a diagnostic marker. Such an indicator may also be capable of presenting a visual signal in response to exposure to the diagnostic marker. As shown in Fig. 2,
  • diagnostic filter 1 10 in at least one embodiment may be comprised of more than one component.
  • diagnostic filter 1 10 is comprised of filter 122 and diagnostic membrane 124. Further, diagnostic filter 1 10, in at least one embodiment, may be removable from chamber 102 for futher processing.
  • Device 100 in at least one exemplary embodiment may also comprise a stabilizing agent capable of completely or substantially preventing the degradation or inactivation of a diagnostic marker contained within the bodily fluid passing through device 100.
  • a stabilizing agent may be integral to or coated on diagnostic filter 1 10 or introduction lumen 1 12 of device 100.
  • the stabilizing agent may be any compound which completely or substantially preventing the degradation or inaclivation of a specified diagnostic marker contained within the bodily fluid.
  • the stabilizing agent may be selected from a group consisting of aluminum sulfate hydrate, benzoic acid, aluminum potassium sulfate dodecahydrate, or a combination thereof.
  • the stabilizing agent may comprise more than one stabilizing agent, such as a exemplary stabilizing agent which contains aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate.
  • the diagnostic marker detected in device 1 00 may be selected from the group consisting of Adenomatous polyposis coli (APC), ⁇ -catenin, Wnt, and Cancer Antigen CA 19-9. In some instances, the diagnostic marker may be indicative of a cancer, such as colorectal cancer.
  • APC Adenomatous polyposis coli
  • ⁇ -catenin ⁇ -catenin
  • Wnt Wnt
  • Cancer Antigen CA 19-9 Cancer Antigen CA 19-9.
  • the diagnostic marker may be indicative of a cancer, such as colorectal cancer.
  • device 100 may be used to collect fecal material or urine to determine whether a predetermined diagnostic marker is present. Such a use may occur thorough at home use, or at a primary care center in conjunction with a procedure, such as a colonoscopy.
  • device 100 may be used in conjunction with a laxative (such as may be prescribed prior to a colonoscopy).
  • a laxative such as may be prescribed prior to a colonoscopy.
  • An embodiment of the laxative of the present disclosure may be used to void a bodily fluid from a mammal while still preserving a specified diagnostic marker contained w ithin the bodily fluid.
  • Such a laxative may comprise any commonly recognized laxative (such as Fleet's* Phospho-soda oral laxative, C.B. Fleet Company) combined w ith at least one stabil izing agent.
  • the diagnostic marker may in some instances be a marker that is present in the solids and/or liquids of the gastrointestinal tract.
  • the contents of the colon may be expelled and collected in device 100 for the detection of the presence of and the level of the diagnostic marker.
  • the laxative contains an embodiment of a stabilizing agent to inhibit the destructive components.
  • a method of detecting a diagnostic marker in bodily fluids is shown in Fig. 3.
  • the method 300 comprises the step 3 10 of mixing a body fluid of the gastrointestinal tract having a diagnostic marker with a stabilizing agent and the step 320 of collecting the stabilized diagnostic marker. Further, method 300 may also comprise the step 330 of analyzing the collected stabilized diagnostic marker.
  • Step 3 10 in at least one embodiment may occur in the patient through use of a laxative or other ingested composition which contains a stabilizing agent. Collecting of the stabilized diagnostic marker in step 320 may occur through use of an embodiment of dev ice 1 00 where the bodily fluid enters the introduction lumen 1 12, passes through diagnostic filter 1 1 0 and exits through outlet 106.
  • Diagnostic markers present in the bodily fluid may be trapped in the diagnostic filter 1 10, or by an indicator contained with in diagnostic filter 1 10 to increase the concentration of the diagnostic marker to a detectable level.
  • diagnostic filter 1 10 may be analyzed in step 330 either internal in device 100 or removed from device 1 00.
  • the analysis of step 330 may use any appropriate analytical 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 embod iment, may be of a modified product, a cleavage product, or cleavage pattern, of a diagnostic marker.
  • the diagnostic marker may bind to an indicator which is capable of displaying a colorimetric or luminescent indicator in recognition of the diagnostic marker.
  • conditioning of body fluids for the preservation of diagnostic markers may be accompl ished 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 embod iment 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 1 0, 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 800ppm to about 1200 ppm.
  • the stabilizing agent may be present at a level of approximately 500ppm.
  • 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 stale or heath/li estyle 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, metabol ic 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 hot linum, Clostridium difficile, Clostridium perfringens, Clostridium tetani, Borrelia burgdorferi, Treponema pallidum, Chlamydia trachomatis, Chlamydophila psittaci, Corynehacterium diphtheriae, Mycobacterium tuberculosis, and Mycobacterium avium, Rickettsia prowazekii, Rickettsia rickettsii, Rickettsia t
  • Ehrlichia chaffeensis Brucella melitensis, Bordetella pertussis, Burkholderia mallei, B. pseudomallei, Neisseria gonorrhoeae, Neisseria meningilides, 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 mirahilis, Yersinia pestis, Yersinia enter ocolitica, 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 D A viruses, including the genus Panwiridae, 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 v irus. Human immunodeficiency virus 1 and 2, and Human T-lymphotropic virus 1).
  • Fungal infections may include a mammalian infection by any bacterial species.
  • Embod iments 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.
  • Embod iments of the parasite may be, but are not limited to, Entamoeba histolytica, Babesia microti, Babesia sp. WAI, 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 (A 1 AT) - G Protein, Cystatin C, Cystatin A, alpha-2- macroglobulin (A2MG), Uteroglobin, Transthyretin (TTR), Annexin Al , Annexin A2, Annexin A3 and Calnexin.
  • a 1 AT Alpha- 1 antitrypsin
  • A2MG alpha-2- macroglobulin
  • TTR Transthyretin
  • Annexin Al Annexin A2 Annexin A2 Annexin A3 and 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, Angiogcnin, Annexin A l , B-cell activating factor (BAFF), B-cell lymphoma 2 (BCL2)-like 2, Beta Human chorionic gonadotropin, Ca l 5-3, Calcyclin, Calvasculin, Cathepsin D, Caveolm- 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 (gp l 30), Glutathione S-transferase Mu 1 (GSTM 1 ), Hepsin, High
  • Diagnostic markers directed towards a bacteria! 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, 1 7-Hydroxyprogesterone, 25-hydroxy- vitamin D, Antineutrophil Cytoplasmic Antibodies, 5-Hydroxy Tryptamine, 5- hydroxyindoleacetic acid, Acetoacctate, 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, Alpha l -antitrypsin, Alzheimer biomarkers (including Tau protein and Amyloid), and Tau protein and Amyloidsin
  • Antiphospholipids Antistreptolysin O titer, Antithrombin, Antithyroglobulin antibody, Antithyroid antibodies, Apolipoprotein ⁇ - ⁇ , 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 I and Breast Cancer Gene 2, c-ANCA, c-erbB-2, C-peptidc, C-Reactive Protein, Caffeine.
  • Direct Antiglobulin Test Direct bilirubin, Direct Low-density lipoprotein cholesterol.
  • 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, Mycophcnolic acid, Mycoplasma. Mycoplasma pneumoniae IgG and IgM antibodies, Myeloperoxidase Antibodies, Myoglobin, N- terminal pro b-type natriuretic peptide.
  • 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 a-amylascs are calciummetalloenzymes that are completely unable to function in the absence of calcium. Through acting at random locations on the starch chain, a-amylase breaks down carbohydrates into maltotriose and maltose from amylase. In animals, a-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 oc-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. I t 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 alum inum potassium sulfate dodecahydrate.
  • 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 dodecahydratc.
  • 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 inactivalion 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 tablets, beverages, coatings, or other applicable vehicles to deliver a stabilizing agent to the site of a diagnostic marker.
  • Embodiments of a carrier 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. Hxemplary embodiments of these components are listed herein ⁇ See section entitled "Additional Components for Rinse").
  • 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 Adm inistration of the United States of America.
  • the preservative may comprise sodium azide.
  • 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 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:
  • ⁇ 1- .AKiNG 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 thiodipropionatc, crythorbic acid, gum guaiac, methylparaben, potassium bisulfite, potassium metabisulfite, potassium sorbate, propionic acid, propyl gallate, propylparaben, sodium ascorbate, sodium benzoate, sodium bisul fite, sod ium metahisulfite, sodium propionate, sodium sorbate, sodium sulfite, sorbic acid, stannous chloride, sulfur dioxide, thiodipropionic acid, tocopherols, and sodium azide.
  • 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 bitarlrate, choline chloride, copper gluconate, cuprous iodide, cysteine, cystine, ferric phosphate, ferric pyrophosphate, ferric sodium pyrophosphate, ferrous gluconate, ferrous lactate, ferrous sulfate, glycine, histidine.
  • 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 c itrate, 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, stcrculia (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, hel ium, hydrochloric acid, hydrogen peroxide, lactic acid, lecithin, magnesium carbonate, magnesium hydroxide, magnesium oxide, magnesium stearate, malic acid, methylcellulose, monoammonium glutamate, mono
  • SYNTHETIC FLAVORING SUBSTANCES acetaldehyde, acetoin, aconitic acid, anethole, benzaldchyde, N-butyric acid, d- or 1-carvone cinnamaldehyde, citral, decanal, diacetyl, ethyl acetate, ethyl butyrate, ethyl vanillin, cugcnol, geraniol, geranyl acetate, glycerol tributyrate limonene, linalool, linalyl acetate, 1-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, such as CA 19-9, 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, MHC 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:
  • AlexaFluor® • AlexaFluor®(AF); o AF405, AF488.AF500, AF514, AF532, AF546, AF555, AF568, AF594, AF610, AF633, AF635, AF647, AF680, AF700, AF710, AF750, AF800;
  • BFP1CFP • GFP and GFP derivated mutant proteins
  • BFP1CFP YFP, DsRcd, Tl , Dimer2,
  • mRFP l 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 peptidc/protcin, Dex, or poly-sacceride. o Any combination of the fluorescent dyes involving in generation of FRET;
  • 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-diabctes, cardiac disease, precardiac 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 (cTnl).
  • cardiac markers such as Creatine kinase-MB (CK-MB), myoglobin, homocysteine, C-reactive protein (CRP), troponin T (cTnT), and troponin I (cTnl).
  • 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, allikrein 3, Vascular endothelial growth factor A (VEGF), Calcitonin, Chromogranin A, Gastrin, S I 00 alpha chain, Somatostatin, Thyroglobulin, V-erb-b2, cyckub-dependent kinase inhibitor 1 (p2 I ), Breast Cancer Antigen 1 and 2 (BRCA l and 2), MutL homolog 1 (MLH 1 ), MutS homolog 2 (MSH2), MutS homolog 6 ( MS H6). and postmeiotic segregation increased 1 and 2 (PMS 1 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.
  • 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 method of biomarker stabilization and detection 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 w ith 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.
  • Infertility Inflammatory Bowel Disease, Influenza, Influenza A, Influenza B, Insulin Resistance, Jaundice, Juven ile 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.
  • 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

Herein are presented devices, systems, and methods for biomarker stabilization and analysis, including formulations which function to preserve the level of markers in body fluids from enzymatic alteration and degradation. In embodiments the methods and formulations disclosed can be used to improve the sampling and testing of a body fluid (including body fluids comprising fecal matter) by conditioning the body fluid. Also provided are embodiments of devices for detecting and indicating diagnostic markers in body fluids as well as the diagnosis of particular disease states.

Description

DEVICES, SYSTEMS, AND METHODS FOR FECAL ANALYSIS
BACKGROUND
Colorectal cancer, also called colon cancer or large bowel cancer, includes cancerous growths in the colon, rectum and appendix. With 655,000 deaths worldwide per year, it is the fourth most common form of cancer in the United States and the third leading cause of cancer- related death in the Western world (National Cancer Institute, 2009. http://www.cancer.gov/cancertopics/commoncancers.). Colorectal cancers arise from adenomatous polyps in the colon. These mushroom-shaped growths are usually benign, but some develop into cancer over time. Localized colon cancer is usually diagnosed through colonoscopy.
During a colonoscopy, a long, flexible tube (colonoscope) is inserted into the rectum. A tiny video camera at the tip of the lube allows the doctor to view the inside of the entire colon. If necessary, polyps or other types of abnormal tissue can be removed through the scope during a colonoscopy. Tissue samples (biopsies) can be taken during a colonoscopy as well. Preparation of the colon to allow for the visualization of abnormal conditions involves the use of laxitives or other processes to cleanse the colon. Because of this invasive nature of the procedure and the costs associated with the procedure, there is a need for an improved method of diagnosis.
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 :
Fig. 1 shows a perspective view of a device for fecal analysis, according to at least one embodiment of the present disclosure;
Fig. 2 shows a perspective view of a device for fecal analysis, according to at least one embodiment of the present d isclosure:
Fig. 3 shows a flowchart indicating the steps of a method for fecal analysis, 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 devices, sy stems, 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 or a device used to test 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, scrum and mixtures of thereof. Body fluids may also comprise cancer cells, peripheral blood mononuclear cells, lymphocytes, lymph fluid, and other tissue secretions or fluid.
Turning to Fig. 1 , at least one embodiment of the present d isclosure of a device for the detection of a diagnostic marker in a bodily fluid is disclosed. The device 100 is comprised of a chamber 102 sized and shaped to define an inlet 104, an outlet 106, and a passage 108 fluidly connecting the in let 104 and outlet 1 06. Device 100 may further comprise a diagnostic filter 1 10 positioned in the passage so as to define a introduction lumen 1 12 between the inlet 104 and the diagnostic filter 1 1 0, and an outlet lumen 1 14 between the outlet 1 06 and the diagnostic fi lter 1 10. The device 1 00 may also comprise a stabilizing bar 1 16 coupled to chamber 1 02, where stabilizing bar 1 16 is operable to affix to a stationary apparatus, such as the side of a toilet. Additionally, device 100 may also comprise a inlet cap 1 18 capable of coupling to the inlet 1 04, and an outlet cap 120 capable of coupling to outlet 1 06. Optionally, one or both of inlet cap 1 1 8 and outlet cap 120 may be coupled to inlet 104 and/or outlet 1 06 respectively to form a fluid- tight seal.
Diagnostic filter 1 1 0 in an exemplary embodiment comprises a filter which serves to prevent passage of particulate material below a predetermined size threshold from passing therethrough. Additionally, diagnostic filter 1 10 may also comprise at least one indicator capable of binding or recognizing a diagnostic marker. Such an indicator may also be capable of presenting a visual signal in response to exposure to the diagnostic marker. As shown in Fig. 2,
Ί diagnostic filter 1 10 in at least one embodiment may be comprised of more than one component. In an exemplary embodiment, diagnostic filter 1 10 is comprised of filter 122 and diagnostic membrane 124. Further, diagnostic filter 1 10, in at least one embodiment, may be removable from chamber 102 for futher processing.
Device 100 in at least one exemplary embodiment may also comprise a stabilizing agent capable of completely or substantially preventing the degradation or inactivation of a diagnostic marker contained within the bodily fluid passing through device 100. Such a stabilizing agent may be integral to or coated on diagnostic filter 1 10 or introduction lumen 1 12 of device 100. The stabilizing agent may be any compound which completely or substantially preventing the degradation or inaclivation of a specified diagnostic marker contained within the bodily fluid. In at least one embodiment, the stabilizing agent may be selected from a group consisting of aluminum sulfate hydrate, benzoic acid, aluminum potassium sulfate dodecahydrate, or a combination thereof. Further, the stabilizing agent may comprise more than one stabilizing agent, such as a exemplary stabilizing agent which contains aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate.
The diagnostic marker detected in device 1 00 may be selected from the group consisting of Adenomatous polyposis coli (APC), β-catenin, Wnt, and Cancer Antigen CA 19-9. In some instances, the diagnostic marker may be indicative of a cancer, such as colorectal cancer.
In at least one embodiment, device 100 may be used to collect fecal material or urine to determine whether a predetermined diagnostic marker is present. Such a use may occur thorough at home use, or at a primary care center in conjunction with a procedure, such as a colonoscopy.
In at least one embodiment of the present disclosure, device 100 may be used in conjunction with a laxative (such as may be prescribed prior to a colonoscopy). An embodiment of the laxative of the present disclosure may be used to void a bodily fluid from a mammal while still preserving a specified diagnostic marker contained w ithin the bodily fluid. Such a laxative may comprise any commonly recognized laxative (such as Fleet's* Phospho-soda oral laxative, C.B. Fleet Company) combined w ith at least one stabil izing agent. The diagnostic marker may in some instances be a marker that is present in the solids and/or liquids of the gastrointestinal tract. Through use of the laxative, the contents of the colon may be expelled and collected in device 100 for the detection of the presence of and the level of the diagnostic marker. Since, the gastrointestingal tract contains many destructive components which degrade biological markers, the laxative contains an embodiment of a stabilizing agent to inhibit the destructive components.
In at least one embodiment of the present disclosure, a method of detecting a diagnostic marker in bodily fluids is shown in Fig. 3. The method 300 comprises the step 3 10 of mixing a body fluid of the gastrointestinal tract having a diagnostic marker with a stabilizing agent and the step 320 of collecting the stabilized diagnostic marker. Further, method 300 may also comprise the step 330 of analyzing the collected stabilized diagnostic marker. Step 3 10 in at least one embodiment may occur in the patient through use of a laxative or other ingested composition which contains a stabilizing agent. Collecting of the stabilized diagnostic marker in step 320 may occur through use of an embodiment of dev ice 1 00 where the bodily fluid enters the introduction lumen 1 12, passes through diagnostic filter 1 1 0 and exits through outlet 106. Diagnostic markers present in the bodily fluid may be trapped in the diagnostic filter 1 10, or by an indicator contained with in diagnostic filter 1 10 to increase the concentration of the diagnostic marker to a detectable level. Once the diagnostic marker has been collected, diagnostic filter 1 10 may be analyzed in step 330 either internal in device 100 or removed from device 1 00. The analysis of step 330, in at least one embodiment, may use any appropriate analytical 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 embod iment, may be of a modified product, a cleavage product, or cleavage pattern, of a diagnostic marker. Futher, in at least one embodiment, the diagnostic marker may bind to an indicator which is capable of displaying a colorimetric or luminescent indicator in recognition of the diagnostic marker.
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 accompl ished 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 embod iment 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 stabi l ization 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 1 0, 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 800ppm to about 1200 ppm. Moreover, in at least one embodiment, the stabilizing agent may be present at a level of approximately 500ppm.
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 6 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 Aluminum potassium sulfate
42 dodecahydrate Sigma-Aldrich
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
Table I - GRAS Compounds as Stabilizing Agents 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.
Figure imgf000009_0001
Table II
As used herein, "diagnostic marker" may be any biological molecule whose presence, specific concentration, or specific activity may be indicative of a disease stale or heath/li estyle 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, metabol ic 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 hot linum, Clostridium difficile, Clostridium perfringens, Clostridium tetani, Borrelia burgdorferi, Treponema pallidum, Chlamydia trachomatis, Chlamydophila psittaci, Corynehacterium diphtheriae, 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 meningilides, 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 mirahilis, Yersinia pestis, Yersinia enter ocolitica, 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 D A viruses, including the genus Panwiridae, 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 v irus. 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. Embod iments 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. Embod iments of the parasite may be, but are not limited to, Entamoeba histolytica, Babesia microti, Babesia sp. WAI, 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 (A 1 AT) - G Protein, Cystatin C, Cystatin A, alpha-2- macroglobulin (A2MG), Uteroglobin, Transthyretin (TTR), Annexin Al , 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, Angiogcnin, Annexin A l , B-cell activating factor (BAFF), B-cell lymphoma 2 (BCL2)-like 2, Beta Human chorionic gonadotropin, Ca l 5-3, Calcyclin, Calvasculin, Cathepsin D, Caveolm- 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 (gp l 30), Glutathione S-transferase Mu 1 (GSTM 1 ), Hepsin, High-mobility group protein B l (I IMGB-1 ), Insulin-like growth factor binding protein 1 (lGFBP- 1 ), Insulin-like growth factor binding protein 4 (1GFBP-4), Insulinlike growth factor bind ing 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, Peroxircdoxin, Phosphoserine aminotransferase (PSATl ), Prostate Specific Antigen, Receptor tyrosinc-protcin kinase erbB-3 (ErbB3), Serpin B3, Vascular smooth muscle cell growth factor R2 (VSGF R2/ DR), 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-β Ι ), Tumor necrosis factor receptor 1 (TNF-R l ), urokinase-type Plasminogen Activator (uPA), urokinase-type Plasminogen Activator Receptor (uPAR), Brcal, Brcall, kallikreins, e-cadhcrin, Hox peptide, and EngraiIed-2.
Diagnostic markers directed towards a bacteria! or virus may include any surface or secreted antigen from the bacteria or virus, or a conserved nucleotide sequence.
Figure imgf000012_0001
Table HI 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, 1 7-Hydroxyprogesterone, 25-hydroxy- vitamin D, Antineutrophil Cytoplasmic Antibodies, 5-Hydroxy Tryptamine, 5- hydroxyindoleacetic acid, Acetoacctate, 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, Alpha l -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 citruilinated peptide antibody, Anti-retroviral drug resistance testing, anti- ribonucleoprotein, anti-Sjogren's Syndrome A, anti-Sjogren'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 Λ-Ι, 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 I and Breast Cancer Gene 2, c-ANCA, c-erbB-2, C-peptidc, C-Reactive Protein, Caffeine. Calcidiol, Calcifidiol, Calcitonin, Calcitriol, Calcium, Calcofluor white stain, Cancer Antigen 125. Cancer Antigen 1 5-3. Cancer Antigen 19-9, Cancer antigen-breast, Cancer antigen-Gl. 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. Chromogran in A, Chymotrypsin, Citrulline antibody. Coagulation Factors, Cobalamin, Complement Component C3, Complement Component C4, Complexed PSA, Conjugated bilirubin. Copper, Corticotropin,
] 1 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 Antinuclcar Antibody, Fluorescent treponemal antibody absorption, Folic Acid, Follicle-stimulating hormone, Fragment D-dimer, Fructosaminc, 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 l 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, Mycophcnolic 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 I I 20210, Red Blood Cel l Antibody Identification, Renin, Respiratory Syncytial Virus, Rheumatoid Factor, Ristocetin Cofactor, Rubella, Scleroderma antibodies, Serine Protease 3, Serotonin, Siderophilin, S irolimus, Smith antibody, Smooth Muscle Antibody, Sodium, Soluble Mesothel in-Related Peptides, Somatomedin C, Somatotropin, Syphilis, Tacrolimus, Tegretol®, Testosterone, Testosterone-estrogen B inding 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, Tripsinogen, Tryptase, Valproic acid, Vancomycin, Vanillylmandelic acid, , Varicella Zoster Virus, Vasopressin, Very Low-Density Lipoprotein Cholesterol, Viral Hepatitis A Antibody, Viral Hepatitis C. Viral Load (HIV), Vitamin B 12, Vitamin B 12 & Folate, Vitamin D, Vitamin D2, Vitamin D3, Vitamin , 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 a-amylascs are calciummetalloenzymes that are completely unable to function in the absence of calcium. Through acting at random locations on the starch chain, a-amylase breaks down carbohydrates into maltotriose and maltose from amylase. In animals, a-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 oc-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. Lvsozvme
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. I t 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 alum inum potassium sulfate dodecahydrate.
4. Cancer Antigen 1 9-9 (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 dodecahydratc.
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 inactivalion 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 tablets, beverages, coatings, 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. Hxemplary 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 Adm inistration of the United States of America. Moreover, in at least one embodiment of the carrier, the preservative may comprise sodium azide.
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 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. ΛΝΊ 1- .AKiNG 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 thiodipropionatc, crythorbic acid, gum guaiac, methylparaben, potassium bisulfite, potassium metabisulfite, potassium sorbate, propionic acid, propyl gallate, propylparaben, sodium ascorbate, sodium benzoate, sodium bisul fite, sod ium metahisulfite, sodium propionate, sodium sorbate, sodium sulfite, sorbic acid, stannous chloride, sulfur dioxide, thiodipropionic acid, tocopherols, and sodium azide.
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 bitarlrate, 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, linolcic 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 B 12, 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 c itrate, 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, stcrculia (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, hel ium, 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- mcthylcellulose, 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, benzaldchyde, N-butyric acid, d- or 1-carvone cinnamaldehyde, citral, decanal, diacetyl, ethyl acetate, ethyl butyrate, ethyl vanillin, cugcnol, geraniol, geranyl acetate, glycerol tributyrate limonene, linalool, linalyl acetate, 1-malic acid, methyl anthranilate, 3-methyl -3- phenyl glycidic acid, ethyl ester, piperonal, vanillin.
II. Indicator Carriers
In at least one add itional 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, such as CA 19-9, 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, MHC 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 isothiocy anate) 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, DsRcd, Tl , Dimer2, mRFP l, 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 peptidc/protcin, 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-diabctes, cardiac disease, precardiac 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 (cTnl). 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, allikrein 3, Vascular endothelial growth factor A (VEGF), Calcitonin, Chromogranin A, Gastrin, S I 00 alpha chain, Somatostatin, Thyroglobulin, V-erb-b2, cyckub-dependent kinase inhibitor 1 (p2 I ), Breast Cancer Antigen 1 and 2 (BRCA l and 2), MutL homolog 1 (MLH 1 ), MutS homolog 2 (MSH2), MutS homolog 6 ( MS H6). and postmeiotic segregation increased 1 and 2 (PMS 1 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. 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).
111. 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 method of biomarker stabilization and detection 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 w ith 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 ery thematosus, Double pneumonia, Down Syndrome, Rncephalitis, 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 H 1 N 1 , 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, Juven ile 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, Sjogren'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 B 12 and Folate Deficiency, Vitamin B 12 Deficiency, Vitamin K Deficiency, Walking pneumonia, West Nile Virus, Wilson's Disease, Wound and Skin infections.
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

CLAIMS:
1. A device for detection of a diagnostic marker in a bodily fluid, the device comprising:
a chamber sized and shaped to defined an inlet, an outlet, and a passage flu idly connecting the inlet and outlet; and
a diagnostic filter capable of binding to a diagnostic marker, the diagnostic filter positioned in the passage so as to define a introduction lumen between the inlet and the diagnostic filter, and an outlet lumen between the outlet and the diagnostic filter.
2. The device of claim I , further comprising a stabilizing agent positioned in the introduction lumen, the stabilizing agent useful to completely or substantially prevent degradation or inaetivation of the diagnostic marker.
3. The device of claim 1 , wherein the stabilizing agent is selected from the group consisting of aluminum sulfate hydrate, benzoic acid, aluminum potassium sulfate dodecahydrate, and a combination thereof.
4. The device of claim I , wherein the stabilizing agent comprises aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate.
5. The device of claim 1 , wherein the diagnostic marker is selected from the group consisting of Adenomatous polyposis coli (APC), β-catenin, Wnt, and Cancer Antigen CA 19-9.
6. The device of claim 1 , wherein the diagnostic marker is indicative of a cancer.
7. The device of claim 6, wherein the cancer is a colorectal cancer.
8. The device of claim 1 further comprising a plurality of arms coupled to the chamber, the plurality of arms sized and shaped receive a stabilizing structure.
9. The device of claim 8, wherein the stabilizing structure is a portion of a toilet.
10. The device of claim 1 , wherein the diagnostic filter is capable of preventing the flow therethrough of particulate matter of a predetermined size.
1 1 . A method of stabilizing a diagnostic marker, the method comprising the step of: mixing a body fluid comprising a diagnostic marker with a stabilizing agent; collecting the body fluid with the stabilized diagnostic marker; and analyzing the collected diagnostic marker: wherein the stabilization agent comprises a Generally Regarded as Safe (GRAS) compound; and
wherein the stabilization agent completely or substantially prevents the cleavage, degradation, or inactivation of the diagnostic marker.
12. The method of claim 1 1 wherein the step of mixing a body fluid with a stabilizing agent comprises introducing the stabilizing agent into a mouth/oral cavity of a patient so that the stabilizing agent mixes with saliva.
13. The method of claim 1 1 , wherein the stabilizing agent is useful to completely or substantially inactivate an enzyme selected from the group consisting of an amylase, a lysozyme, a peroxidase, a glycosidase, an esterase, a protease, and a peptidase.
14. The method of claim I I , wherein the body fluid is selected from the group consisting of urine and an anal secretion.
15. The method of claim 1 1 , wherein the stabilizing agent is selected from a group consisting of Fixanal® Buffer 6.0 (Sigma-Aldrich Co.), acetic acid, aluminum hydroxide bentonite, alum inum sulfate hydrate, aluminum potassium sulfate dodecahydrate, benzoic acid, caffeine, and 3-tert-butyl-hydroxyanisole, or a combination thereof.
16. The method of claim 1 1 , wherein the stabilizing agent is capable of inhibiting degradation of the diagnostic marker to an inhibitory degree, wherein the inhibitory degree is selected from a group consisting of 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%.
17. The method of claim 1 1, wherein the stabilization agent has a concentration selected from the group consisting of about 200 parts per mill ion (ppm) to about 2000ppm, about 400ppm to about 1600ppm, about 600ppm to about 1400ppm, about 800ppm to about 1200ppm, and about 400ppm to about 600ppm.
18. The method of claim 1 1 , wherein the diagnostic marker is selected from the group consisting Adenomatous polyposis coli (APC), β-catenin, Wnt, and Cancer Antigen CA 19-9.
PCT/US2011/066616 2010-12-22 2011-12-21 Devices, systems, and methods for fecal analysis WO2012088330A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201061425784P 2010-12-22 2010-12-22
US61/425,784 2010-12-22

Publications (2)

Publication Number Publication Date
WO2012088330A2 true WO2012088330A2 (en) 2012-06-28
WO2012088330A3 WO2012088330A3 (en) 2012-09-13

Family

ID=46314904

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/066616 WO2012088330A2 (en) 2010-12-22 2011-12-21 Devices, systems, and methods for fecal analysis

Country Status (1)

Country Link
WO (1) WO2012088330A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11458476B2 (en) 2019-10-17 2022-10-04 Marvin Liu In-situ fecal specimen sampling device

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5066463A (en) * 1990-10-01 1991-11-19 Chang Maw Guay Multiple-purpose fecal examination apparatus
US20050069900A1 (en) * 2003-09-25 2005-03-31 Cytyc Corporation Analyte sample detection
US20050232813A1 (en) * 2004-04-16 2005-10-20 Karmali Rashida A Specimen collecting, processing and analytical assembly
US20070172823A1 (en) * 2003-09-23 2007-07-26 Universitaet Potsdam Method for conducting non-invasive early detection of colon cancer and/or of colon cancer precursor cells
US20070248989A1 (en) * 2006-04-21 2007-10-25 Prasad Devarajan Method and Kit for the Early Detection of Impaired Renal Status

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5066463A (en) * 1990-10-01 1991-11-19 Chang Maw Guay Multiple-purpose fecal examination apparatus
US20070172823A1 (en) * 2003-09-23 2007-07-26 Universitaet Potsdam Method for conducting non-invasive early detection of colon cancer and/or of colon cancer precursor cells
US20050069900A1 (en) * 2003-09-25 2005-03-31 Cytyc Corporation Analyte sample detection
US20050232813A1 (en) * 2004-04-16 2005-10-20 Karmali Rashida A Specimen collecting, processing and analytical assembly
US20070248989A1 (en) * 2006-04-21 2007-10-25 Prasad Devarajan Method and Kit for the Early Detection of Impaired Renal Status

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11458476B2 (en) 2019-10-17 2022-10-04 Marvin Liu In-situ fecal specimen sampling device

Also Published As

Publication number Publication date
WO2012088330A3 (en) 2012-09-13

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
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11851897

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11851897

Country of ref document: EP

Kind code of ref document: A2