WO2011127467A1 - Devices, systems, and methods for biomarker stabilization - Google Patents

Devices, systems, and methods for biomarker stabilization Download PDF

Info

Publication number
WO2011127467A1
WO2011127467A1 PCT/US2011/031935 US2011031935W WO2011127467A1 WO 2011127467 A1 WO2011127467 A1 WO 2011127467A1 US 2011031935 W US2011031935 W US 2011031935W WO 2011127467 A1 WO2011127467 A1 WO 2011127467A1
Authority
WO
WIPO (PCT)
Prior art keywords
agent
detection
diagnostic marker
binding
stabilizing
Prior art date
Application number
PCT/US2011/031935
Other languages
French (fr)
Inventor
Richard H. Selinfreund
Rakesh Vig
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
Priority claimed from US12/906,239 external-priority patent/US20110195423A1/en
Application filed by Companion Diagnostics, Inc. filed Critical Companion Diagnostics, Inc.
Publication of WO2011127467A1 publication Critical patent/WO2011127467A1/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
    • G01N33/573Immunoassay; Biospecific binding assay; Materials therefor for enzymes or isoenzymes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere

Definitions

  • Biomarkers are molecular indicators of a specific biological property, a biochemical feature or facet that can be used to measure the progress of a disease or the effects of a treatment.
  • serum low-density lipoprotein (LDL) is a biomarker of cholesterol and blood pressure
  • P53 gene is a biomarker for cancer.
  • LDL low-density lipoprotein
  • accurate diagnosis is particularly important, especially where the side effects of a treatment are severe.
  • biomarkers as molecular indicators not only detect the presence or absence of the biomarker, but often must measure the exact concentration of a biomarker to determine whether an abnormal condition exists. Because of the requirement for accuracy, the process of sample collection, preparation, and analysis are often complicated and time consuming. Currently, blood-based assays for biomarker presence or activity are considered to be the "gold standard" for biomarker-type assays.
  • the disclosure of the present application provides computer-implemented methods and systems of improving diagnostic marker availability.
  • the system comprises a stabilizing agent useful to completely or substantially prevent degradation or inactivation of a diagnostic marker present within a mammalian body, the diagnostic marker indicative of a mammalian condition, and a detection agent capable of detecting the diagnostic marker.
  • an embodiment of the system may further comprise a detection platform comprising a plurality of detection sites each capable of receiving the diagnostic marker, the stabilizing agent, and the detection agent, a computer database capable of receiving a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of the diagnostic marker to the detection agent, a processor operably coupled to the computer database and the detection platform, the processor having and executing a software program operational to (1) determine a binding characteristic between the detection agent and the stabilized diagnostic agent in each of the plurality of detection sites, (2) compare the binding characteristic among each of the plurality of detection sites, wherein the comparison of binding characteristics is capable of determining the stabilizing agent with the greatest effect on the binding characteristic between the detection agent and the diagnostic agent, (3) generate a binding record using the compared binding characteristics, and (4)deliver the binding record to a recipient.
  • the software program is further operational to compare the binding characteristic with at least one of a plurality of stored binding characteristics in the computer database.
  • the system further comprises a detection platform comprising a plurality of detection sites each capable of receiving the diagnostic marker, the stabilizing agent, and the detection agent, a detection device capable of determining a binding characteristic between the detection agent and the diagnostic marker in the detection sites, a computer database capable of receiving a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of the diagnostic marker to the detection agent, a processor operably coupled to the computer database and the detection device, the processor having and executing a software program operational to (1) determine the binding characteristic of the detection agent to the diagnostic marker in each of the plurality of detection sites, (2) compare the binding characteristic among each of the plurality of detection sites to order the level of effect of the stabilizing agents assayed on the binding characteristic between the detection agent and the diagnostic agent, (3)generate a binding report using the compared binding characteristics, and (4) deliver the binding report to a recipient.
  • a detection platform comprising a plurality of detection sites each capable of receiving the diagnostic marker, the stabilizing agent, and the detection agent, a detection device capable
  • the stabilizing system further comprises a detection platform comprising a plurality of detection sites each capable of receiving at the stabilizing agent, the detection agent, and a body fluid from a patient, the body fluid comprising the diagnostic marker, a computer database capable of storing a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of the diagnostic marker to the detection agent, and a processor operably coupled to the computer database and the detection platform, the processor having and executing a software program operational to determine a binding characteristic of the detection agent and a stabilized diagnostic marker in each of the plurality of detection sites compare the binding characteristic among each of the plurality of detection sites, wherein the comparison of binding characteristics is capable of determining the stabilizing agent with the greatest effect on the binding characteristic between the detection agent and the diagnostic marker, generate a binding record using the compared binding characteristics, and deliver the binding record to a recipient.
  • the binding record may be used by to assist in determining a therapeutic course for the patient.
  • the system further comprises a fluid collector to collect a body fluid, and a housing to test and retain the body fluid, the housing comprising a collection chamber, having an open end, to receive the fluid collector and contain the stabilization agent, at least one membrane test strip, in fluid communication with the collection chamber, to indicate the presence or absence of the diagnostic marker, and an immunoassay-based fingerprint acquisition pad in fluid communication with the collection chamber, the acquisition pad having at least one control zone including a control reagent to identify a donor of the body fluid, the reagent including a member of a predetermined ligand/receptor binding pair, and a plurality of reaction zones, each of which includes the detection agent to determine the presence of the diagnostic marker in the fluid sample, the detection agent including a member of a predetermined ligand/receptor binding pair.
  • the system further comprises a detection platform comprising a plurality of detection sites each capable of receiving the diagnostic marker, the stabilizing agent, and the detection agent, a computer database capable of receiving a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of the diagnostic marker to the detection agent, a processor operably coupled to the computer database and the detection platform, the processor having and executing a software program.
  • the software program operational to determine a binding characteristic of the detection agent and a stabilized detection agent in each of the plurality of detection sites, compare the binding characteristic among each of the plurality of detection sites, wherein the comparison of binding characteristics is capable of determining the stabilizing agent with the greatest effect on the binding characteristic between the detection agent and the diagnostic agent, generate a binding record using the compared binding characteristics, and deliver the binding record to a recipient.
  • the system further comprises a detection platform comprising a plurality of detection wells housed in a solid matrix, the detection wells containing a plurality of the diagnostic markers and at least one competitor molecule, wherein the plurality of the diagnostic markers and at least one control molecule are each bound to a paramagnetic particle, a detection system capable of receiving the detection platform, exerting a magnetic field upon a surface of the detection platform, and determining at least one characteristic of the diagnostic marker.
  • An embodiment of the solid matrix may be comprised of material selected for the group consisting of quartz, glass, metal, silica-based materials, resins, polymers, or combinations thereof.
  • the magnetic field may be able to force the paramagnetic particle towards the surface of the detection platform.
  • the an embodiemt of the system may further comprise a second paramagnetic particle, such as Europium, linked to a detection antibody, and optionally coupled to a second detection agent.
  • the system further comprises a diagnostic device comprising a plurality of test wells, wherein each test well is capable of containing the detection agent, and a detection device in communication with the diagnostic device, wherein the detection device is capable of detecting an interaction between the at least one detection agent and the diagnostic marker in at least one of the plurality of test wells, wherein the stabilizing agent is contained within at least one of the plurality of test wells.
  • the diagnostic device may be comprised of a material selected for the group consisting of quartz, glass, metal, silica-based materials, resins, polymers, or combinations thereof.
  • an embodiment of the detection device may be capable of detecting a colorimetric or fluorometric signal from at least one of the plurality of test wells.
  • the system further comprises a microarray product comprising at least 100 diagnostic markers/cm2, a microarray identifier, and the stabilizing agent, the system also having a control microarray product comprising a first specific binding pair member that binds to a first detectable label, and a processor for providing information regarding the identification and concentration of markers on the microarray based on the identity of the array provided by the microarray identifier.
  • the microarray product may be comprised of a compound selected from the group consisting of gel, nitrocellulose, nylon, quartz, glass, metal, silica based materials, silica, resins, polymers, or combinations thereof.
  • the diagnostic marker may comprises a DNA fragment having a length of about 10 base pairs to about 50 base pairs of DNA or R A, or a length of at least about about 50 amino acids.
  • the system further comprises a latex particle having a carboxylate group, the latex particle functionally coupled to the stabilizing agent.
  • the latex particle may further comprising at least one of human serum albumin (HSA) and bovine serum albumin (BSA), and/or a linker group coupled to latex particle, where the linker group may comprise a maleimide compound.
  • the maleimide compound is selected from the group consisting of Succinimidyl 4-[N-Maleimidomethyl] Cyclohexane-1 -Carboxylate (SMCC) and N- Hydroxysuccinimide-activated hexa(ethylene glycol) undecane Thiol (NHS).
  • the linker group may be coupled to the HSA or BSA.
  • the system may further comprise antibody functionally linked to the latex particle.
  • the latex particle in at least one embodiment may have a diameter of about 0.02 ⁇ to about 7.0 ⁇ .
  • the stabilizing agent is selected from the group consisting of a protease inhibitor, a DNase inhibitor, and a RNase inhibitor. Further, the stabilizing agent may be 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.
  • an embodiment of the stabilizing agent may be selected from the group consisting of Fixanal® Buffer 6.0 (Sigma-Aldrich Co.), acetic acid, aluminum hydroxide bentonite, aluminum sulfate hydrate, aluminum potassium sulfate dodecahydrate, benzoic acid, caffeine, and 3- tert-butyl-hydroxyanisole, or a combination thereof.
  • the stabilizing agent may comprise a plurality of stabilizing agents, each present in approximately the same concentration.
  • the stabilizing agent may be capable of inhibiting degradation of the diagnostic marker to an inhibitory degree, wherein the inhibitory degree is selected from the 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%, and at least about 99%.
  • the stabilizing agent has a concentration selected from the group consisting of about 200 parts per million (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.
  • the stabilizing agent may be able to inhibit the degradation or inactivation of the diagnostic marker for an inhibitory period selected from the group consisting of at least one minute, at least about five minutes, at least about ten minutes, at least about fifteen minutes, at least about thirty minutes, at least about one hour, at least about two hours, at least about four hours, and at least about eight hours.
  • the diagnostic marker is selected from the group consisting of a protein, a glycoprotein, a nucleic acid, an enzyme, an enzyme inhibitor, and a metabolite. Further, in at least one embodiment of the system or method of the present discosure, the diagnostic marker is selected from the group consisting a-hydroxybutyrate, 1-linoleoyl-GPC, palmitate, Glycine, and 3-methyl-2- oxybutyrate, glycosylated hemoglobin (HbAlc), IgE, and IgG, Aldose reductase, Angiogenin, Annexin Al, B-cell activating factor (BAFF), B-cell lymphoma 2 (BCL2)-like 2, Beta Human chorionic gonadotropin, Cal5-3, Calcyclin, Calvasculin, Cancer Antigen CA 19-9, Cancer Antigen CA 15-3, Cathepsin D, Caveolin-1, Chromogranin A, Alpha-crystallin B chain
  • the diagnostic marker may be indicative of a drug selected from the group consisting alcohol, cocaine, marijuana, opiates, amphetamine, methamphetamine, amphetamines, phencyclidine, benzodiazepines, barbiturates, methadone, tricyclic antidepressants, heroin, steroids, niacin, xanan, vicodin, oxycontin, adderall, morphine, and nicotine.
  • a drug selected from the group consisting alcohol, cocaine, marijuana, opiates, amphetamine, methamphetamine, amphetamines, phencyclidine, benzodiazepines, barbiturates, methadone, tricyclic antidepressants, heroin, steroids, niacin, xanan, vicodin, oxycontin, adderall, morphine, and nicotine.
  • the condition is selected from a group consisting of insulin resistance, glucose intolerance, cancer, cardiovascular disease, a bacterial infection, a viral infection, a fungal infection, a parasite infection, a food allergy, a non-food allergy, drug use, fertility, pregnancy, a level of circulating calcium.
  • the degradation or inactivation of the diagnostic marker for a condition may occur by a destructive agent, such as galactose oxidase.
  • the body fluid is selected from the group consisting of saliva, a mucous secretion, tears, sweat, semen, urine, a vaginal secretion, exhalate, blood, serum, and an anal secretion.
  • the method comprises the step of mixing a mammalian body fluid comprising a diagnostic marker indicative of a condition with an effective amount of a stabilizing agent, wherein the stabilizing agent completely or substantially prevents the cleavage, degradation, or inactivation of the diagnostic marker.
  • the step of mixing a body fluid with the stabilizing agent may comprise introducing the stabilizing agent into a mouth/oral cavity of a patient so that the stabilizing agent mixes with saliva.
  • the method comprises the steps of introducing a predetermined diagnostic marker for cancer into a plurality of detection sites of a detection platform, introducing a stabilization agent into each of the plurality of detection sites containing the predetermined diagnostic marker for cancer, wherein the stabilization agent in each of the plurality of detection sites is capable of completely or substantially preventing the degradation or inactivation of the diagnostic marker, introducing a detection agent into each of the plurality of detection sites having a stabilized diagnostic agent, determining a binding characteristic of the detection agent and the stabilized diagnostic agent in each of the plurality of detection sites with a processor, and computationally comparing the binding characteristic among each of the plurality of detection sites with the processor, wherein the comparison of binding characteristics is capable of determining the stabilizing agent with the greater effect on the binding characteristic between the detection agent and the diagnostic agent.
  • At least one embodiment of the method of the present disclosure may further comprise the step of computationally comparing the binding characteristics to at least one stored binding characteristic contained in the computer database in communication with the processor.
  • the method comprises the steps of contacting a body fluid of a diseased subject having a cancer with a stabilizing agent capable of completely or substantially preventing the degradation or inactivation of a diagnostic marker indicative of the cancer, the body fluid comprising the diagnostic agent, analyzing the stabilized body fluid to determine at last one property of the diagnostic marker to create a cancer profile; comparing the cancer profile with a control profile of a healthy subject not having the cancer, wherein the control profile is created in a like manner to the cancer profile, and generating a cancer identifier panel from the compared cancer profile, wherein the cancer identifier panel contains at least one cancer diagnostic marker indicative of the cancer.
  • the stabilizing agent may prevent the degradation or inactivation of the diagnostic marker for at least a twenty four hours period following interaction with the diagnostic agent.
  • the method comprises the steps of introducing a diagnostic marker into each of a plurality of detection sites of a detection platform, the detection platform comprising the plurality of detection sites each capable of receiving a diagnostic marker, a stabilization agent, and a detection agent, a detection device capable of determining a binding characteristic between the detection agent and the diagnostic marker in the detection sites, a computer database capable of receiving a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of a diagnostic marker to a detection agent, and a processor operably coupled to the computer database and the detection device.
  • the processor in at least one embodiment, having and executing a software program operational to (l)determine the binding characteristic of the detection agent to the diagnostic marker in each of the plurality of detection sites, (2) compare the binding characteristic among each of the plurality of detection sites to order the level of effect of the stabilizing agents assayed on the binding characteristic between the detection agent and the diagnostic agent, (3) generate a binding record using the compared binding characteristics, and (4) deliver the binding record to a recipient.
  • the at least one embodiment of the method further comprising the steps of introducing the stabilization agent to each of the plurality of detection sites containing the diagnostic marker, combining the detection agent with the stabilization agent and diagnostic marker in the detection sites, comparing the binding characteristic with the processor among each of the plurality of detection sites to order the level of effect of the stabilizing agents assayed on the binding characteristic between the detection agent and the diagnostic agent, generating a binding report using the compared binding characteristics, and delivering the binding report to a recipient.
  • the method comprises the steps of treating a subject having a disease state with an effective amount of therapeutic compound, harvesting at least one cell from the subject following a treatment with the therapeutic compound, bringing the at least one cell , such as a tumor cell, into contact with a stabilizing compound capable of completely or substantially preventing the degradation or inactivation of a diagnostic marker for the disease state, analyzing the at least one stabilized cell for the at least one diagnostic marker, creating a marker profile from at least one result determined from analyzing the at least one stabilized cell, and comparing the marker profile with at least one previous marker profile to determine efficacy of the therapeutic compound on the disease state.
  • the step of harvesting the at least one cell comprises the step of collecting a body fluid from the subject, wherein the body fluid comprises the at least one cell.
  • the method comprises the steps of harvesting a body fluid from a patient having a disease state, introducing the body fluid to a detection platform comprising a plurality of detection sites each capable of receiving a stabilization agent, a detection agent, and a body fluid from a patient comprising a diagnostic marker, a computer database capable of storing a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of a diagnostic marker to a detection agent, a processor operably coupled to the computer database and the detection platform, the processor having and executing a software program.
  • the software program operational to determine a binding characteristic of the detection agent and a stabilized diagnostic marker in each of the plurality of detection sites, compare the binding characteristic among each of the plurality of detection sites, wherein the comparison of binding characteristics is capable of determining the stabilizing agent with the greatest effect on the binding characteristic between the detection agent and the diagnostic marker, generate a binding record using the compared binding characteristics, and deliver the binding record to a recipient.
  • the method further comprises the steps of contacting the body fluid with a stabilizing agent capable of completely or substantially preventing the degradation or inactivation of a diagnostic marker for the disease state, analyzing the at least one stabilized body fluid for the at least one diagnostic marker to create a marker profile, comparing the marker profile with a marker profile library stored in the computer database to determine a therapeutic potential of the therapeutic compound on the disease state, generating a binding record using the compared binding characteristics, and delivering the binding record to a recipient.
  • a stabilizing agent capable of completely or substantially preventing the degradation or inactivation of a diagnostic marker for the disease state
  • analyzing the at least one stabilized body fluid for the at least one diagnostic marker to create a marker profile
  • comparing the marker profile with a marker profile library stored in the computer database to determine a therapeutic potential of the therapeutic compound on the disease state
  • generating a binding record using the compared binding characteristics and delivering the binding record to a recipient.
  • the method comprises the steps of combining a body fluid comprising a diagnostic marker with a stabilization agent for an incubation period, the stabilization agent capable of completely or substantially preventing the degradation or inactivation of the diagnostic marker, introducing the stabilized body fluid into a detection platform, combining a detection agent with the stabilized body fluid, determining an interaction characteristic of the detection agent and the diagnostic marker of the stabilized body fluid using a detection device in communication with a processor, comparing the interaction characteristic with a plurality of indexed interaction records contained on a database in communication with the processor to determine an identity and concentration of the detection marker, generating a report of the identity and concentration of the detection marker in the body fluid, and delivering the report to a recipient.
  • the detection platform may be selected from the group consisting of a microarray, a multi-well plate, a high-performance liquid chromato graph, and a mass spectrometer.
  • a microfluidic device for analyzing the presence or absence of the diagnostic marker to be detected in a body fluid is disclosed.
  • An embodiment of the microfluidic device comprising a container, the container housing a sample reservoir having a sample input port in fluid connection with at least one detector array, wherein the detector array comprises at least one detector comprising a reaction chamber comprising an immobilized capture molecule, the stabilizing agent contained within the sample reservoir, the stabilizing agent useful to completely or substantially prevent degradation or inactivation of the diagnostic marker, and a reagent contained within the detector array, the reagent capable of undergoing a colorimetric reaction or displaying an optically detectable signal and capable of reacting with the diagnostic marker.
  • An embodiment of the microfluidic device may further comprise at least one of a filter capable of separating at least one unwanted component from a body fluid, a results display, wherein a result from the detector array is visually detectable, a plurality of displays capable to visualizing a plurality of detected diagnostic markers, and a control display, wherein the detection of at least one control reagent by the at least one detector is visualized.
  • a filter capable of separating at least one unwanted component from a body fluid
  • a results display wherein a result from the detector array is visually detectable
  • a plurality of displays capable to visualizing a plurality of detected diagnostic markers
  • a control display wherein the detection of at least one control reagent by the at least one detector is visualized.
  • the system comprises a sample container having an opening and least one rigid chamber for containing a body fluid, the body fluid comprising at least one diagnostic marker, a stabilizing agent contained within the sample container, the stabilizing agent capable of completely or substantially preventing the degradation or inactivation of the diagnostic marker, and a securing device for closing the opening of the sample container, wherein the stabilizing agent is coupled to a solid support.
  • the stabilizing agent may also be adsorbed to the solid support.
  • Figs. 1A and B show a diagram of a detection system, according to at least one embodiment of the present disclosure
  • Figs. 2-4 shows a diagram of a method of producing a bead system, according to at least one embodiment of the present disclosure
  • Fig. 5 shows a flowchart of a method for creating a bead system, according to at least one embodiment of the present disclosure
  • Figs. 6A and B show a diagram of a test strip, according to at least one embodiment of the present disclosure
  • Fig. 6C show a diagram of a microarray, according to at least one embodiment of the present disclosure
  • Fig. 6D shows a diagram of a microfluidic system, according to at least one embodiment of the present disclosure
  • Fig. 6E shows a diagram of a sample collection device, according to at least one embodiment of the present disclosure
  • Fig. 6F shows a diagram of a sample collection device, according to at least one embodiment of the present disclosure
  • Fig. 6G shows a diagram of a sample container, according to at least one embodiment of the present disclosure
  • Fig. 6H shows a diagram of a sample container, according to at least one embodiment of the present disclosure
  • Fig. 7 shows a flowchart of a method of stabilizing diagnostic markers, according to at least one embodiment of the present disclosure
  • Fig. 8 shows a flowchart of a method analyzing a diagnostic markers, according to at least one embodiment of the present disclosure
  • Fig. 9 shows a flowchart of a method of improving biomarker availability, according to at least one embodiment of the present disclosure
  • Fig. 10 shows a flowchart of a method of identifying diagnostic markers of a disease state, according to at least one embodiment of the present disclosure
  • Fig. 11 shows a flowchart of a method for screening biomarkers, according to at least one embodiment of the present disclosure
  • Fig. 12 shows a flowchart of a method of stabilizing diagnostic markers, according to at least one embodiment of the present disclosure
  • Fig. 13 shows a an exemplary system framework, according to at least one embodiment of the present disclosure
  • Fig. 14 shows a graphical depiction of a-amylase inhibition according to at least one embodiment of the present disclosure
  • Fig. 15 shows a graphical depiction of a-amylase inhibition according to at least one embodiment of the present disclosure
  • Fig. 16 shows a graphical depiction of lysozyme inhibition according to at least one embodiment of the present disclosure
  • Fig. 17 shows a graphical depiction of a galactose oxidase screen according to at least one embodiment of the present disclosure
  • Fig. 18A shows a three dimensional plot of glycated Hemoglobin Ale (HbAlc) analyzed by high pressure liquid chromatography (HPLC) according to at least one embodiment of the present disclosure
  • Fig. 18B shows a ultraviolet (UV) image of HbAlc analyzed by HPLC according to at least one embodiment of the present disclosure
  • Fig. 18C shows a chromatogram of HbAlc analyzed by HPLC according to at least one embodiment of the present disclosure
  • Fig. 19A shows a three dimensional plot of HbAlc analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure
  • Fig. 19B shows a UV image of HbAlc analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure
  • Fig. 19C shows a chromatogram of HbAlc analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure
  • Fig. 20A shows a three dimensional plot of HbAlc analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure
  • Fig. 20B shows a UV image of HbAlc analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure
  • Fig. 20C shows a chromatogram of HbAlc analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure
  • Fig. 21 shows a comparison of chromatograms from HPLC analyzed HbAlc (Fig.
  • Fig. 22 shows a comparison of UV images from HPLC analyzed HbAlc (Fig. 22A), HbAlc + Saliva (Fig. 22B), and HbAlc + Saliva + Stabilizing mixture (Fig. 22C) according to at least one embodiment of the present disclosure;
  • Fig 23 shows a comparison of three dimensional plots from HPLC analyzed HbAlc (Fig. 23A), HbAlc + Saliva (Fig. 23B), and HbAlc + Saliva + Stabilizing mixture (Fig. 23C) according to at least one embodiment of the present disclosure
  • Fig. 24A shows a three dimensional plot of Cancer Antigen 19-9 (CA 19-9) analyzed by high pressure liquid chromatography (HPLC) according to at least one embodiment of the present disclosure
  • Fig. 24B shows a ultraviolet (UV) image of CA 19-9 analyzed by HPLC according to at least one embodiment of the present disclosure
  • Fig. 25 A shows a three dimensional plot of CA 19-9 analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure
  • Fig. 25B shows a UV image of CA 19-9 analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure
  • Fig. 26A shows a three dimensional plot of CA 19-9 analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure
  • Fig. 26B shows a UV image of CA 19-9 analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure
  • Fig. 27 shows a comparison of three dimensional plots from HPLC analyzed CA 19-9 (Fig. 27A), CA 19-9 + Saliva (Fig. 27B), and CA 19-9 + Saliva + Stabilizing mixture (Fig. 27C) according to at least one embodiment of the present disclosure;
  • Fig. 28 shows a comparison of UV images from HPLC analyzed CA 19-9 (Fig. 28 A), CA 19-9 + Saliva (Fig. 28B), and CA 19-9 + Saliva + Stabilizing mixture (Fig. 28C) according to at least one embodiment of the present disclosure;
  • Fig. 29 shows a graphical depiction of Peroxidase activity inhibition by an inhibitor cocktail according to at least one embodiment of the present disclosure
  • Fig. 30 shows a graphical depiction of Peroxidase activity inhibition by caffeine according to at least one embodiment of the present disclosure
  • Fig. 31 shows a graphical depiction of Peroxidase activity inhibition by 3-tert-butyl- hydroxyanisole according to at least one embodiment of the present disclosure
  • Fig. 32 shows a graphical depiction of Peroxidase activity inhibition by benzoic acid according to at least one embodiment of the present disclosure
  • Fig. 33 shows a graphical depiction of Peroxidase activity inhibition by aluminum potassium sulfate dodecahydrate according to at least one embodiment of the present disclosure
  • Fig. 34 shows a schematic of a diagnostic system, according to at least one embodiment of the present disclosure
  • Fig. 35 shows a graphical representation of an assay component, according to at least one embodiment of the present disclosure.
  • Fig. 36 shows a graphical depiction of a fluid collection device, according to at least one embodiment of the present disclosure.
  • compositions, systems, and methods for biomarker stabilization and analysis provide various compositions, systems, and methods for biomarker stabilization and analysis.
  • formulations are disclosed herein, which function to preserve the level of diagnostic markers, such as biomarkers, in body fluids, from enzymatic alteration, or degradation.
  • methods for the collection and analysis of body fluids are disclosed.
  • the methods and formulations disclosed herein can be used to improve the sampling and testing of a body fluid by conditioning the body fluids for the stabilization of specific biomolecules and/or drug metabolites.
  • body fluids includes fluids produced by the body, such as saliva, or fractions thereof, mucous secretions, tears, sweat, bile, semen, urine, vaginal secretions, exhalations, anal secretions, blood, plasma, serum and mixtures of thereof.
  • Body fluids may also comprise cancer cells, peripheral blood mononuclear cells, lymphocytes, lymph fluid, and other tissue secretions or fluid.
  • Saliva is clear, viscous fluid with a slightly alkaline pH and a pi range from 11.5-3.0. It is hypotonic, composed of about 99.5% water, and also contains ions (e.g., K + , Na + , Ca ,
  • Saliva has a complex "proteome”- 10 6 D glycoproteins to 1000D peptides. It contains secretory products of salivary glands, products of B cells, PMNs, epithelial cells, and bacteria.
  • Major e.g., parotid, submandibular, and sublingual
  • minor e.g., palatine and retromolar
  • saliva contributes to the composition of saliva, along with extraneous contributors such as gingival crevicular fluid, serum proteins, white blood cells and their byproducts, oral epithelial cells, oral bacteria, food debris and dissolved food components.
  • extraneous contributors such as gingival crevicular fluid, serum proteins, white blood cells and their byproducts, oral epithelial cells, oral bacteria, food debris and dissolved food components.
  • Saliva from different glands may differ in composition. For example, saliva from the parotid gland is dominated by serous secretory cells, whereas saliva from the submandibular and sublingual glands and minor glands are mixed serous or mostly mucous. There can also be qualitative and quantitative differences in saliva output that affect its composition. Glandular contribution to saliva is affected by level of gland activity. The amount of saliva secreted per minute or the salivary flow rate influences the concentration of the constituents as well as the proportion of the constituents from each of the three pairs of major salivary glands and the minor salivary glands.
  • conditioning of body fluids for the preservation of diagnostic markers may be accomplished through the use of stabilizing agents, which may be incorporated into one or more carrier vehicles, such as rinses, gums, beverages, and confectionaries.
  • stabilizing agents such as rinses, gums, beverages, and confectionaries.
  • specific rinses and/or pre-rinses specially formulated according to the specific molecule or molecules (the diagnostic marker) to be detected in the body fluid may contain a stabilization agent.
  • the use of rinses and pre- rinses of the present disclosure to condition the body fluid may enhance the sensitivity for detection of specific diagnostic markers for clinical diagnosis. Additionally, this process may improve the signal-to-noise ratio for a better diagnostic yield. Further, each rinse/pre-rinse can be specifically formulated to reduce or prevent false positive and false negative results.
  • the stabilizing agent may act to prevent/decrease the degradation, or reduction of activity, of diagnostic markers.
  • the stabilizing agent may be comprised of one or more Generally Regarded as Safe (GRAS) compound, as determined by the Food and Drug Administration of the United States of America, such as those listed in Table I.
  • GRAS Generally Regarded as Safe
  • An exemplary embodiment of a stabilization agent may act to stabilize a diagnostic marker, and/or to bind to a destructive component to prevent the destructive component from degrading or decreasing the activity of the diagnostic marker.
  • the stabilizing agent may comprise at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, or at least 25 compounds which acts to stabilize the diagnostic marker, and/or to bind to a destructive component to prevent the destructive component from degrading or decreasing the activity of the diagnostic marker.
  • the stabilizing agent of the present disclosure may be present in at a level of between about 200ppm to about 2,000 ppm, about 400ppm to about 1600 ppm, about 600ppm to about 1400 ppm, about 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.
  • Lima beans 8-10 kDa There are six different lima bean inhibitors.
  • Ovomucoids are the glycoprotein protease inhibitors
  • Soybeans considered the primary one. All of them bind
  • diagnostic marker may be any biological molecule whose presence, specific concentration, or specific activity may be indicative of a disease state or heath/lifestyle characteristic (which may collectively be referred to as a "condition").
  • indicator of a disease state refers to the presence, progression, prevention, remission, amelioration, or prognosis of a disease. This also includes, but is not limited to, the effects of a therapeutic on a disease.
  • the disease state analyzed through use of the diagnostic marker may in an exemplary embodiment be directed towards any cancer, metabolic disease (such as diabetes I or II), cardiovascular disease, or the predisposition of any of the same.
  • a disease state may also include the infection of the patient by a bacterial, virus, yeast, fungus, or parasite.
  • Bacterial infections may include a mammalian infection by any bacterial species.
  • Embodiments of the bacterial species may include, but are not limited to, Bacillus anthracis, Bacillus cereus, Staphylococcus aureus, Listeria monocytogenes, Streptococcus pneumoniae, Streptococcus pyogenes, Clostridium botulinum, Clostridium difficile, Clostridium perfringens, Clostridium tetani, Borrelia burgdorferi, Treponema pallidum, Chlamydia trachomatis, Chlamydophila psittaci, Corynebacterium diphtheriae, Mycobacterium tuberculosis, and Mycobacterium avium, Rickettsia prowazekii, Rickettsia rickettsii, Rickettsia typhi,
  • pseudomallei Neisseria gonorrhoeae, Neisseria meningitides, Campylobacter jejuni, Helicobacter pylori, Legionella pneumophila, Acinetobacter baumannii, Moraxella catarrhalis, Pseudomonas aeruginosa, Aeromonas sp., Vibrio cholerae, Vibrio parahaemolyticus, Thiotrichales sp., Haemophilus influenzae, Klebsiella pneumoniae, Proteus mirabilis, Yersinia pestis, Yersinia 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 DNA viruses, including the genus Parvoviridae, double-stranded DNA viruses, including the genus Papillomaviridae, Polyomaviridae, Poxviridae, Herpesviridae (ex. Herpes Simplex Virus 1, 2, 6, Varicella-zoster virus, Epstein-Barr virus EBV, Human cytomegalovirus, and human herpesvirus 7), single-stranded R A viruses, Reo- and Retroviruses (ex. Hepatitis B virus, Human immunodeficiency virus 1 and 2, and Human T-lymphotropic virus 1).
  • Fungal infections may include a mammalian infection by any bacterial species.
  • Embodiments of the fungal species, as used herein, may include, but are not limited to, Fusarium oxysporum, Pneumocystis jirovecii, Aspergillus spp., Coccidioides immitis/posadasii, Candida albicans, Filobasidiella neoformans, Trichosporon, Encephalitozoon cuniculi, Enterocytozoon bieneusi, Mucor circinelloides, Rhizopus oryzae, and Lichtheimia corymbifera.
  • Parasite infections may include a mammalian infection by any parasite.
  • Embodiments of the parasite may be, but are not limited to, Entamoeba histolytica, Babesia microti, Babesia sp. WA1, Trypanosoma cruzi, Taenia solium, Echinococcus granulosus, Leishmania braziliensis, L. donovani, L. tropica, Plasmodium falciparum, Plasmodium vivax, Plasmodium ovale and Plasmodium malariae, Plasmodium knowlesi, Paragonimus westermani, chistosoma sp., S. mansoni, S.
  • Exemplary embodiments of diagnostic markers include those listed in Table III.
  • exemplary embodiments for the diagnosis of type-II diabetes include, but are not limited to, Albumin, Alpha-1 antitrypsin (A1AT) - G Protein, Cystatin C, Cystatin A, alpha- 2-macroglobulin (A2MG), Uteroglobin, Transthyretin (TTR), Annexin Al, 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, Angiogenin, Annexin Al, B-cell activating factor (BAFF), B-cell lymphoma 2 (BCL2)-like 2, Beta Human chorionic gonadotropin, Cal5-3, Calcyclin, Calvasculin, Cathepsin D, Caveolin-1, Chromogranin A, Alpha-crystallin B chain (CRYAB), Endostatin, Eotaxin-2, Epithelial cell adhesion molecule (EpCAM), Ezrin, fatty acid binding protein 4 (FABP4), Galectin-3, ⁇ -glutamylcysteine ligase regulatory chain (GCLR), Gelsolin, Glucose 6-phosphate (G6P), Glycoprotein 130 (gpl30), Glutathione S-transferase Mu 1 (GSTM1), Hepsin, High-mobility group protein Bl (HMGB- 1),
  • BAFF B-cell activating
  • Diagnostic markers directed towards a bacterial or virus may include any surface or secreted antigen from the bacteria or virus, or a conserved nucleotide sequence.
  • diagnostic markers may include one or more of: 1,25 dihydroxy-vitamin D, 17-Hydroxyprogesterone, 25-hydroxy- vitamin D, Antineutrophil Cytoplasmic Antibodies, 5-Hydroxy Tryptamine, 5- hydroxyindoleacetic acid, Acetoacetate, Activated Partial Thromboplastin Time, Adrenocorticotropic Hormone, Alanine aminotransferase, Alanine transaminase, Albumin, Albumin-to-Creatinine ratio, Albumin/Globulin ratio, Alcohol, Aldolase, Aldosterone, Aldosterone and plasma renin activity, Aldosterone and Renin, Alkaline Phosphatase, Allergen-specific IgE, Alpha tryptase, Alpha- 1 Antitrypsin, Alpha- fetoprotein, Alphal- antitrypsin, Alzheimer biomarkers (including Tau protein and Amyloid Beta 42 peptid
  • Viral Load HAV
  • Vitamin B12 Vitamin B12 & Folate
  • Vitamin D Vitamin D2, Vitamin D3, Vitamin K, von Willebrand Factor, West Nile Virus, Westergren sedimentation rate, and Zinc Protoporphyrin.
  • 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-amylases 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 a-amylase. Further, the stabilizing agent may be active in a biological fluid, such as in saliva.
  • a stabilizing agent may comprise one or more of Fixanal® Buffer 6.0 (Sigma-Aldrich Co.), aluminum sulfate hydrate, aluminum hydroxide, bentonite, and aluminum potassium sulfate dodecahydrate.
  • Lysozyme also known as muramidase or N-acetylmuramide glycanhydrolase, is a glycoside hydrolase, that damage bacterial cell walls by catalyzing hydrolysis of 1,4-beta- linkages between N-acetylmuramic acid and N-acetyl-D-glucosamine residues in a peptidoglycan and between N-acetyl-D-glucosamine residues in chitodextrins. Lysozyme is abundant in a number of secretions, such as tears, saliva, human milk, and mucus. It is also present in cytoplasmic granules of the polymorphonuclear neutrophils (PMN).
  • PMN polymorphonuclear neutrophils
  • a stabilizing agent of the present disclosure may be an inhibitor of lysozyme. Further, the stabilizing agent may be active in a biological fluid, such as saliva.
  • a stabilizing agent may comprise one or more of Fixanal® Buffer 6.0 (Sigma-Aldrich Co.), bentonite, benzoic acid, and acetic acid.
  • Peroxidase are a class of oxidoreductase enzymes that catalyze the oxidation of a compound by the decomposition of hydrogen peroxide or an organic peroxide. These peroxidases may be found in many different bodily fluids. For example, saliva contains both salivary peroxidase (SPX) and myeloperoxidase (MPO). These peroxides may act on a number of different diagnostic markers found in bodily fluids to mask the detection or analysis of the markers.
  • SPX salivary peroxidase
  • MPO myeloperoxidase
  • the stabilizing agent may be an inhibitor peroxidase activity. Further, the stabilizing agent may be active in a biological fluid, such as saliva, blood, serum, or cancer cells.
  • a stabilizing agent may comprise one or more of aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate.
  • the stabilizing agent may preserve a diagnostic marker for insulin resistance and/or glucose intolerance.
  • the stabilizing agent may be active in a biological fluid, such as saliva.
  • a diagnostic marker may be one or more of a-hydroxybutyrate, 1-linoleoyl-GPC, palmitate, Glycine, 3-methyl-2-oxybutyrate.
  • a diagnostic marker may be any marker of early stage insulin resistance and glucose intolerance in a non- diabetic individual.
  • Galactose oxidase is a member of the family of oxidoreductaases, and has been shown to participate in galactose metabolism. Because of this activity, galactose oxidase may be used to detect mucin-like glycoproteins. These glycoproteins are a major component of mucus secreted by epithelial and glandular cells and are primarily responsible for the protective properties of the viscoeleastic mucous barrier. Mucins have been implicated in the process of cholesterol gallstone formation, and has been identified as having abnormal expression in some cancers.
  • the stabilizing agent may be an inhibitor of galactose oxidase. Further, the stabilizing agent may be active in a biological fluid, such as saliva.
  • a stabilizing agent may comprise one or more of aluminum potassium sulfate dodecahydrate, 3-tert-butyl- hydroxyanisole, benzoic acid, and acetic acid.
  • HbAlc Glycated hemoglobin
  • the stabilizing agent may be an inhibitor of HbAlc degradation. Further, the stabilizing agent may be active in a biological fluid, such as saliva.
  • a stabilizing agent may comprise one or more of acetic acid, aluminum hydroxide bentonite, aluminum sulfate hydrate, aluminum potassium sulfate dodecahydrate, benzoic acid, caffeine, and 3- tert-butyl-hydroxyanisole, or a combination thereof.
  • Cancer Antigen 19-9 (CA 19-9), which may in some instances be referred to as Cancer Angigen 19.9, Cancer antigen-GI or CA-GI, is an antigen associated with various cancers, such as prostate and colon cancer. At least one use of CA 19-9 may be see whether a pancreatic tumor is secreting antigen. If that is the case, then the levels may decrease when the tumor is treated, and they may rise again if the disease recurs.
  • the stabilizing agent may be an inhibitor of CA 19-9 degradation. Further, the stabilizing agent may be active in a biological fluid, such as saliva, blood, serum, or cancer cells.
  • a stabilizing agent may comprise one or more of aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate.
  • the components of a stabilizing agent may be present at or about equal amounts, such as for example 1 : 1 : 1 in a stabilizing agent with three active components.
  • the stabilizing agents may in at least one embodiment inhibit degradation or inactivation of a diagnostic marker by at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%o, at least about 70%>, at least about 75%, at least about 80%>, at least about 85%, at least about 90%, at least about 95%, or at least about 99%.
  • the stabilizing agent may, in at least one embodiment, inhibit degradation of a diagnostic marker for at least about 1 minute, at least about 5 minutes, at least about 10 minutes, at least about 15 minutes, at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 4 hours, or at least about 8 hours.
  • stabilizing agents may be incorporated into various carriers for their use with mammals.
  • Such carriers may include various rinses, gums, lozenge, mouthwash, beverages, confectionary, washes, or other applicable vehicles to deliver a stabilizing agent to the site of a diagnostic marker.
  • Embodiments of a carrier, such as a rinse, according to the present disclosure may also comprise one or more additional component which may include an anti-caking agent, a chemical preservative, an emulsifying agent, a nutrient and dietary supplement, a sequestrant, a stabilizer, an additive, a synthetic and flavoring substance. Exemplary embodiments of these components are listed herein (See section entitled “Additional Components for Rinse”). Further, the one or more additional component may be a substance which has been labeled as Generally Recognized As Safe (GRAS) by the Food and Drug Administration of the United States of America.
  • GRAS Generally Recognized As Safe
  • a carrier of the present application may comprise a liquid.
  • the liquid in at least one embodiment, is a non-toxic liquid. Further, the liquid may comprise water, a beverage containing water, or glycerin. Moreover, in at least one embodiment, a stabilizing agent may be at least partially dissolved in the liquid.
  • the following components in at least one embodiment of the rinse of the present disclosure, may include:
  • ANTI-CAKING AGENTS aluminum calcium silicate, calcium silicate, magnesium silicate, sodium calcium aluminosilicate, and tricalcium silicate.
  • CHEMICAL PRESERVATIVES ascorbic acid, ascorbyl palmitate, benzoic acid, butylated hydroxyanisole, butylated hydroxytoluene, calcium ascorbate, calcium propionate, calcium sorbate, caprylic acid, dilauryl thiodipropionate, erythorbic acid, gum guaiac, methylparaben, potassium bisulfite, potassium metabisulfite, potassium sorbate, propionic acid, propyl gallate, propylparaben, sodium ascorbate, sodium benzoate, sodium bisulfite, sodium metahisulfite, sodium propionate, sodium sorbate, sodium sulfite, sorbic acid, stannous chloride, sulfur dioxide, thiodipropionic acid, tocopherols.
  • EMULSIFYING AGENTS cholic acid, desoxycholic acid, diacetyl tartaric acid esters of (M)mono- and diglycerides, glycocholic acid, mono- and diglycerides, monosodium phosphate derivatives of above, propylene glycol, ox bile extract, taurocholic acid.
  • NUTRIENTS AND DIETARY SUPPLEMENTS alanine, arginine, ascorbic acid, aspartic acid, biotin, calcium carbonate, calcium citrate, calcium glycerophosphate, calcium oxide, calcium pantothenate, calcium phosphate, calcium pyrophosphate, calcium sulfate, carotene, choline bitartrate, choline chloride, copper gluconate, cuprous iodide, cysteine, cystine, ferric phosphate, ferric pyrophosphate, ferric sodium pyrophosphate, ferrous gluconate, ferrous lactate, ferrous sulfate, glycine, histidine, inositol, iron (reduced), isoleucine, leucine, linoleic acid, lysine, magnesium oxide, magnesium phosphate, magnesium sulfate, manganese chloride, manganese citrate, manganese gluconate, manganese glycerophosphat
  • SEQUESTRANTS calcium acetate, calcium chloride, calcium citrate, calcium diacetate, calcium gluconate, calcium hexametaphosphate, calcium phosphate, monobasic, calcium phytate, citric acid, dipotassium phosphate, disodium phosphate, isopropyl citrate, monoisopropyl citrate, potassium citrate, sodium acid phosphate, sodium citrate, sodium diacetate, sodium gluconate, sodium hexametaphosphate, sodium metaphosphate, sodium phosphate, sodium potassium tartrate, sodium pyrophosphate, sodium pyrophosphate, tetra, sodium tartrate, sodium thiosulfate, sodium tripolyphosphate, stecaryl citrate, tartaric acid.
  • STABILIZERS acacia (gum arabic) , agar-agar, ammonium alginate, calcium alginate, carob bean gum, chondrus extract, ghatti gum, guar gum, potassium alginate, sodium alginate, sterculia (or karava) gum, tragacanth.
  • ADDITIVES acetic acid, adipic acid, aluminum ammonium sulfate, aluminum potassium sulfate aluminum sodium sulfate, aluminum sulfate, ammonium bicarbonate, ammonium carbonate, ammonium hydroxide, ammonium phosphate, ammonium sulfate, bees wax, bentonite, butane, caffeine, calcium carbonate, calcium chloride, calcium citrate, calcium gluconate, calcium hydroxide, calcium lactate, calcium oxide, calcium phosphate, caramel, carbon dioxide, carnauba wax, citric acid, dextrans, ethyl formate, glutamic acid, glutamic acid hydrochloride, glycerin, glyceryl monostearate, helium, hydrochloric acid, hydrogen peroxide, lactic acid, lecithin, magnesium carbonate, magnesium hydroxide, magnesium oxide, magnesium stearate, malic acid, methylcellulose, monoammonium glutamate, monopotassi
  • SYNTHETIC FLAVORING SUBSTANCES acetaldehyde, acetoin, aconitic acid, anethole, benzaldehyde, N-butyric acid, d- or 1-carvone cinnamaldehyde, citral, decanal, diacetyl, ethyl acetate, ethyl butyrate, ethyl vanillin, eugenol, 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, 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.
  • MHC multimers including MHC- dextramers, MHC-tetramers, MHC-pentamers and other MHC- multimers
  • 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 BlueTM, Pacific OrangeTM, Cascade YellowTM; • 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, DsRed, Tl , Dimer2, mPvFPl, MBanana, mOrange, dTomato, tdTomato, mTangerine, mStrawberry, mCherry Tandem dyes: o RPE-Cy5, RPE-Cy5.5, RPE-Cy7, RPE-AlexaFluor® tandem conjugates;
  • RPE-Alexa610, RPE-TxRed o APC-Aleca600, APC-Alexa610, APC-Alexa750, APC-Cy5, APC-Cy5.5 Multi fluorochrome assemblies o Multiple fluorochromes attached to a polymer molecule, such as a peptide/protein, Dex, or poly-sacceride. o Any combination of the fluorescent dyes involving in generation of FRET;
  • the target of an exemplary indicator compound of the present disclosure may be any diagnostic marker or diagnostic condition for a disease state (or pre-disease state) of an individual.
  • the disease state may be diabetes, pre-diabetes, cardiac disease, 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, Kallikrein 3, Vascular endothelial growth factor A (VEGF), Calcitonin, Chromogranin A, Gastrin, SI 00 alpha chain, Somatostatin, Thyro globulin, V-erb-b2, cyckub-dependent kinase inhibitor 1 (p21), Breast Cancer Antigen 1 and 2 (BRCA1 and 2), MutL homolog 1 (MLH1), MutS homolog 2 (MSH2), MutS homolog 6 (MSH6), and postmeiotic segregation increased 1 and 2 (PMS1 and 2).
  • the target may be the presence or level of any diagnostic marker included herein.
  • the indictor signal in at least one embodiment of the present disclosure may comprise any detectable signal, including but not limited to, color change, fluorescence, and chemical/structural change of the target (and/or indicator compound) so as to be amenable to reacting with a secondary detection marker. Further, the detection of a signal may involve a secondary reactive molecule.
  • glucose may be detected through an indicator rinse by a chemical or enzymatic method.
  • exemplary embodiments of this method may include alkaline copper reduction, alkaline ferricyanide reduction, glucose oxidase, and/or hexokinase.
  • the rinse may further comprise one or more additional component as described herein.
  • additional components may include stabilizing agents, as well as GRAS components (such as those listed herein).
  • glycated Hemoglobin Ale may be detected through any known method for the detection of HbAlc or fragments thereof, such as high pressure liquid chromatography, immunoassays, and an indicator rinse by a chemical or enzymatic method.
  • the rinse may further comprise one or more additional component as described herein. These additional components may include stabilizing agents, as well as GRAS components (such as those listed herein).
  • cancer antigen 19-9 may be detected through any known method for the detection of CA 19-9 or fragments thereof, such as high pressure liquid chromatography, immunoassays, and an indicator rinse by a chemical or enzymatic method.
  • the rinse may further comprise one or more additional component as described herein. These additional components may include stabilizing agents, as well as GRAS components (such as those listed herein).
  • a diagnostic marker susceptible to degradation or alteration by a destructive component may be detected through any known method, including, but not limited to, high pressure liquid chromatography, immunoassays, and an indicator rinse by a chemical or enzymatic method. Further, the detection may be conducted in a micro well, deep well, microarray, microfluidic device, high throughput screen, or any applicable diagnostic device.
  • a detection platform 100 may comprise a platform 102 having a plurality of test wells 104, where each well is capable of containing a plurality of test spots 106.
  • the plurality of test wells 104 may include at least 2, at least 4, at least 8, at least 16, at least 32, at least 64, or at least 256 wells.
  • the plurality of tests spots 106 may include at least 2, at least 4, at least 8, at least 16, at least 32, or at least 64 test spots 106. Further, each of the plurality of test spots 106 may comprise one or more diagnostic marker or control marker.
  • An embodiment of detection platform 100 may further be sized and shaped to allow for automated detection and analysis of a diagnostic or control marker.
  • the detection system 100 may comprise an identifier 108, such as a barcode or Radio-frequency identification (RFID) tag, to allow for the identification of the detection system 100 and data collected from the analysis of test spots 106.
  • RFID Radio-frequency identification
  • detection platform 100 may be comprised of any applicable material sufficient to house or define test wells 104.
  • applicable materials for detection platform 100 may be a polymer, glass, metal, quartz, nylon, silica-based material, resin, or combination thereof.
  • embodiments of test wells 104 may include not only a depression defined or housed by platform 102, but may also include a defined area or region of the platform 102 (such as may be found on a microarray based platform).
  • An exemplary embodiment of detection system 150 comprises an embodiment of detection platform 100, a detection device 120 capable of determining a binding characteristic between a detection agent and a diagnostic marker on the detection platform 100, and a computer processor 130 coupled to a computer database 135 and to the detection device 120.
  • the computer processor 130 controlling the detection device 120 is able to (1) determine the binding characteristic of a detection agent to a diagnostic agent, (2) compare the binding characteristic among each of a plurality of samples tested, (3) generate a binding report using he compared binding characteristics, and deliver the binding report to a recipient or external computer (not shown) in communication with the computer processor 130.
  • a binding characteristic as used herein may include any measurement of the attraction (or repulsion) of two molecules (such as a detection agent and a diagnostic marker).
  • Detection system 100 may also comprise an embodiment of a bead-based system of the present disclosure having at least one bead type.
  • a dual bead system may be used wherein at least one bead type is magnetic or paramagnetic.
  • These beads may also be bound to a carrier protein with a conjugated hapten. Further, the beads may have an analyte capture monoclonal antibody reagent bound to it. Additionally, the beads, in at least one embodiment, do not retain their magnetic properties when removed from a magnetic field.
  • the beads may be Europium micro-particles. The sizes of the Europium micro-particles may be varied as needed to accommodate the method of detection used.
  • At least one embodiment of a bead based system may comprise a latex particle (See Figs. 2-4).
  • the latex particle may be a Carboxylate modified latex (CML) bead.
  • CML Carboxylate modified latex
  • an exemplary embodiment of a bead may include bovine serum albumin (BSA) and/or human serum albumin (HSA).
  • BSA bovine serum albumin
  • HSA human serum albumin
  • an embodiment of a bead based system of the present disclosure may include a linker coupled to HSA and/or BAS.
  • the linker according to an embodiment, may comprise a maleimide compound.
  • At least one exemplary embodiment of a maleimide compound may be Succinimidyl 4-[N-Maleimidomethyl] Cyclohexane-1 -Carboxylate (SMCC) or N- Hydroxysuccinimide-activated hexa(ethylene glycol) undecane Thiol (NHS).
  • SMCC Cyclohexane-1 -Carboxylate
  • NHS N- Hydroxysuccinimide-activated hexa(ethylene glycol) undecane Thiol
  • at least one exemplary embodiment of a bead based system may further comprise an antibody linked to maleimide on the bead.
  • at least one embodiment of the bead based system comprises a latex particle coupled to BAS or HSA, maleimide coupled to BAS or HSA, and an antibody coupled to maleimide.
  • the latex particle may be in a size range from about 0.02 ⁇ to about 7.0 ⁇ .
  • at least one at least one method 500 of coupling an antibody to a bead comprises the step 502 of modifying an embodiment of a bead (such as a latex bead/particle) with BSA or HAS, wherein the BSA or HSA may further comprise a maleimide containing molecule, such as SMCC or NHS.
  • an embodiment of the method 500 may further comprise the step 504 of attaching a diagnostic marker binding agent (such as an antibody) to the bead.
  • Step 504 of attaching the diagnostic marker binding agent to the bead may be accomplished through binding the diagnostic marker binding agent to maleimide.
  • method 500 may also comprise the step 506 of attaching one or more stabilizing agent to an embodiment of a bead.
  • an embodiment of method 500 may use latex beads for attachment of a maleimide containing molecule and/or a stabilizing agent, alternate embodiments of the bead may comprise silicon, a polymer, or other applicable materials.
  • one or more stabilizing agent may be attached or adsorbed to any one of an embodiment of a detection platform, test strip, microchip, or micro fluidic device.
  • An exemplary embodiment of diagnostic testing device 600 comprises housing structure 602 which comprises a collection chamber 604 capable of receiving body fluid, at least one membrane strip 606 in fluid communication with collection chamber 604, an immunoassay-based fingerprint acquisition pad 608 in fluid communication with the collection chamber 604, and a plurality of reaction zones 610 which may allow the visual display of the presence of a predetermined diagnostic marker.
  • reaction zone 610 may in an exemplary embodiment be capable of displaying a characteristic of the predetermined diagnostic marker, such as the presence of, the concentration of, or a concentration above or below a predetermined value for the predetermined diagnostic agent.
  • diagnostic testing device 600 further comprises a fluid collector 612 which is operable to collect body fluid from a subject, and wherein the fluid collector is capable of supply collected body fluid to collection chamber 604.
  • test strip 606 may comprise any material capable of adsorbing or attaching a stabilizing agent and may bind a diagnostic marker. Further, exemplary embodiments of test strip 606 may comprise compositions comprising a polyvinyl chloride-silica combination, nitrocellulose, or any suitable synthetic, resinous material. At least one embodiment, housing 602 is capable of reducing the exposure of the test strip 606 at sites other than the collection chamber 604 to foreign material. Test strip 606 in at least one embodiment may be capable of interfacing with an analysis device 615. The analysis device 615 in an exemplary embodiment may be capable of measuring the level of the predetermined diagnostic marker in the sample.
  • An exemplary embodiment of microarray system 620 may comprise a microarray product 622 having a microarray identifier 624, and a plurality of microarray sites 626, a control microarray product 628 located on at least one microarray site 626 and bound to microarray product 622, and operably connected to a computer processor 130 for providing information regarding the identification and concentration of markers on the microarray product 622 based on the microarray identifier 624.
  • microarray product 622 may further comprise, in at least one embodiment, an embodiment of a stabilization agent 630 located on at least one microarray site 626 and bound to microarray product 622.
  • microarray product 622 may comprise one or more diagnostic marker 632 located on at least one microarray site 626 and bound to microarray product 622.
  • microarray system 620 in at least one embodiment, may further comprise a microarray detector 634 operable to detect a signal from at least one of the control microarray product 628 and/or diagnostic marker 632.
  • a microchip or microarrays may refer to an array of distinct polynucleotides affixed to a substrate, such as glass, plastic, paper, nylon or other type of membrane, filter, chip, or any other suitable solid support.
  • the polynucleotides can be synthesized directly on the substrate, or synthesized separate from the substrate and then affixed to the substrate.
  • the microarray may be prepared and used at least according to the methods described in U.S. Pat. No. 5,837,832, Chee et al, PCT application W095/11995 (Chee et al), Lockhart, D. J. et al. (1996; Nat. Biotech. 14: 1675-1680) and Schena, M. et al. (1996; Proc. Natl. Acad. Sci. 93: 10614-10619), all of which are incorporated herein in their entirety by reference.
  • microfluidic device 640 comprises a sample reservoir 642 for receiving a body fluid (or other fluid sample) through a sample input 644, where the sample reservoir 642 is fluidly connected to a detector array 646. Additionally, in at least one embodiment of microfluidic device 640, detector array 646 comprises a reaction site 647 and a results display 648. Moreover, an exemplary embodiment of microfluidic device 640 may further comprise a filter device 650 capable of separating at least one unwanted component from a fluid sample and fluidly coupled between sample reservoir 642 and detector array 646. Detector array 646 may also, in at least one embodiment comprise a control reagent display 652, where at least one control reagent may be visually detected.
  • microfluidic device 640 may also, in at least one embodiment, be able to couple to a processor 655.
  • Processor 655 may be able to compare at least one reading, such as from results display 648 or control reagent display 652 from an embodiment of microfluidic device 640 to at least one additional stored reading on a computer database 657 of the processor 655.
  • An embodiment of microfluidic system 660 of the present disclosure may comprise one or more microfluidic device 640 operationally coupled to a processor 655.
  • Embodiments of microfluidic devices 640 which may also be referred to as "lab-on-a- chip” systems, biomedical micro-electro-mechanical systems (bioMEMs), or multicomponent integrated systems, are exemplary kits/systems for analyzing polynucleotide regions. Such systems may miniaturize and compartmentalize processes such as probe/target hybridization, nucleic acid amplification, and capillary electrophoresis reactions in a single functional device. Such microfluidic devices typically utilize detection reagents in at least one aspect of the system, and such detection reagents may be used to detect one or more polynucleotide region of the present disclosure.
  • Exemplary microfluidic devices 640 may comprise a pattern of microchannels designed onto a glass, silicon, quartz, or plastic wafer included on a microchip.
  • the movements of the samples may be controlled by electric, electroosmotic or hydrostatic forces applied across different areas of the microchip to create functional microscopic valves and pumps with no moving parts. Varying the voltage can be used as a means to control the liquid flow at intersections between the micro-machined channels and to change the liquid flow rate for pumping across different sections of the microchip.
  • the microarray or microfluidic device may comprise one or more stabilizing agent capable of reducing the degradation of a predefined diagnostic marker.
  • a stabilizing agent may be incorporated onto the surface of the microarray device, and/or on at least one part of the microfluidic device.
  • a sample collection device 665 of the present disclosure comprises an intake tube (or first tube) 666 having a first end and a second end, where the second end is coupled to a suction device 667.
  • Suction device in at least one embodiment is also coupled to a dispensing tube 668 (or second tube) having a first and second end at the first end.
  • intake tube 666 comprises a filter 669 capable of removing at least one component of a body fluid before entering dispensing tube 668.
  • sample collection device 665 may also comprise securing devices 670 to fluidly seal either, or both of, the first end of the intake tube 666 and the second end of the dispensing tube 668.
  • securing devices 670 to fluidly seal either, or both of, the first end of the intake tube 666 and the second end of the dispensing tube 668.
  • an embodiment of sample collection device 665 may also comprise an effective amount of an embodiment of a stabilizing agent of the present disclosure.
  • device 665 may be operated by placing device 665 in contact with a fluid source, using the suction device 667 to provide negative pressure so as to pull a portion of the fluid source into sample collection device 665.
  • the portion of the fluid source may pass through filter 669 where at least one component of the body fluid may be removed.
  • the process of intake of the portion of fluid source may also initiate the contact of the of the portion of the fluid source with a stabilization compound housed within the sample collecting device. The stabilized portion of the body fluid may then be dispensed in a controlled fashion through the dispensing tube 668 by positive pressure provided by suction device 667.
  • the dispensing tube 668 is structured so as to deliver a predetermined amount of liquid per drop.
  • the predetermined amount of liquid per drop may be ⁇ , 30 ⁇ 1, 50 ⁇ 1, or ⁇ in at least one embodiment of sample device 665.
  • FIG. 6F at least one embodiment of fluid collection device 672 is shown.
  • sample collection device 672 comprises a housing 673 having a top and bottom opening (not shown), a tubular compartment 675 within housing 673 and in fluid communication with both top and bottom openings, a swab portion 674 coupled to the bottom opening and in fluid communication with tubular compartment 675, and a stabilizing agent contained within the tubular compartment 675 .
  • fluid collection device 672 may also comprise a sealing mechanism 680 capable of fluidly sealing the tubular compartment 675 and one or both of the top and bottom openings.
  • housing 673 may comprise a sample compartment 675 coupled to the top and bottom openings, and a stabilization compartment 676 containing an embodiment of stabilization agent 677.
  • compartments 675 and 676 are connected by mixing tube 678, but fluid communication is prevented by removable barrier 679.
  • Barrier 679 may be operable to allow fluid communication between compartments 675 and 676 upon mechanical or electrical activation by a user.
  • an embodiment of a stabilization agent 677 may also be provided in fluid collection device 672 in at least one of swab portion 674 and sample compartment 675.
  • transport system 682 comprises an inner container 684 contained within outer container 685.
  • Inner container 684 in an exemplary embodiment, has an inner compartment 686 and an opening 687.
  • Inner compartment 686 may be sized and shaped to be capable of receipt of a sample through opening 687.
  • inner compartment 686 may comprise a solid support 689, where solid support 689 comprises or has attached an embodiment of a stabilizing agent 690.
  • Inner container 686 may be reversibly sealed to prevent liquid release from the inner compartment 686.
  • transport system 682 may further comprise an absorbent material 691 contained within outer container 685 and sufficient to absorb the liquid contents of inner container 684 in the event of an unexpected release of the liquids contained therein.
  • transport system 682 is structured in such a manner, and stabilizing agent 690 contained therein has the properties, to prevent the degradation of a diagnostic agent in a liquid sample stored within the inner container 684 for a period of at least twenty-four hours.
  • stabilizing system 692 comprises a sample container 694 having an opening 695 and at least one rigid chamber 696 for containing a fluid sample. Additionally, stabilizing system 692 may also contain an effective amount of an embodiment of stabilizing agent 697 that may be adsorbed to, or part of, solid support 698. Stabilizing system 692 may be reversibly sealed with securing device 699 that may seal the opening 695.
  • the method 700 comprises the step 702 of mixing a body fluid with an embodiment of a stabilizing agent.
  • the step of mixing a body fluid with a stabilizing agent may comprise the step 704 of introducing the stabilizing agent into the patient, such as through the oral cavity, so that the stabilizing agent mixes with the bodily fluid, and the step 706 of retrieving a body fluid comprising a stabilized diagnostic marker.
  • the method comprises the step 708 of isolating a body fluid having a diagnostic marker, the step 710 of treating the body fluid with an embodiment of a carrier, such as a rinse, gum, or beverage, as described herein, and the step 712 of analyzing the diagnostic marker.
  • Isolation of the body fluid may occur through any customary mechanism, including, but not limited to, rinsing, swabbing, suction, collection, and lavage.
  • the rinse in an exemplary embodiment of a method 700 of the present disclosure may be ingested to stabilize a diagnostic marker present in the bodily fluid, such as in urine, semen, anal secretions, and vaginal secretions.
  • the step 702 of mixing a body fluid with a carrier may occur prior to, or after isolation step 708 of the body fluid.
  • a rinse may be used to rinse the mouth.
  • the rinse containing the treated body fluid may be collected by spitting, suction, or other means.
  • expirated saliva may be mixed with a rinse to treat the saliva ex vivo. Similar treatment of body fluids may be performed with any type of body fluid.
  • the diagnostic marker may be analyzed through any known means.
  • the analysis of the treated body fluid may include the separation of solid materials from soluble materials through means such as filtration, or by centrifugation.
  • Analysis of the treated body fluid may use any appropriate technique, such as western blot analysis, Enzyme-Linked Immunosorbent Assay (ELISA), protein activity assays, reverse transcription polymerase chain reaction (RT-PCR), microarray, high pressure liquid chromatography, or any comparable assay to determine a characteristic of the diagnostic marker.
  • ELISA Enzyme-Linked Immunosorbent Assay
  • RT-PCR reverse transcription polymerase chain reaction
  • microarray a microarray
  • high pressure liquid chromatography or any comparable assay to determine a characteristic of the diagnostic marker.
  • Such an analysis in an exemplary embodiment, may be of a modified product, a cleavage product, or cleavage pattern, of a diagnostic marker.
  • the treated body fluid may be placed in contact with an embodiment of a test strip, such as a nitrocellulose strip, prior to detection with an appropriate probe specific for the diagnostic marker.
  • a test strip such as a nitrocellulose strip
  • the nitrocellulose strip may be designed to trap or filter particulates in the body fluid. This may reduce or eliminate potential contaminants or other substances in the oral fluid that would otherwise reduce the signal to noise ration during the detection step.
  • a probe (such as an antibody) with affinity to the diagnostic marker may be affixed or immobilized to a specific location on the nitrocellulose membrane for detection of the analyte. Utilizing standard principles of immunoassay detection (or nucleotide detection), the signal generated may be visible to the eye or detectable by an instrument.
  • the method 800 comprises the step 802 of operating a system for the detection of a diagnostic marker in a body fluid, where the system comprises a diagnostic device having a plurality of test wells each capable of containing at least one diagnostic marker binding agent, and a detection device capable of interacting with diagnostic device, wherein the detection device is capable of detecting an interaction between the at least one diagnostic marker binding agent and a diagnostic marker.
  • the sample of bodily fluid may be diluted in step 803 with a reagent which may contain an embodiment of stabilizing agent as described herein.
  • At least one embodiment of method 800 further comprises the steps of: contacting 804 a sample of a bodily fluid with at least one diagnostic marker binding agent, incubating 806 the contacted sample in the test well at a first temperature for a pre-determined period of time.
  • the step 804 of contacting of the sample of bodily fluid with at least one diagnostic marker binding agent may be accomplished through a magnetic field applied to the test well.
  • the first temperature may be at room or at body temperature (such as about 22°C or about 37°C).
  • the pre-determined period of time may, for example, be between five and fifteen minutes.
  • the incubated and contacted sample may then be removed in step 808 from the plurality of test wells, and the test wells analyzed in step 810 to detect a binding event with the detection device.
  • a stabilizing agent may be incubated with the diagnostic marker binding agent (also referred to as a detection agent) prior to introduction of a bodily fluid.
  • each diagnostic marker binding agent may have a different monoclonal antibody designed to bind a specific hapten molecule.
  • the characteristic may in step 812 be compared to a standard value to determine the presence or absence of a disease state.
  • the characteristic determined may be an activity level, the concentration of the diagnostic marker, or a particular modification, such as glycosylation or methylation.
  • an embodiment of the method 800 may further comprise the step 814 of customizing a detection method through screening of applicable pre-clinical samples through a database of stabilizing agents/cocktails.
  • an embodiment of method 800 of diagnostic marker detection may further comprise the step 816 of comparing a level or characteristic (such as a predictable degradation products) of a diagnostic agent with a library of known characteristics.
  • an exemplary embodiment of method 800 may additionally comprise the step 818 of determining a diagnosis of a disease state using the compared characteristic.
  • a method of diagnostic marker detection may be automated, and capable of detecting at least about ten thousand samples in a twenty-four hour period.
  • Method 800 in at least one embodiment, may be at least partially completed by a computer processor.
  • the processor may be in communication with at least one more processor and/or a computer database. For instance, in at least one embodiment of method 800, most of the steps of the method may be completed with or monitored by a computer processor. Moreover, method 800, in at least one embodiment, may use a computer processor to perform additional step 820 of generating a report by using a comparison of determined characteristics (such as a binding report where a comparison is performed of binding characteristics), and step 822 of delivering the report to a recipient (such as a user, or secondary processor).
  • a computer processor may use a computer processor to perform additional step 820 of generating a report by using a comparison of determined characteristics (such as a binding report where a comparison is performed of binding characteristics), and step 822 of delivering the report to a recipient (such as a user, or secondary processor).
  • a method of biomarker stabilization may, in an exemplary embodiment, may be used to diagnose a particular disease state or condition of a patient.
  • a disease state or condition as used with this method may include one or more of Acid-Base Disorders, Acidosis and Alkalosis, Acidosis/Alkalosis, Acute inflammatory demyelinating polyneuropathy, Acute myocardial infarct, Addison's Disease, Adrenal Insufficiency, Adrenal Insufficiency & Addison's Disease, Alcohol dependence, Alcoholism, Allergies, Alzheimer's Disease, Anemia, Angina Pectoris, Anthrax, Arthritis, Asthma, Atypical Pneumonia, Autoimmune Disorders, Autoimmune thyroiditis, Avian flu, Benign Prostatic Hyperplasia, Benign Prostatic Hypertrophy, Bioterrorism Agents, Bleeding Disorders, Bone Marrow Disorders, Breast Cancer, Cardiovascular Disease, Celiac Disease, Cerv
  • Method 900 comprises the step 902 of contacting a diagnostic marker from a patient with a stabilization agent, the step 904 of introducing a therapeutic agent to the diagnostic marker and the stabilization agent, and the step 906 of analyzing the interaction of the therapeutic agent with the diagnostic marker to determine a therapeutic profile, such as the binding affinity of the agent to the diagnostic marker. Further, method 900 may also comprise the step 908 of comparing the therapeutic profile with at least one other therapeutic profile to determine a stabilizing impact from the stabilizing agent on the diagnostic marker.
  • Step 908 of comparing a plurality of therapeutic profiles may be performed using a computer processor and may further utilize a computer database of previously documented therapeutic profiles. Moreover, in an exemplary embodiment of method 900, the step 902 of contacting a diagnostic marker with a stabilization agent may be repeated with a plurality of different stabilization agents and/or therapeutic agents. Additionally, the therapeutic profile may be used to determine a therapeutic course to treat the patient.
  • Method 1000 comprises the step 1010 of contacting a bodily fluid from a subject having a disease state with a stabilizing agent, and the step 1020 of analyzing the stabilized bodily fluid to determine at least one property of a diagnostic agent, to create a disease state profile.
  • An exemplary embodiment of method 1000 may further comprise the step 1030 of comparing the disease state profile with a profile generated in a like manner from a subject not having the disease state, and the step 1040 of generating a disease state identifier panel from the compared disease state profile.
  • the disease state is cancer.
  • Exemplary embodiments of cancer may include at least bladder cancer, lung cancer, breast cancer, melanoma, colon and rectal cancer, non-hodgkin lymphoma, endometrial cancer, pancreatic cancer, kidney (renal cell) cancer, prostate cancer, leukemia, and thyroid cancer.
  • the subject is currently receiving treatment with an original treatment agent, and the disease state profile may be compared with a disease profile of the same subject while receiving a different treatment agent, a different concentration of the same treatment agent, or at an earlier point of time.
  • Method 1100 comprises the steps of (a) contacting 1110 a cell from a subject having a disease state with a stabilizing agent, (b) incubating 1120 the stabilized cell with a therapeutic compound, and (c) analyzing 1130 the incubated cell for at least one diagnostic marker to create a marker profile.
  • method 1100 may further comprise the step 1140 of comparing the marker profile with at least one additional marker profile to determine the efficacy of the therapeutic compound.
  • the method 1200 comprises the step 1210 of measuring the level of diagnostic marker in a subject with a disease state, the step 1220 of treating the subject with a therapeutic agent, and the step 1230 of determining the post-treatment level of diagnostic marker.
  • the step 1210 of measuring the level of diagnostic marker includes the step of incubation of the diagnostic marker with a stabilization agent.
  • the method may be used with a device as described herein, such as an embodiment of diagnostic system 100, test strip 600, a microarray 640, or microfluidic device 660, any of which may also be performed in conjunction with a computer processor.
  • a device as described herein, such as an embodiment of diagnostic system 100, test strip 600, a microarray 640, or microfluidic device 660, any of which may also be performed in conjunction with a computer processor.
  • a computer processor and a database may be used in or during the system, method, or device.
  • An exemplary embodiment of a system framework comprising at least one computer processor and at least one database, as may be used in the embodiments of the present disclosure is shown in Fig. 13.
  • a result from a diagnostic method may be compared using a computer processor to at least one additional result from the diagnostic test, and/or to a result stored on a database.
  • Such a comparison in at least one embodiment of the present disclosure, may reveal the stabilizing agent with a greater effect on the binding characteristic, may allow for the diagnosis of a disease state or health/lifestyle characteristic. Further, such a comparison, in at least one embodiment, may be used to develop, confirm, or modify a therapeutic treatment of a patient having a disease state or health/lifestyle characteristic.
  • one or more user computers 1302 may be operably connected to a system server 1304.
  • a user computer 1302 may be a computer, computing device, or system of a type known in the art, such as a personal computer, mainframe computer, workstation, notebook computer, laptop computer, hand-held computer, wireless mobile telephone, personal digital assistant device, and the like.
  • One or more administrator computers 1306 may also be operably connected to system server 1304 including through a network 1308 such as the Internet.
  • Administrator computers 1306, similar to user computers, may be computers, computing devices, or systems of a type known in the art, such as personal computers, mainframe computers, workstations, notebook computers, laptop computers, hand-held computers, wireless mobile telephones, personal digital assistant devices, and the like.
  • user computers and administrator computers may each comprise such software (operational and application), hardware, and componentry as would occur to one of skill of the art, such as, for example, one or more microprocessors, memory, input/output devices, device controllers, and the like.
  • User computers and administrator computers may also comprise one or more data entry means (not shown in Fig.
  • User computers and administrator computers operable by a user of client computer and/or an administrator computer, such as, for example, a keyboard, keypad, pointing device, mouse, touchpad, touchscreen, microphone, and/or other data entry means known in the art.
  • User computers and administrator computers also may comprise an audio display means (not shown in Fig. 13) such as one or more loudspeakers and/or other means known in the art for emitting an audibly perceptible output.
  • the configuration of user computers and administrator computers in a particular implementation of one or more systems of the present disclosure is left to the discretion of the practitioner.
  • System server 1304 may comprise one or more server computers, computing devices, or systems of a type known in the art.
  • System server 1304 may comprise server memory.
  • System server 1304 may comprise one or more components of solid-state electronic memory, such as random access memory.
  • System server 1304 may also comprise an electromagnetic memory such as one or more hard disk drives and/or one or more floppy disk drives or magnetic tape drives, and may comprise an optical memory such as a Compact Disk Read Only Memory (CD-ROM) drive.
  • System server 1304 may further comprise such software (operational and application), hardware, and componentry, as would occur to one of skill of the art, such as, for example, microprocessors, input/output devices, device controllers, video display means, and the like.
  • System server 1304 may comprise one or more host servers, computing devices, or computing systems configured and programmed to carry out the functions allocated to system server 1304.
  • System server 1304 may be operated by, or under the control of, a "system operator," which may be an individual or a business entity.
  • System server 1304 is shown in Fig. 13 and referred to herein as a single server.
  • System server 1304 need not, however, be a single server.
  • System server 1304 may comprise a plurality of servers or other computing devices or systems connected by hardware and software that collectively are operable to perform the functions allocated to the various systems of present disclosure.
  • system server 1304 may be operable to be a web server, configured and programmed to carry out the functions allocated to a system server according to the present disclosure.
  • user computers 1302 and administrator computers 1306 may be connected directly to system server 1304, these computers may be connected to system server 1304 through any suitable network, such as network 1308. Further, in one embodiment, the users need not be provided access to system server 1304, but instead the content posts from users are made by the user(s) and saved to one or more particular locations and the posts are accessed or harvested by the administrator or system automatically.
  • System server 1304 may be operably connected to the various user computers 1302 and/or an administrator computers 1306 by network 1308, which in an embodiment of the present disclosure comprises the Internet, a global computer network.
  • network 1308 need not comprise the Internet.
  • Network 1308 may comprise any means for electronically interconnecting system server 1304 and a user computer 1302 and/or an administrator computer 1306.
  • the network 1308 may comprise the Internet, the commercial telephone network, one or more local area networks, one or more wide area networks, one or more wireless communications networks, coaxial cable, fiber optic cable, twisted-pair cable, the equivalents of any of the foregoing, or the combination of any two or more of the foregoing.
  • network 1308 comprises the hardware and software means interconnecting system server 1304 and user computer 1302 and/or an administrator computer 1306 within the single computing device.
  • Network 1308 may comprise packet switched facilities, such as the Internet, circuit switched facilities, such as the public switched telephone network, radio based facilities, such as a wireless network, etc.
  • the various systems, methods, schema, ontologies, and architectures of the present disclosure may be used for purposes outside of the medical transcription field as referenced in the various examples cited herein.
  • the system for analyzing verbal records may comprise various components and relationships suitable for use in any number of areas where various experiences are utilized and processed, with feedback being fed back into system componentry to improve overall system outcomes.
  • various components described herein may share a name (or a portion thereof) but have duplicative reference numbers, and therefore the descriptions for the various components should read in view of one another.
  • such systems may be operable, as desired by a user of such systems, to generate visual, electronic (video, audio, database, transcript, etc.), and/or printed reports, outputs, outcomes, and the like.
  • Such exemplary outputs may be used for any number of purposes, and may be useful generally to "report" results, data, and/or knowledge contained within and generated from such systems.
  • the disclosure of the present application further encompasses uses of the various methods, systems, architectures, etc., to perform various tasks in connection therewith.
  • At least one assay used for the detection of a-amylases activity includes the use of a chromagenic substrate, 2-chloro-p-nitrophenol linked to maltotriose. Enzymatic activity of a-amylase on the substrate yields 2-chloro-p-nitrophenol, which can be measured spectrophotometrically at 405nm. The amount of a-amylase activity present in the experimental sample is directly proportional to the increase in absorbance at 405nm.
  • test compounds are mixed with saliva, prior to the addition of a- amylase substrate.
  • the test compounds, such as GRAS materials are prepared at a concentration of 5,000 ppm in distilled water.
  • a- amylase substrate (Salimetrics a-amylase Assay Kit) that has been pre-heated to 37°C is added to 20 ⁇ 1 of the test compound.
  • ⁇ of a dilute (3.5%) or full strength (100%) saliva solution is added to the mixture.
  • each mixture is measured at 1 minute intervals using a spectrophotometer (such as Molecular Devices M5 reader) at 405nm. Temperatures of the mixtures are kept constant at 37°C during the assay.
  • CDI-034 and CDI-42 were shown to inhibit a-Amylase in 100% saliva samples (See Fig. 15).
  • At least one assay used for the detection of a-amylases activity includes the use of a Micrococcus lysodeikticus labeled with fluorescein.
  • the assay measures lysozyme activity on Micrococcus lysodeikticus cell walls, which are labeled to such a degree that the fluorescence is quenched. Lysozyme activity can relieve this quenching; yielding increased fluorescence that is proportional to lysozyme activity.
  • test compounds are mixed with saliva, prior to the addition of lysozyme substrate.
  • the test compounds such as GRAS materials, are prepared at a concentration of 5,000 ppm in distilled water.
  • 50 ⁇ 1 of lysozyme substrate at lmg/ml (Molecular Probes EnzChek Lysozyme Assay Kit) that has been pre-heated to 37°C is added to 50 ⁇ 1 of the test compound.
  • 50 ⁇ 1 of a dilute (3.5%) or full strength (100%) saliva solution is added to the mixture.
  • each mixture is measured at 1 minute intervals using a spectrophotometer (such as Molecular Devices M5 reader) for absorption at 494 nm and fluorescence emission at 518 nm. Temperatures of the mixtures are kept constant at 37°C during the assay.
  • a spectrophotometer such as Molecular Devices M5 reader
  • At least one assay used for the detection of galactose oxidase activity includes the use of a chromagenic or fluorogenic substrate, such as with Amplex® Red
  • galactose oxidase catalyzes the oxidation of galactose at the C6 position and generates hydrogen peroxide (H202).
  • H202 then, in the presence of horseradish peroxidase (HRP), reacts with 1 : 1 stoichiometry with Amplex® Red reagent to generate the red-fluorescent oxidation product, resorufin.
  • Resorufin has absorption and fluorescence emission maxima of approximately 571 nm and 585 nm, respectively, and because the extinction coefficient is high (54,000 cm-lM-1), the assay can be performed either fluorometrically or spectrophotometrically.
  • test compounds are mixed with Amplex Red, reaction buffer, horseradish peroxidase (HRP) at lOOU/ml, and galactose stock solution (according to manufacturers recommended protocol, Molecular Probes A22179), prior to the addition of saliva.
  • the test compounds such as GRAS materials, are prepared under various concentration for testing. Following this preparation, 50 ⁇ 1 of the test compound solution (with Amplex Red, reaction buffer, and HRP) is added to 50 ⁇ 1 of saliva. Following the initiation, each mixture is measured at 1 minute intervals using a spectrophotometer (such as Molecular Devices M5 reader) using excitation in the range of 530-560nm and emission detection at about 590nm or absorbance at 560nm.. Temperatures of the mixtures are kept constant at 37°C during the assay.
  • At least one procedure used for the stabilization and detection of glycosylated hemoglobin includes the incubation of HbAlc with a stabilizing agent prior to, or concurrently with, detection.
  • Analysis of samples was conducted on a HPLC 1100 series (Agilent Technologies) with a 2.1X150 (3.5 ⁇ ) CI 8 reverse phase column. The mobile phase used was acetonitrile with 6.5mM Ammonium Carbonate.
  • a HbAlc standard was prepared by dissolving HbAlc (Sigma Aldrich, 405 RM) with 1.0ml of deionized water.
  • Fig. 18 100ml HbAlc mixed with 100ml of fresh saliva
  • Fig. 19 200ml HbAlc mixed with 100ml of fresh saliva and 100ml of a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1 : 1 : 1 ratio)
  • a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1 : 1 : 1 ratio)
  • Fig. 20 While the pure HbAlc sample was analyzed fresh, samples of HbAlc with saliva were incubated overnight at room temperature prior to analysis. For each sample analyzed, the data was displayed through a three dimensional plot (Figs 18 A, 19A, and 20A), a UV image (Figs. 18B, 19B, and 20B), and a chromatogram (Figs. 18C, 19C, and 20
  • a series of GRAS compounds were tested, as shown in Figs. 18-20, for the ability to protect HbAlc from degradation by components in saliva.
  • the tested compounds are included in Table I.
  • a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1 :1 : 1 ratio) minimized the degradation of HbAl c, and yielded a defined pattern for HbAl c.
  • At least one procedure used for the stabilization and detection of Cancer Antigen 19-9 includes the incubation of CA 19-9 with a stabilizing agent prior to, or concurrently with, detection. Analysis of samples was conducted on a HPLC 1100 series (Agilent Technologies) with a 2.1X150 (3.5 ⁇ ) CI 8 reverse phase column. The mobile phase used was acetonitrile with 6.5mM Ammonium Carbonate. A CA 19-9 standard was prepared by dissolving CA 19-9 with 1.0ml of deionized water. The samples analyzed by HPLC included pure CA 19-9 (Fig. 24), 100ml CA 19-9 mixed with 100ml of fresh saliva (Fig.
  • test compounds such as GRAS materials or cocktails of same, were prepared at a concentration of 500ppm in distilled water.
  • 50 ⁇ samples of test compounds were mixed with 20-30 ⁇ of saliva (either “pooled” or “unpooled") prior to the addition of Amplex® Red.
  • 50 ⁇ 1 of Amplex® Red/HRP working solution See Protocol for Invitrogen Assay #A22188, which is incorporated herein in its entirety, which has a concentration of 100 ⁇ of Amplex® Red, is added to the test compound/saliva mixture.
  • each mixture is measured at 1 minute intervals using a spectrophotmoter (such as a Molecular Devices M5 reader) for excitation in the range of 530-560 nm and fluorescence emission detection at approximately 590nl, or for absorption at approximately 560nm. Reactions were allowed to run for 30 minutes during these assays.
  • a spectrophotmoter such as a Molecular Devices M5 reader
  • a test slide with 28 test wells are spotted with 12 to 18 sets of diagnostic markers.
  • These reagents may include diagnostic markers, or controls for the diagnosis.
  • the diagnostic device in this example, has four diagnostic markers spotted in duplicate, along with two positive and two negative control marker. Each marker is spotted with a competitor, such as anti-haptin, which binds the capture antigen particle to the marker.
  • the device may have four different hatpin/anti- haptin systems to serve as the generic capture system.
  • a unique haptin may be conjugated to a fully paramagnetic particle (PmMp).
  • the PmMp is also coated with a capture monoclonal antibody (MAb) specific for the marker.
  • MAb capture monoclonal antibody
  • a second particle (polystyrene microparticle with Europium) is conjugated with the second or detection MAb.
  • the hash marks on the side of the diagnostic device are indentations molded into the plastic, which may be used as cog grooves. Additionally the slide is designed to fit into a processor that may have more than one processing station, as shown in Fig. 36.
  • the user In performing a method of detecting a diagnostic marker using an embodiment of the diagnostic device, such as that in Fig. 34 and 36, the user adds a pre-determined amount of specimen (such as 120 ⁇ 1) to a specimen diluent (such as 40 to 120 ⁇ ), and adds the mixture to each test well. Following addition of the diluted specimen, the test slide is incubated at 37°C for about 5 to about 15 minutes with agitation. Following the incubation period, the test wells move over a magnetic source that brings the PmMp to the bottom of the well, so that there is maximal binding of the PmMp with the anti-haptin bound in the test well. Next, specimen and assay reagents are removed, wash buffer is added and aspirated off. Lastly, each well is scanned using a visualization device, such as a CCD camera, and then the fluroescent signal (or other signal) of each marker is determined.
  • a visualization device such as a CCD camera
  • a collection device for the collection of an bodily fluids is depicted in Fig. 35.
  • the collection device is capable of collecting at least 500 ⁇ 1 of bodily fluid, such as saliva.
  • the cap is first removed from the tube. Squeezing the bulb provides suction adequate to pull an amount of saliva into the bulb.
  • a filter may be included in the tube to remove unwanted materials (such as particulate and mucous).
  • the filter may be of any known filter material, such as rayon, that is appropriate for the purposes described herein.
  • the dispensing tube is in fluid communication with the bulb and is sized and shaped to allow for drops of a predetermined size to be formed when the bulb is squeezed.
  • the dispensing tube may allow drops of 30 ⁇ 1 to be formed when the bulb is squeezed.
  • the disclosure may have presented a method and/or process as a particular sequence of steps.
  • the method or process should not be limited to the particular sequence of steps described.
  • Other sequences of steps may be possible. Therefore, the particular order of the steps disclosed herein should not be construed as limitations of the present disclosure.
  • disclosure directed to a method and/or process should not be limited to the performance of their steps in the order written. Such sequences may be varied and still remain within the scope of the present disclosure.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Microbiology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

Devices, systems and methods for biomarker stabilization. In at least one embodiment of a stabilizing system, the system comprises a stabilizing agent useful to completely or substantially prevent degradation or inactivation of a diagnostic marker present within a mammalian body, the diagnostic marker indicative of a mammalian condition, and a detection agent capable of detecting the diagnostic marker. In at least one embodiment a method of the present disclosure, the method comprises the steps of mixing a mammalian body fluid comprising a diagnostic marker indicative of a condition with an effective amount of a stabilizing agent, and wherein the stabilizing agent completely or substantially prevents the cleavage, degradation, or inactivation of the diagnostic marker.

Description

DEVICES, SYSTEMS, AND METHODS FOR BIOMARKER
STABILIZATION
PRIORITY
This international patent application claims the priority benefit of U.S. Patent
Application Serial No. 12/906,157, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,158, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,159, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,161, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,162, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,164, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,168, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,170, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,196, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,199, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,201, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,204, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,202, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,208, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,223, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,224, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,231, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,239, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,245, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,253, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,260, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,211, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,221, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,227, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,235, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,225, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,236, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,255, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,263, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,268, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,273, filed October 18, 2010; U.S. Patent Application Serial
No. 12/906,166, filed October 18, 2010; U.S. Patent Application Serial No. 12/906,167, filed
October 18, 2010; U.S. Patent Application Serial No. 12/906,169, filed October 18, 2010;
U.S. Patent Application Serial No. 12/906,171, filed October 18, 2010; U.S. Patent
Application Serial No. 12/906,173, filed October 18, 2010; U.S. Provisional Patent Application Serial No. 61/390,002, filed October 5, 2010; U.S. Provisional Patent Application Serial No. 61/385,347, filed September 22, 2010; U.S. Provisional Patent Application Serial No. 61/383,401, filed September 16, 2010; U.S. Provisional Patent Application Serial No. 61/379,598, filed September 2, 2010; U.S. Provisional Patent Application Serial No. 61/378,960, filed September 1, 2010; U.S. Provisional Patent Application Serial No. 61/373,619, filed August 13, 2010; U.S. Provisional Patent Application Serial No. 61/364,964, filed July 16, 2010; U.S. Provisional Patent Application Serial No. 61/364,969, filed July 16, 2010; U.S. Provisional Patent Application Serial No. 61/364,975, filed July 16, 2010; U.S. Provisional Patent Application Serial No. 61/364,978, filed July 16, 2010; U.S. Provisional Patent Application Serial No. 61/364,982, filed July 16, 2010; U.S. Provisional Patent Application Serial No. 61/365,179, filed July 16, 2010; and U.S. Provisional Patent Application Serial No. 61/322,768, filed April 9, 2010. The contents of each of these applications are hereby incorporated by reference in their entirety into this disclosure.
BACKGROUND
Biomarkers are molecular indicators of a specific biological property, a biochemical feature or facet that can be used to measure the progress of a disease or the effects of a treatment. For example, serum low-density lipoprotein (LDL) is a biomarker of cholesterol and blood pressure, while the P53 gene is a biomarker for cancer. For chronic diseases and conditions, such as diabetes and allergies, accurate diagnosis is particularly important, especially where the side effects of a treatment are severe.
Diagnostic tests using biomarkers as molecular indicators not only detect the presence or absence of the biomarker, but often must measure the exact concentration of a biomarker to determine whether an abnormal condition exists. Because of the requirement for accuracy, the process of sample collection, preparation, and analysis are often complicated and time consuming. Currently, blood-based assays for biomarker presence or activity are considered to be the "gold standard" for biomarker-type assays.
Despite the desire for accurate results, rapid, point of care analysis of biological samples using biomarkers is becoming increasingly important in the present medical environment due to the need for quick results.
BRIEF SUMMARY
The disclosure of the present application provides computer-implemented methods and systems of improving diagnostic marker availability.
In at least one embodiment of a stabilizing system of the present disclosure, the system comprises a stabilizing agent useful to completely or substantially prevent degradation or inactivation of a diagnostic marker present within a mammalian body, the diagnostic marker indicative of a mammalian condition, and a detection agent capable of detecting the diagnostic marker. Optionally, an embodiment of the system may further comprise a detection platform comprising a plurality of detection sites each capable of receiving the diagnostic marker, the stabilizing agent, and the detection agent, a computer database capable of receiving a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of the diagnostic marker to the detection agent, a processor operably coupled to the computer database and the detection platform, the processor having and executing a software program operational to (1) determine a binding characteristic between the detection agent and the stabilized diagnostic agent in each of the plurality of detection sites, (2) compare the binding characteristic among each of the plurality of detection sites, wherein the comparison of binding characteristics is capable of determining the stabilizing agent with the greatest effect on the binding characteristic between the detection agent and the diagnostic agent, (3) generate a binding record using the compared binding characteristics, and (4)deliver the binding record to a recipient. In at least one embodiment of the system, the software program is further operational to compare the binding characteristic with at least one of a plurality of stored binding characteristics in the computer database.
In at least one embodiment of the system of the present disclosure, the system further comprises a detection platform comprising a plurality of detection sites each capable of receiving the diagnostic marker, the stabilizing agent, and the detection agent, a detection device capable of determining a binding characteristic between the detection agent and the diagnostic marker in the detection sites, a computer database capable of receiving a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of the diagnostic marker to the detection agent, a processor operably coupled to the computer database and the detection device, the processor having and executing a software program operational to (1) determine the binding characteristic of the detection agent to the diagnostic marker in each of the plurality of detection sites, (2) compare the binding characteristic among each of the plurality of detection sites to order the level of effect of the stabilizing agents assayed on the binding characteristic between the detection agent and the diagnostic agent, (3)generate a binding report using the compared binding characteristics, and (4) deliver the binding report to a recipient.
In at least one embodiment of the system of the present disclosusre, the stabilizing system further comprises a detection platform comprising a plurality of detection sites each capable of receiving at the stabilizing agent, the detection agent, and a body fluid from a patient, the body fluid comprising the diagnostic marker, a computer database capable of storing a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of the diagnostic marker to the detection agent, and a processor operably coupled to the computer database and the detection platform, the processor having and executing a software program operational to determine a binding characteristic of the detection agent and a stabilized diagnostic marker in each of the plurality of detection sites compare the binding characteristic among each of the plurality of detection sites, wherein the comparison of binding characteristics is capable of determining the stabilizing agent with the greatest effect on the binding characteristic between the detection agent and the diagnostic marker, generate a binding record using the compared binding characteristics, and deliver the binding record to a recipient. Optionally, the binding record may be used by to assist in determining a therapeutic course for the patient.
In at least one embodiment of the system of the present disclosure, the system further comprises a fluid collector to collect a body fluid, and a housing to test and retain the body fluid, the housing comprising a collection chamber, having an open end, to receive the fluid collector and contain the stabilization agent, at least one membrane test strip, in fluid communication with the collection chamber, to indicate the presence or absence of the diagnostic marker, and an immunoassay-based fingerprint acquisition pad in fluid communication with the collection chamber, the acquisition pad having at least one control zone including a control reagent to identify a donor of the body fluid, the reagent including a member of a predetermined ligand/receptor binding pair, and a plurality of reaction zones, each of which includes the detection agent to determine the presence of the diagnostic marker in the fluid sample, the detection agent including a member of a predetermined ligand/receptor binding pair.
In at least one embodiment of the system of the present disclosure, the system further comprises a detection platform comprising a plurality of detection sites each capable of receiving the diagnostic marker, the stabilizing agent, and the detection agent, a computer database capable of receiving a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of the diagnostic marker to the detection agent, a processor operably coupled to the computer database and the detection platform, the processor having and executing a software program. The software program operational to determine a binding characteristic of the detection agent and a stabilized detection agent in each of the plurality of detection sites, compare the binding characteristic among each of the plurality of detection sites, wherein the comparison of binding characteristics is capable of determining the stabilizing agent with the greatest effect on the binding characteristic between the detection agent and the diagnostic agent, generate a binding record using the compared binding characteristics, and deliver the binding record to a recipient.
In at least one embodiment of the system of the present disclosure, the system further comprises a detection platform comprising a plurality of detection wells housed in a solid matrix, the detection wells containing a plurality of the diagnostic markers and at least one competitor molecule, wherein the plurality of the diagnostic markers and at least one control molecule are each bound to a paramagnetic particle, a detection system capable of receiving the detection platform, exerting a magnetic field upon a surface of the detection platform, and determining at least one characteristic of the diagnostic marker. An embodiment of the solid matrix may be comprised of material selected for the group consisting of quartz, glass, metal, silica-based materials, resins, polymers, or combinations thereof. Further, the magnetic field may be able to force the paramagnetic particle towards the surface of the detection platform. Optionally, the an embodiemt of the system may further comprise a second paramagnetic particle, such as Europium, linked to a detection antibody, and optionally coupled to a second detection agent.
In at least one embodiment of the system of the present disclosure, the system further comprises a diagnostic device comprising a plurality of test wells, wherein each test well is capable of containing the detection agent, and a detection device in communication with the diagnostic device, wherein the detection device is capable of detecting an interaction between the at least one detection agent and the diagnostic marker in at least one of the plurality of test wells, wherein the stabilizing agent is contained within at least one of the plurality of test wells. Optionally, the diagnostic device may be comprised of a material selected for the group consisting of quartz, glass, metal, silica-based materials, resins, polymers, or combinations thereof. Further, an embodiment of the detection device may be capable of detecting a colorimetric or fluorometric signal from at least one of the plurality of test wells.
In at least one embodiment of the system of the present disclosure, the system further comprises a microarray product comprising at least 100 diagnostic markers/cm2, a microarray identifier, and the stabilizing agent, the system also having a control microarray product comprising a first specific binding pair member that binds to a first detectable label, and a processor for providing information regarding the identification and concentration of markers on the microarray based on the identity of the array provided by the microarray identifier. The microarray product may be comprised of a compound selected from the group consisting of gel, nitrocellulose, nylon, quartz, glass, metal, silica based materials, silica, resins, polymers, or combinations thereof. Further, the diagnostic marker may comprises a DNA fragment having a length of about 10 base pairs to about 50 base pairs of DNA or R A, or a length of at least about about 50 amino acids.
In at least one embodiment of the system of the present disclosure, the system further comprises a latex particle having a carboxylate group, the latex particle functionally coupled to the stabilizing agent. Optionally, the latex particle may further comprising at least one of human serum albumin (HSA) and bovine serum albumin (BSA), and/or a linker group coupled to latex particle, where the linker group may comprise a maleimide compound. In at least one example, the maleimide compound is selected from the group consisting of Succinimidyl 4-[N-Maleimidomethyl] Cyclohexane-1 -Carboxylate (SMCC) and N- Hydroxysuccinimide-activated hexa(ethylene glycol) undecane Thiol (NHS). Further the linker group may be coupled to the HSA or BSA. In at least one embodiment, the system may further comprise antibody functionally linked to the latex particle. The latex particle in at least one embodiment may have a diameter of about 0.02μιη to about 7.0μιη.
In at least one embodiment of the stabilizing agent of the present disclosure, the stabilizing agent is selected from the group consisting of a protease inhibitor, a DNase inhibitor, and a RNase inhibitor. Further, the stabilizing agent may be 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. An embodiment of the stabilizing agent may be selected from the group consisting of Fixanal® Buffer 6.0 (Sigma-Aldrich Co.), acetic acid, aluminum hydroxide bentonite, aluminum sulfate hydrate, aluminum potassium sulfate dodecahydrate, benzoic acid, caffeine, and 3- tert-butyl-hydroxyanisole, or a combination thereof. Optionally, the stabilizing agent may comprise a plurality of stabilizing agents, each present in approximately the same concentration. Moreover the stabilizing agent may be capable of inhibiting degradation of the diagnostic marker to an inhibitory degree, wherein the inhibitory degree is selected from the 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%, and at least about 99%.
In at least one embodiment of the stabilizing agent, the stabilizing agent has a concentration selected from the group consisting of about 200 parts per million (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. Further, the stabilizing agent may be able to inhibit the degradation or inactivation of the diagnostic marker for an inhibitory period selected from the group consisting of at least one minute, at least about five minutes, at least about ten minutes, at least about fifteen minutes, at least about thirty minutes, at least about one hour, at least about two hours, at least about four hours, and at least about eight hours.
In at least one embodiment of the system or methodof the present disclosure, the diagnostic marker is selected from the group consisting of a protein, a glycoprotein, a nucleic acid, an enzyme, an enzyme inhibitor, and a metabolite. Further, in at least one embodiment of the system or method of the present discosure, the diagnostic marker is selected from the group consisting a-hydroxybutyrate, 1-linoleoyl-GPC, palmitate, Glycine, and 3-methyl-2- oxybutyrate, glycosylated hemoglobin (HbAlc), IgE, and IgG, Aldose reductase, Angiogenin, Annexin Al, B-cell activating factor (BAFF), B-cell lymphoma 2 (BCL2)-like 2, Beta Human chorionic gonadotropin, Cal5-3, Calcyclin, Calvasculin, Cancer Antigen CA 19-9, Cancer Antigen CA 15-3, Cathepsin D, Caveolin-1, Chromogranin A, Alpha-crystallin B chain (CRYAB), Endostatin, Eotaxin-2, Epithelial cell adhesion molecule (EpCAM), Ezrin, fatty acid binding protein 4 (FABP4), Galectin-3, γ-glutamylcysteine ligase regulatory chain (GCLR), Gelsolin, Glucose 6-phosphate (G6P), Glycoprotein 130 (gpl30), Glutathione S-transferase Mu 1 (GSTM1), Hepsin, High-mobility group protein Bl (HMGB-1), Insulinlike growth factor binding protein 1 (IGFBP-1), Insulin- like growth factor binding protein 4 (IGFBP-4), Insulin-like growth factor binding protein 5 (IGFBP-5), Insulin-like growth factor binding protein 6 (IGFBP-6), LGL, latency associated peptide (LAP), macrophage stimulating protein (MSP), MHC class I polypeptide-related sequence A (MICA), Nucleoside diphosphate kinase B (NME2), Neuron-specific Enolase (NSE), Osteopontin, Osteoprotegerin, Pepsinogen, Peroxiredoxin, Phosphoserine aminotransferase (PSAT1), Prostate Specific Antigen, Receptor tyrosine-protein kinase erbB-3 (ErbB3), Serpin B3, Vascular smooth muscle cell growth factor R2 (VSGF R2/KDR), Vascular endothelial growth factor R3 (VEGF R3/Flt-4), Thyroglobulin, Tyrosine kinase with immunoglobulin- like and EGF-like domains 2 (TIE-2), Tissue plasminogen activator (tPA), Transforming growth factor beta (TGF-βΙ), Tumor necrosis factor receptor 1 (TNF-R1), urokinase-type Plasminogen Activator (uPA), urokinase-type Plasminogen Activator Receptor (uPAR), Brcal, Brcall, kallikreins, e-cadherin, Hox peptide, Engrailed-2, creatine kinase, troponin I and T, LD, Myoglobin, Alanine aminotransferase (ALT), Aspartate transaminase(AST), ALT/AST ratio, Heart-type fatty acid binding protein (H-FABP), Glycogen phosphorylase B, a bacterial protein, a bacterial outer membrane protein, a bacterial secreted protein, a bacteria-specific DNA sequence, a bacteria-specific RNA sequence, a bacterial metabolite, a host-reaction product to a bacteria, a viral protein, a viral outer membrane protein, a viral secreted protein, a viral-specific DNA sequence, a viral-specific RNA sequence, a viral metabolite, and a host-reaction product to a virus, a fungal protein, a fungal outer membrane protein, a fungal secreted protein, a fungal-specific DNA sequence, a fungal-specific RNA sequence, a fungal metabolite, and a host-reaction product to a fungus, a parasite protein, a parasite outer membrane protein, a parasite secreted protein, a parasite-specific DNA sequence, a parasite-specific RNA sequence, a parasite metabolite, a host-reaction product to a parasite, lutenizing hormone, progesterone, human chorionic gonadotropin (hCG), early pregnancy factor (EPF), calcium, vitamin D, phosphorus, and magnesium.
In at least one embodiment of the present disclosure, the diagnostic marker may be indicative of a drug selected from the group consisting alcohol, cocaine, marijuana, opiates, amphetamine, methamphetamine, amphetamines, phencyclidine, benzodiazepines, barbiturates, methadone, tricyclic antidepressants, heroin, steroids, niacin, xanan, vicodin, oxycontin, adderall, morphine, and nicotine.
In at least one embodiment of the condition of the present disclosure, the condition is selected from a group consisting of insulin resistance, glucose intolerance, cancer, cardiovascular disease, a bacterial infection, a viral infection, a fungal infection, a parasite infection, a food allergy, a non-food allergy, drug use, fertility, pregnancy, a level of circulating calcium. Further, in at least one embodiment of the system, the degradation or inactivation of the diagnostic marker for a condition may occur by a destructive agent, such as galactose oxidase.
In at least one embodiment of the body fluid of the present disclosure, the body fluid is selected from the group consisting of saliva, a mucous secretion, tears, sweat, semen, urine, a vaginal secretion, exhalate, blood, serum, and an anal secretion.
In at least one method of the present disclosure, the method comprises the step of mixing a mammalian body fluid comprising a diagnostic marker indicative of a condition with an effective amount of a stabilizing agent, wherein the stabilizing agent completely or substantially prevents the cleavage, degradation, or inactivation of the diagnostic marker.
Optionally, the step of mixing a body fluid with the stabilizing agent may comprise introducing the stabilizing agent into a mouth/oral cavity of a patient so that the stabilizing agent mixes with saliva. In at least one embodiment of the method of the present disclosure, the method comprises the steps of introducing a predetermined diagnostic marker for cancer into a plurality of detection sites of a detection platform, introducing a stabilization agent into each of the plurality of detection sites containing the predetermined diagnostic marker for cancer, wherein the stabilization agent in each of the plurality of detection sites is capable of completely or substantially preventing the degradation or inactivation of the diagnostic marker, introducing a detection agent into each of the plurality of detection sites having a stabilized diagnostic agent, determining a binding characteristic of the detection agent and the stabilized diagnostic agent in each of the plurality of detection sites with a processor, and computationally comparing the binding characteristic among each of the plurality of detection sites with the processor, wherein the comparison of binding characteristics is capable of determining the stabilizing agent with the greater effect on the binding characteristic between the detection agent and the diagnostic agent. The detection platform in at least one embodiment is selected from the group consisting of a microtitre plate, a microarray, and a multi-well plate.
At least one embodiment of the method of the present disclosure may further comprise the step of computationally comparing the binding characteristics to at least one stored binding characteristic contained in the computer database in communication with the processor.
In at least one embodiment of the method of the present disclosure, the method comprises the steps of contacting a body fluid of a diseased subject having a cancer with a stabilizing agent capable of completely or substantially preventing the degradation or inactivation of a diagnostic marker indicative of the cancer, the body fluid comprising the diagnostic agent, analyzing the stabilized body fluid to determine at last one property of the diagnostic marker to create a cancer profile; comparing the cancer profile with a control profile of a healthy subject not having the cancer, wherein the control profile is created in a like manner to the cancer profile, and generating a cancer identifier panel from the compared cancer profile, wherein the cancer identifier panel contains at least one cancer diagnostic marker indicative of the cancer. In at least one exemplary embodiment, the stabilizing agent may prevent the degradation or inactivation of the diagnostic marker for at least a twenty four hours period following interaction with the diagnostic agent.
In at least one embodiment of the method of the present disclosure, the method comprises the steps of introducing a diagnostic marker into each of a plurality of detection sites of a detection platform, the detection platform comprising the plurality of detection sites each capable of receiving a diagnostic marker, a stabilization agent, and a detection agent, a detection device capable of determining a binding characteristic between the detection agent and the diagnostic marker in the detection sites, a computer database capable of receiving a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of a diagnostic marker to a detection agent, and a processor operably coupled to the computer database and the detection device. The processor, in at least one embodiment, having and executing a software program operational to (l)determine the binding characteristic of the detection agent to the diagnostic marker in each of the plurality of detection sites, (2) compare the binding characteristic among each of the plurality of detection sites to order the level of effect of the stabilizing agents assayed on the binding characteristic between the detection agent and the diagnostic agent, (3) generate a binding record using the compared binding characteristics, and (4) deliver the binding record to a recipient. The at least one embodiment of the method further comprising the steps of introducing the stabilization agent to each of the plurality of detection sites containing the diagnostic marker, combining the detection agent with the stabilization agent and diagnostic marker in the detection sites, comparing the binding characteristic with the processor among each of the plurality of detection sites to order the level of effect of the stabilizing agents assayed on the binding characteristic between the detection agent and the diagnostic agent, generating a binding report using the compared binding characteristics, and delivering the binding report to a recipient.
In at least one embodiment of the method of the present disclosure, the method comprises the steps of treating a subject having a disease state with an effective amount of therapeutic compound, harvesting at least one cell from the subject following a treatment with the therapeutic compound, bringing the at least one cell , such as a tumor cell, into contact with a stabilizing compound capable of completely or substantially preventing the degradation or inactivation of a diagnostic marker for the disease state, analyzing the at least one stabilized cell for the at least one diagnostic marker, creating a marker profile from at least one result determined from analyzing the at least one stabilized cell, and comparing the marker profile with at least one previous marker profile to determine efficacy of the therapeutic compound on the disease state. Optionally, the step of harvesting the at least one cell comprises the step of collecting a body fluid from the subject, wherein the body fluid comprises the at least one cell.
In at least one embodiment of the method of the present disclosure, the method comprises the steps of harvesting a body fluid from a patient having a disease state, introducing the body fluid to a detection platform comprising a plurality of detection sites each capable of receiving a stabilization agent, a detection agent, and a body fluid from a patient comprising a diagnostic marker, a computer database capable of storing a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of a diagnostic marker to a detection agent, a processor operably coupled to the computer database and the detection platform, the processor having and executing a software program. The software program operational to determine a binding characteristic of the detection agent and a stabilized diagnostic marker in each of the plurality of detection sites, compare the binding characteristic among each of the plurality of detection sites, wherein the comparison of binding characteristics is capable of determining the stabilizing agent with the greatest effect on the binding characteristic between the detection agent and the diagnostic marker, generate a binding record using the compared binding characteristics, and deliver the binding record to a recipient. Further, in at least one embodiment, the method further comprises the steps of contacting the body fluid with a stabilizing agent capable of completely or substantially preventing the degradation or inactivation of a diagnostic marker for the disease state, analyzing the at least one stabilized body fluid for the at least one diagnostic marker to create a marker profile, comparing the marker profile with a marker profile library stored in the computer database to determine a therapeutic potential of the therapeutic compound on the disease state, generating a binding record using the compared binding characteristics, and delivering the binding record to a recipient.
In at least one embodiment of the method of the present disclosure, the method comprises the steps of combining a body fluid comprising a diagnostic marker with a stabilization agent for an incubation period, the stabilization agent capable of completely or substantially preventing the degradation or inactivation of the diagnostic marker, introducing the stabilized body fluid into a detection platform, combining a detection agent with the stabilized body fluid, determining an interaction characteristic of the detection agent and the diagnostic marker of the stabilized body fluid using a detection device in communication with a processor, comparing the interaction characteristic with a plurality of indexed interaction records contained on a database in communication with the processor to determine an identity and concentration of the detection marker, generating a report of the identity and concentration of the detection marker in the body fluid, and delivering the report to a recipient. Optionally, the the detection platform may be selected from the group consisting of a microarray, a multi-well plate, a high-performance liquid chromato graph, and a mass spectrometer. In at least one embodiment of the present disclosure, a microfluidic device for analyzing the presence or absence of the diagnostic marker to be detected in a body fluid is disclosed. An embodiment of the microfluidic device comprising a container, the container housing a sample reservoir having a sample input port in fluid connection with at least one detector array, wherein the detector array comprises at least one detector comprising a reaction chamber comprising an immobilized capture molecule, the stabilizing agent contained within the sample reservoir, the stabilizing agent useful to completely or substantially prevent degradation or inactivation of the diagnostic marker, and a reagent contained within the detector array, the reagent capable of undergoing a colorimetric reaction or displaying an optically detectable signal and capable of reacting with the diagnostic marker. An embodiment of the microfluidic device may further comprise at least one of a filter capable of separating at least one unwanted component from a body fluid, a results display, wherein a result from the detector array is visually detectable, a plurality of displays capable to visualizing a plurality of detected diagnostic markers, and a control display, wherein the detection of at least one control reagent by the at least one detector is visualized.
In at least one embodiment of a system of the present disclosure, the system comprises a sample container having an opening and least one rigid chamber for containing a body fluid, the body fluid comprising at least one diagnostic marker, a stabilizing agent contained within the sample container, the stabilizing agent capable of completely or substantially preventing the degradation or inactivation of the diagnostic marker, and a securing device for closing the opening of the sample container, wherein the stabilizing agent is coupled to a solid support. Optionally, the the stabilizing agent may also be adsorbed to the solid support.
DESCRIPTION OF THE DRAWINGS
The features and advantages of the present disclosure, and the manner of attaining them, will be more apparent and better understood by reference to the following descriptions taken in conjunction with the accompanying figures, wherein:
Figs. 1A and B show a diagram of a detection system, according to at least one embodiment of the present disclosure;
Figs. 2-4 shows a diagram of a method of producing a bead system, according to at least one embodiment of the present disclosure;
Fig. 5 shows a flowchart of a method for creating a bead system, according to at least one embodiment of the present disclosure; Figs. 6A and B show a diagram of a test strip, according to at least one embodiment of the present disclosure;
Fig. 6C show a diagram of a microarray, according to at least one embodiment of the present disclosure;
Fig. 6D shows a diagram of a microfluidic system, according to at least one embodiment of the present disclosure;
Fig. 6E shows a diagram of a sample collection device, according to at least one embodiment of the present disclosure;
Fig. 6F shows a diagram of a sample collection device, according to at least one embodiment of the present disclosure;
Fig. 6G shows a diagram of a sample container, according to at least one embodiment of the present disclosure;
Fig. 6H shows a diagram of a sample container, according to at least one embodiment of the present disclosure;
Fig. 7 shows a flowchart of a method of stabilizing diagnostic markers, according to at least one embodiment of the present disclosure;
Fig. 8 shows a flowchart of a method analyzing a diagnostic markers, according to at least one embodiment of the present disclosure;
Fig. 9 shows a flowchart of a method of improving biomarker availability, according to at least one embodiment of the present disclosure;
Fig. 10 shows a flowchart of a method of identifying diagnostic markers of a disease state, according to at least one embodiment of the present disclosure;
Fig. 11 shows a flowchart of a method for screening biomarkers, according to at least one embodiment of the present disclosure;
Fig. 12 shows a flowchart of a method of stabilizing diagnostic markers, according to at least one embodiment of the present disclosure;
Fig. 13 shows a an exemplary system framework, according to at least one embodiment of the present disclosure;
Fig. 14 shows a graphical depiction of a-amylase inhibition according to at least one embodiment of the present disclosure;
Fig. 15 shows a graphical depiction of a-amylase inhibition according to at least one embodiment of the present disclosure;
Fig. 16 shows a graphical depiction of lysozyme inhibition according to at least one embodiment of the present disclosure; Fig. 17 shows a graphical depiction of a galactose oxidase screen according to at least one embodiment of the present disclosure;
Fig. 18A shows a three dimensional plot of glycated Hemoglobin Ale (HbAlc) analyzed by high pressure liquid chromatography (HPLC) according to at least one embodiment of the present disclosure;
Fig. 18B shows a ultraviolet (UV) image of HbAlc analyzed by HPLC according to at least one embodiment of the present disclosure;
Fig. 18C shows a chromatogram of HbAlc analyzed by HPLC according to at least one embodiment of the present disclosure;
Fig. 19A shows a three dimensional plot of HbAlc analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure;
Fig. 19B shows a UV image of HbAlc analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure;
Fig. 19C shows a chromatogram of HbAlc analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure;
Fig. 20A shows a three dimensional plot of HbAlc analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure;
Fig. 20B shows a UV image of HbAlc analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure;
Fig. 20C shows a chromatogram of HbAlc analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure;
Fig. 21 shows a comparison of chromatograms from HPLC analyzed HbAlc (Fig.
21 A), HbAlc + Saliva (Fig. 2 IB), and HbAlc +Saliva + Stabilizing mixture (Fig. 21C) according to at least one embodiment of the present disclosure;
Fig. 22 shows a comparison of UV images from HPLC analyzed HbAlc (Fig. 22A), HbAlc + Saliva (Fig. 22B), and HbAlc + Saliva + Stabilizing mixture (Fig. 22C) according to at least one embodiment of the present disclosure;
Fig 23 shows a comparison of three dimensional plots from HPLC analyzed HbAlc (Fig. 23A), HbAlc + Saliva (Fig. 23B), and HbAlc + Saliva + Stabilizing mixture (Fig. 23C) according to at least one embodiment of the present disclosure; Fig. 24A shows a three dimensional plot of Cancer Antigen 19-9 (CA 19-9) analyzed by high pressure liquid chromatography (HPLC) according to at least one embodiment of the present disclosure;
Fig. 24B shows a ultraviolet (UV) image of CA 19-9 analyzed by HPLC according to at least one embodiment of the present disclosure;
Fig. 25 A shows a three dimensional plot of CA 19-9 analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure;
Fig. 25B shows a UV image of CA 19-9 analyzed by HPLC following exposure to saliva according to at least one embodiment of the present disclosure;
Fig. 26A shows a three dimensional plot of CA 19-9 analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure;
Fig. 26B shows a UV image of CA 19-9 analyzed by HPLC following exposure to saliva and a stabilizing mixture according to at least one embodiment of the present disclosure;
Fig. 27 shows a comparison of three dimensional plots from HPLC analyzed CA 19-9 (Fig. 27A), CA 19-9 + Saliva (Fig. 27B), and CA 19-9 + Saliva + Stabilizing mixture (Fig. 27C) according to at least one embodiment of the present disclosure;
Fig. 28 shows a comparison of UV images from HPLC analyzed CA 19-9 (Fig. 28 A), CA 19-9 + Saliva (Fig. 28B), and CA 19-9 + Saliva + Stabilizing mixture (Fig. 28C) according to at least one embodiment of the present disclosure;
Fig. 29 shows a graphical depiction of Peroxidase activity inhibition by an inhibitor cocktail according to at least one embodiment of the present disclosure;
Fig. 30 shows a graphical depiction of Peroxidase activity inhibition by caffeine according to at least one embodiment of the present disclosure;
Fig. 31 shows a graphical depiction of Peroxidase activity inhibition by 3-tert-butyl- hydroxyanisole according to at least one embodiment of the present disclosure;
Fig. 32 shows a graphical depiction of Peroxidase activity inhibition by benzoic acid according to at least one embodiment of the present disclosure;
Fig. 33 shows a graphical depiction of Peroxidase activity inhibition by aluminum potassium sulfate dodecahydrate according to at least one embodiment of the present disclosure;
Fig. 34 shows a schematic of a diagnostic system, according to at least one embodiment of the present disclosure; Fig. 35 shows a graphical representation of an assay component, according to at least one embodiment of the present disclosure; and
Fig. 36 shows a graphical depiction of a fluid collection device, according to at least one embodiment of the present disclosure.
DETAILED DESCRIPTION
For the purposes of promoting an understanding of the principles of the present disclosure, reference will now be made to the embodiments illustrated in the drawings, and specific language will be used to describe the same. It will nevertheless be understood that no limitation of the scope of this disclosure is thereby intended.
The disclosure of the present application provides various compositions, systems, and methods for biomarker stabilization and analysis. Specifically, formulations are disclosed herein, which function to preserve the level of diagnostic markers, such as biomarkers, in body fluids, from enzymatic alteration, or degradation. Additionally, methods for the collection and analysis of body fluids are disclosed. The methods and formulations disclosed herein can be used to improve the sampling and testing of a body fluid by conditioning the body fluids for the stabilization of specific biomolecules and/or drug metabolites. As used herein, the term "body fluids" includes fluids produced by the body, such as saliva, or fractions thereof, mucous secretions, tears, sweat, bile, semen, urine, vaginal secretions, exhalations, anal secretions, blood, plasma, serum and mixtures of thereof. Body fluids may also comprise cancer cells, peripheral blood mononuclear cells, lymphocytes, lymph fluid, and other tissue secretions or fluid.
Saliva
Saliva is clear, viscous fluid with a slightly alkaline pH and a pi range from 11.5-3.0. It is hypotonic, composed of about 99.5% water, and also contains ions (e.g., K+, Na+, Ca ,
2_|_ _|_ _ _ _ _ _
Mg , H , CI", HCO3 ", I ", F", HPO4 "), and small organic molecules (e.g., ureas, hormones, lipids, DNA, and R A). There are multiple contributors to the composition of saliva. Saliva has a complex "proteome"- 106 D glycoproteins to 1000D peptides. It contains secretory products of salivary glands, products of B cells, PMNs, epithelial cells, and bacteria. Major (e.g., parotid, submandibular, and sublingual) and minor (e.g., palatine and retromolar) glands contribute to the composition of saliva, along with extraneous contributors such as gingival crevicular fluid, serum proteins, white blood cells and their byproducts, oral epithelial cells, oral bacteria, food debris and dissolved food components.
Saliva from different glands may differ in composition. For example, saliva from the parotid gland is dominated by serous secretory cells, whereas saliva from the submandibular and sublingual glands and minor glands are mixed serous or mostly mucous. There can also be qualitative and quantitative differences in saliva output that affect its composition. Glandular contribution to saliva is affected by level of gland activity. The amount of saliva secreted per minute or the salivary flow rate influences the concentration of the constituents as well as the proportion of the constituents from each of the three pairs of major salivary glands and the minor salivary glands.
I. Stabilizing Agents
According to at least one embodiment of the present disclosure, conditioning of body fluids for the preservation of diagnostic markers may be accomplished through the use of stabilizing agents, which may be incorporated into one or more carrier vehicles, such as rinses, gums, beverages, and confectionaries. For example, specific rinses and/or pre-rinses specially formulated according to the specific molecule or molecules (the diagnostic marker) to be detected in the body fluid, may contain a stabilization agent. The use of rinses and pre- rinses of the present disclosure to condition the body fluid may enhance the sensitivity for detection of specific diagnostic markers for clinical diagnosis. Additionally, this process may improve the signal-to-noise ratio for a better diagnostic yield. Further, each rinse/pre-rinse can be specifically formulated to reduce or prevent false positive and false negative results.
In at least one embodiment of a stabilizing agent of the present disclosure, the stabilizing agent may act to prevent/decrease the degradation, or reduction of activity, of diagnostic markers. In at least one embodiment, the stabilizing agent may be comprised of one or more Generally Regarded as Safe (GRAS) compound, as determined by the Food and Drug Administration of the United States of America, such as those listed in Table I. An exemplary embodiment of a stabilization agent may act to stabilize a diagnostic marker, and/or to bind to a destructive component to prevent the destructive component from degrading or decreasing the activity of the diagnostic marker.
In additional exemplary embodiments of the stabilization agent, the stabilizing agent may comprise at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, or at least 25 compounds which acts to stabilize the diagnostic marker, and/or to bind to a destructive component to prevent the destructive component from degrading or decreasing the activity of the diagnostic marker. Further, in various embodiments the stabilizing agent of the present disclosure may be present in at a level of between about 200ppm to about 2,000 ppm, about 400ppm to about 1600 ppm, about 600ppm to about 1400 ppm, about 800ppm to about 1200 ppm. Moreover, in at least one embodiment, the stabilizing agent may be present at a level of approximately 500ppm. Agar Sigma-Aldrich
Alginic acid sodium salt Sigma-Aldrich
L-Ascorbic acid Sigma-Aldrich
(+)-Sodium ascorbate Sigma-Aldrich
Buffer solution Fluka
Sodium citrate Sigma-Aldrich
Tragacanth Sigma-Aldrich
Gum arabic, from acacia tree Sigma-Aldrich
Sodium tripolyphosphate Sigma-Aldrich
Potassium citrate monobasic Sigma-Aldrich
Sodium pyrophosphate tetrabasic Sigma-Aldrich
Calcium citrate Sigma-Aldrich
Citric acid Sigma-Aldrich
Potassium sodium tartrate tetrahydrate Sigma-Aldrich
Sodium benzoate Sigma-Aldrich
Benzoic acid Sigma-Aldrich
D-aspartic acid Sigma-Aldrich
Sodium phosphate dibasic Sigma-Aldrich
Malic acid Sigma-Aldrich
Sodium propionate Sigma-Aldrich
Sodium L- ascorbate Sigma-Aldrich
DL-Phenylalanine Sigma-Aldrich
Calcium L-ascorbate dihydrate Sigma-Aldrich
Choline chloride Sigma-Aldrich
L-Ascorbic acid Sigma-Aldrich
Sodium diacetate SAFC
PH buffer 4.01 Orion
PH Buffer 7.00 Orion
PH buffer 10.0 Orion
Fixanal Buffer 6.0 Sigma-Aldrich
Ammonium Chloride Sigma-Aldrich
Ammonium Citrate tribasic, anhydrous Sigma-Aldrich
Caffeine Sigma-Aldrich
Aluminum sulfate hydrate Sigma-Aldrich
Beeswax refined Sigma-Aldrich Aluminum Hydroxide Sigma-Aldrich Bentonite Sigma-Aldrich Ammonium bicarbonate Sigma-Aldrich 3-tert-butyl-hydroxyanisole Sigma-Aldrich 40 Benzoic acid Sigma-Aldrich
41 trans-aconitic acid Sigma-Aldrich
Aluminum potassium sulfate
42 dodeca hydrate 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.
Molecular Inhibitory
Material Details
weight power
2.2 times
Lima beans 8-10 kDa There are six different lima bean inhibitors.
weight
Ovomucoids are the glycoprotein protease inhibitors
1.2 times
Ovomucoid 8-10 kDa found in raw avian egg white. There are other protease weight
inhibitors in ovomucoids as well.
Soybeans contain several inhibitors; the one in the chart is
20.7-22.3 1.2 times
Soybeans considered the primary one. All of them bind
kDa weight
chymotrypsin to a lesser degree.
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 state or heath/lifestyle characteristic (which may collectively be referred to as a "condition"). As used herein, "indicative of a disease state" refers to the presence, progression, prevention, remission, amelioration, or prognosis of a disease. This also includes, but is not limited to, the effects of a therapeutic on a disease. The disease state analyzed through use of the diagnostic marker may in an exemplary embodiment be directed towards any cancer, metabolic disease (such as diabetes I or II), cardiovascular disease, or the predisposition of any of the same. Additionally, a disease state may also include the infection of the patient by a bacterial, virus, yeast, fungus, or parasite.
A. Disease States
Bacterial infections, as referenced herein, may include a mammalian infection by any bacterial species. Embodiments of the bacterial species, as used herein, may include, but are not limited to, Bacillus anthracis, Bacillus cereus, Staphylococcus aureus, Listeria monocytogenes, Streptococcus pneumoniae, Streptococcus pyogenes, Clostridium botulinum, Clostridium difficile, Clostridium perfringens, Clostridium tetani, Borrelia burgdorferi, Treponema pallidum, Chlamydia trachomatis, Chlamydophila psittaci, Corynebacterium 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 meningitides, Campylobacter jejuni, Helicobacter pylori, Legionella pneumophila, Acinetobacter baumannii, Moraxella catarrhalis, Pseudomonas aeruginosa, Aeromonas sp., Vibrio cholerae, Vibrio parahaemolyticus, Thiotrichales sp., Haemophilus influenzae, Klebsiella pneumoniae, Proteus mirabilis, Yersinia pestis, Yersinia 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 DNA viruses, including the genus Parvoviridae, double-stranded DNA viruses, including the genus Papillomaviridae, Polyomaviridae, Poxviridae, Herpesviridae (ex. Herpes Simplex Virus 1, 2, 6, Varicella-zoster virus, Epstein-Barr virus EBV, Human cytomegalovirus, and human herpesvirus 7), single-stranded R A viruses, Reo- and Retroviruses (ex. Hepatitis B virus, Human immunodeficiency virus 1 and 2, and Human T-lymphotropic virus 1).
Fungal infections, as referenced herein, may include a mammalian infection by any bacterial species. Embodiments of the fungal species, as used herein, may include, but are not limited to, Fusarium oxysporum, Pneumocystis jirovecii, Aspergillus spp., Coccidioides immitis/posadasii, Candida albicans, Filobasidiella neoformans, Trichosporon, Encephalitozoon cuniculi, Enterocytozoon bieneusi, Mucor circinelloides, Rhizopus oryzae, and Lichtheimia corymbifera.
Parasite infections, as referenced herein, may include a mammalian infection by any parasite. Embodiments of the parasite may be, but are not limited to, Entamoeba histolytica, Babesia microti, Babesia sp. WA1, Trypanosoma cruzi, Taenia solium, Echinococcus granulosus, Leishmania braziliensis, L. donovani, L. tropica, Plasmodium falciparum, Plasmodium vivax, Plasmodium ovale and Plasmodium malariae, Plasmodium knowlesi, Paragonimus westermani, chistosoma sp., S. mansoni, S. haematobium, S. japonicum, Strongyloides stercoralis, Toxocara canis, Toxoplasma gondii, Trichinella spiralis African trypanosomiasis, Ancylostoma, Angiostrongylus, Anisakis, Baylisascaris procyonis, Clonorchis (Opisthorchis) sinensis, Echinococcus multilocularis, Fasciola hepatica, Filariasis, Gnathostoma
Exemplary embodiments of diagnostic markers include those listed in Table III.
Further exemplary embodiments for the diagnosis of type-II diabetes include, but are not limited to, Albumin, Alpha-1 antitrypsin (A1AT) - G Protein, Cystatin C, Cystatin A, alpha- 2-macroglobulin (A2MG), Uteroglobin, Transthyretin (TTR), Annexin 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, Angiogenin, Annexin Al, B-cell activating factor (BAFF), B-cell lymphoma 2 (BCL2)-like 2, Beta Human chorionic gonadotropin, Cal5-3, Calcyclin, Calvasculin, Cathepsin D, Caveolin-1, Chromogranin A, Alpha-crystallin B chain (CRYAB), Endostatin, Eotaxin-2, Epithelial cell adhesion molecule (EpCAM), Ezrin, fatty acid binding protein 4 (FABP4), Galectin-3, γ-glutamylcysteine ligase regulatory chain (GCLR), Gelsolin, Glucose 6-phosphate (G6P), Glycoprotein 130 (gpl30), Glutathione S-transferase Mu 1 (GSTM1), Hepsin, High-mobility group protein Bl (HMGB- 1), Insulin- like growth factor binding protein 1 (IGFBP-1), Insulin-like growth factor binding protein 4 (IGFBP-4), Insulin-like growth factor binding protein 5 (IGFBP-5), Insulin-like growth factor binding protein 6 (IGFBP-6), LGL, latency associated peptide (LAP), macrophage stimulating protein (MSP), MHC class I polypeptide-related sequence A (MICA), Nucleoside diphosphate kinase B (NME2), Neuron-specific Enolase (NSE), Osteopontin, Osteoprotegerin, Pepsinogen, Peroxiredoxin, Phosphoserine aminotransferase (PSATl), Prostate Specific Antigen, Receptor tyro sine-protein kinase erbB-3 (ErbB3), Serpin B3, Vascular smooth muscle cell growth factor R2 (VSGF R2/KDR), Vascular endothelial growth factor R3 (VEGF R3/Flt-4), Thyroglobulin, Tyrosine kinase with immunoglobulin- like and EGF-like domains 2 (TIE-2), Tissue plasminogen activator (tPA), Transforming growth factor beta (TGF-βΙ), Tumor necrosis factor receptor 1 (TNF-R1), urokinase-type Plasminogen Activator (uPA), urokinase-type Plasminogen Activator Receptor (uPAR), Brcal, Brcall, kallikreins, e-cadherin, Hox peptide, and Engrailed-2.
Diagnostic markers directed towards a bacterial or virus may include any surface or secreted antigen from the bacteria or virus, or a conserved nucleotide sequence.
Tumor Marker Relevant Present in Present in Class of
Cancer Serum/Plasma Saliva Molecules a-fetoprotein Heptacellular Yes Yes Protein
carcinoma
Cancer Antigen Breast cancer Yes Yes Protein
(CA15-3)
Cancer Antigen Ovarian cancer Yes Yes Protein Tumor Marker Relevant Present in Present in Class of
Cancer Serum/Plasma Saliva Molecules
(CA 125)
Carcinoembryonic Many epithelial Yes Yes
antigen cancers
Prostate specific Prostate cancer Yes Yes Protein antigen
c-erb-2 Breast cancer Yes Nucleic acid
P16, p53 Oral SCC
BRACA1, Breast cancer Yes Yes Nucleic acid BRACA2
Hemoglobin- Ale Yes Protein
Table III
B. Diagnostic Markers
Further, diagnostic markers according to an embodiment of the present disclosure may include one or more of: 1,25 dihydroxy-vitamin D, 17-Hydroxyprogesterone, 25-hydroxy- vitamin D, Antineutrophil Cytoplasmic Antibodies, 5-Hydroxy Tryptamine, 5- hydroxyindoleacetic acid, Acetoacetate, Activated Partial Thromboplastin Time, Adrenocorticotropic Hormone, Alanine aminotransferase, Alanine transaminase, Albumin, Albumin-to-Creatinine ratio, Albumin/Globulin ratio, Alcohol, Aldolase, Aldosterone, Aldosterone and plasma renin activity, Aldosterone and Renin, Alkaline Phosphatase, Allergen-specific IgE, Alpha tryptase, Alpha- 1 Antitrypsin, Alpha- fetoprotein, Alphal- antitrypsin, Alzheimer biomarkers (including Tau protein and Amyloid Beta 42 peptides), Amylase, ANCA Antibodies, ANCA/MPO/PR3 Antibodies, Angiotensin-Converting Enzyme, Anti-citrulline antibody, anti-cyclic citrullinated peptide antibody, Anti-retroviral drug resistance testing, anti-ribonucleoprotein, anti-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 A-I, Apolipoprotein B-100, Arginine Vasopressin, Aspartate aminotransferase, Aspartate transaminase, B-type natriuretic peptide, Beta hCG, Beta tryptase, Beta-2 Microglobulin, Beta-hydroxybutyrate, Beta-hydroxybutyric acid, Beta2 Microglobulin, Bicarbonate, Bilirubin, cholesterol, Blood clotting factors, Bordetella pertussis Antibodies, Borrelia burgdorferi antibodies, IgM/IgG, Brain natriuretic peptide, Breast Cancer Gene 1 and Breast Cancer Gene 2, c-ANCA, c-erbB-2, C-peptide, C-Reactive Protein, Caffeine, Calcidiol, Calcifidiol, Calcitonin, Calcitriol, Calcium, Calcofluor white stain, Cancer Antigen 125, Cancer Antigen 15-3, Cancer Antigen 19-9, Cancer antigen-breast, Cancer antigen-GI, Carbamazepine, Carcinoembryonic Antigen, Cardiac-specific Troponin I and Troponin T, Cardiolipin Antibodies, Catecholamines, Celiac Disease Tests, Ceruloplasmin, Chickenpox, Chickenpox and Shingles Tests, Chlamydia, Chloride, Cholesterol, Chromogranin A, Chymotrypsin, Citrulline antibody, Coagulation Factors, Cobalamin, Complement Component C3, Complement Component C4, Complexed PSA, Conjugated bilirubin, Copper, Corticotropin, Cortisol, Cotinine, Creatine Kinase, Creatine Kinase-MB, Creatinine, Cryoglobulin, Cryoprotein, Cyclic Citrullinated Peptide Antibody, Cyclosporine, Cystatin C, Cystic Fibrosis Gene Mutation Panel, Cystic fibrosis genotyping, Cytomegalovirus, Cytotoxic T-cells, Dehydroepiandrosterone Sulfate, Delta-aminolevulinic acid, Depakene, Depakote, Des-gamma-carboxy prothrombin, DHEA Sulfate, Diabetes mellitus autoantibody panel, Digoxin, Dilantin, Direct Anti-human Globulin test, Direct Antiglobulin Test, Direct bilirubin, Direct Low-density lipoprotein cholesterol, Dopamine, Drug screen, eGFR, Epidermal Growth Factor Receptor, Epinephrine, Epstein-Barr Virus Antibodies, Erythropoietin, Estradiol, Estriol, Estrogens, Estrone, Ethanol, F-Actin Antibody, Factor I, Factor V Leiden, Factor V Leiden Mutation and PT 20210 Mutation, Factor V Leiden mutation: Activated protein C resistance, Factor V R506Q, Ferritin, Fetal fibronectin, Fibrin degradation fragment, Fibrinogen, Fluorescent Antinuclear Antibody, Fluorescent treponemal antibody absorption, Folic Acid, Follicle-stimulating hormone, Fragment D-dimer, Fructosamine, Gamma-glutamyl transferase, Gamma-glutamyl transpeptidase, Gastrin, Genital Human Papillomavirus, Glucose-6-Phosphate Dehydrogenase, Glutamic Acid Decarboxylase Autoantibodies, Gluten-Sensitive Enteropathy Tests, Glycated Albumin, Glycated hemoglobin, Heavy Metals (such as lead, mercury, iron, copper, and zinc), Hemoglobin, Hemoglobin-binding Protein, Hemogram, Heparin Anti-Xa, Hepatitis A, Hepatitis B, Hepatitis C, Herpes Simplex Virus, Typel and Type 2, Herpes Zoster, Heterophile Antibodies, High-density lipoprotein cholesterol, High-sensitivity C-reactive protein, Homocysteine, Human calcitonin, Human chorionic gonadotropin, Human epidermal growth factor receptor, Human Growth Hormone, Human immunodeficiency virus antibody test, Human Immunodeficiency Virus Genotypic Resistance Testing, Human Leukocyte Antigen, Immunoreactive trypsinogen, Insulin Autoantibodies, Insulin C-peptide, Insulin-like Growth Factor - 1, Insulinoma- Associated-2 Autoantibodies, Intact PTH, Interstitial Cell Stimulating Hormone, Iron, Ischemia-Modified Albumin, Islet autoantibodies Islet Cell Cytoplasmic Autoantibodies, Janus Kinase 2, Ketone bodies Ketones, blood, Lactate, Lactate dehydrogenase, Lactic Acid, Lactic dehydrogenase, Lead, Lipoprotein, Lithium, Low-density lipoprotein cholesterol, Lupus Antibody, Luteinizing hormone, Lyme antibodies IgM/IgG by Western blot, Magnesium, Mast cell tryptase, Measles or Mumps IgM and IgG Antibodies, Mercury, Metanephrine and Normetanephrine, Methotrexate, Methylmalonic Acid, Microalbumin, Mitochondrial Antibody, Mononuclear heterophile test, Mycophenolic acid, Mycoplasma, Mycoplasma pneumoniae IgG and IgM antibodies, Myeloperoxidase Antibodies, Myoglobin, N-terminal pro b-type natriuretic peptide, Neonatal bilirubin, Nicotine, Norepinephrine, p24 antigen, Parathyroid Hormone, Parvovirus, Pertussis, Phenobarbital, Phenytoin, Phosphate, Phosphorus, Platelet-activating factor acetylhydrolase, porphobilinogen, Potassium, Prealbumin, Presenilin 1 gene, Procalcitonin, Progesterone, Proinsulin C-peptide, Prolactin, Prostate Specific Antigen, Protein C, Protein C and Protein S, Protein Tyrosine Phosphatase-like Autoantibodies, Proteinase 3 Antibodies, Prothrombin 20210 mutation, Prothrombin 20210 mutation: PT G20210A, factor II 20210, Red Blood Cell Antibody Identification, Renin, Respiratory Syncytial Virus, Rheumatoid Factor, Ristocetin Cofactor, Rubella, Scleroderma antibodies, Serine Protease 3, Serotonin, Siderophilin, Sirolimus, Smith antibody, Smooth Muscle Antibody, Sodium, Soluble Mesothelin-Related Peptides, Somatomedin C, Somatotropin, Syphilis, Tacrolimus, Tegretol®, Testosterone, Testosterone-estrogen Binding Globulin, Theophylline, Theophylline and Caffeine, Thiopurine methyltransferase, Thiopurine S-methyltransferase, Thymotaxin, Thyro calcitonin, Thyro globulin, Thyroid peroxidase antibody, Thyroid stimulating hormone receptor antibody, Thyroid stimulating immunoglobulin, Thyroid-stimulating hormone, Thyroperoxidase antibody, Thyrotropin, Thyroxine, Thyroxine-binding prealbumin, Transferrin, Transthyretin test, Treponema pallidum particle agglutination assay, Trichomonas, Trichomoniasis, Triglycerides, Triiodothyronine, Troponins, Trypsin, Trypsin and Chymotrypsin, Trypsinogen, Tryptase, Valproic acid, Vancomycin, Vanillylmandelic acid, , Varicella Zoster Virus, Vasopressin, Very Low-Density Lipoprotein Cholesterol, Viral Hepatitis A Antibody, Viral Hepatitis C. Viral Load (HIV), Vitamin B12, Vitamin B12 & Folate, Vitamin D, Vitamin D2, Vitamin D3, Vitamin K, von Willebrand Factor, West Nile Virus, Westergren sedimentation rate, and Zinc Protoporphyrin. C. Exemplary Targets for Stabilizing Agent(s)
1. Amylase
Amylase is an enzyme that breaks down starch into sugar, and is found in such places as human saliva, where it begins the chemical process of digestion. The a-amylases 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 a-amylase. Further, the stabilizing agent may be active in a biological fluid, such as in saliva. In an exemplary embodiment, a stabilizing agent may comprise one or more of Fixanal® Buffer 6.0 (Sigma-Aldrich Co.), aluminum sulfate hydrate, aluminum hydroxide, bentonite, and aluminum potassium sulfate dodecahydrate.
2. Lysozyme
Lysozyme, also known as muramidase or N-acetylmuramide glycanhydrolase, is a glycoside hydrolase, that damage bacterial cell walls by catalyzing hydrolysis of 1,4-beta- linkages between N-acetylmuramic acid and N-acetyl-D-glucosamine residues in a peptidoglycan and between N-acetyl-D-glucosamine residues in chitodextrins. Lysozyme is abundant in a number of secretions, such as tears, saliva, human milk, and mucus. It is also present in cytoplasmic granules of the polymorphonuclear neutrophils (PMN).
In at least one embodiment, a stabilizing agent of the present disclosure may be an inhibitor of lysozyme. Further, the stabilizing agent may be active in a biological fluid, such as saliva. In an exemplary embodiment, a stabilizing agent may comprise one or more of Fixanal® Buffer 6.0 (Sigma-Aldrich Co.), bentonite, benzoic acid, and acetic acid.
3. Peroxidase
Peroxidase are a class of oxidoreductase enzymes that catalyze the oxidation of a compound by the decomposition of hydrogen peroxide or an organic peroxide. These peroxidases may be found in many different bodily fluids. For example, saliva contains both salivary peroxidase (SPX) and myeloperoxidase (MPO). These peroxides may act on a number of different diagnostic markers found in bodily fluids to mask the detection or analysis of the markers.
In at least one embodiment of a stabilizing agent of the present disclosure, the stabilizing agent may be an inhibitor peroxidase activity. Further, the stabilizing agent may be active in a biological fluid, such as saliva, blood, serum, or cancer cells. In an exemplary embodiment, a stabilizing agent may comprise one or more of aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate.
4. Insulin Resistance
In at least one embodiment of a stabilizing agent of the present disclosure, the stabilizing agent may preserve a diagnostic marker for insulin resistance and/or glucose intolerance. Further, the stabilizing agent may be active in a biological fluid, such as saliva. In an exemplary embodiment, a diagnostic marker may be one or more of a-hydroxybutyrate, 1-linoleoyl-GPC, palmitate, Glycine, 3-methyl-2-oxybutyrate. Moreover, a diagnostic marker may be any marker of early stage insulin resistance and glucose intolerance in a non- diabetic individual.
5. Galactose Oxidase
Galactose oxidase is a member of the family of oxidoreductaases, and has been shown to participate in galactose metabolism. Because of this activity, galactose oxidase may be used to detect mucin-like glycoproteins. These glycoproteins are a major component of mucus secreted by epithelial and glandular cells and are primarily responsible for the protective properties of the viscoeleastic mucous barrier. Mucins have been implicated in the process of cholesterol gallstone formation, and has been identified as having abnormal expression in some cancers.
In at least one embodiment of a stabilizing agent of the present disclosure, the stabilizing agent may be an inhibitor of galactose oxidase. Further, the stabilizing agent may be active in a biological fluid, such as saliva. In an exemplary embodiment, a stabilizing agent may comprise one or more of aluminum potassium sulfate dodecahydrate, 3-tert-butyl- hydroxyanisole, benzoic acid, and acetic acid.
6. Glycated hemoglobin
Glycated hemoglobin (HbAlc) is a form of hemoglobin that is indicative of the average plasma glucose concentration over a period of time. Due to the transport of components of the plasma into bodily fluids, such as saliva, the level of HbAlc may be determined in bodily fluids in addition to plasma.
In at least one embodiment of a stabilizing agent of the present disclosure, the stabilizing agent may be an inhibitor of HbAlc degradation. Further, the stabilizing agent may be active in a biological fluid, such as saliva. In an exemplary embodiment, a stabilizing agent may comprise one or more of acetic acid, aluminum hydroxide bentonite, aluminum sulfate hydrate, aluminum potassium sulfate dodecahydrate, benzoic acid, caffeine, and 3- tert-butyl-hydroxyanisole, or a combination thereof.
7. Cancer Antigen 19-9 (CA 19-9)
Cancer Antigen 19-9 (CA 19-9), which may in some instances be referred to as Cancer Angigen 19.9, Cancer antigen-GI or CA-GI, is an antigen associated with various cancers, such as prostate and colon cancer. At least one use of CA 19-9 may be see whether a pancreatic tumor is secreting antigen. If that is the case, then the levels may decrease when the tumor is treated, and they may rise again if the disease recurs.
In at least one embodiment of a stabilizing agent of the present disclosure, the stabilizing agent may be an inhibitor of CA 19-9 degradation. Further, the stabilizing agent may be active in a biological fluid, such as saliva, blood, serum, or cancer cells. In an exemplary embodiment, a stabilizing agent may comprise one or more of aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate.
Further, in an additional exemplary embodiment, the components of a stabilizing agent may be present at or about equal amounts, such as for example 1 : 1 : 1 in a stabilizing agent with three active components. Additionally, the stabilizing agents may in at least one embodiment inhibit degradation or inactivation of a diagnostic marker by at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%o, at least about 70%>, at least about 75%, at least about 80%>, at least about 85%, at least about 90%, at least about 95%, or at least about 99%. Moreover, the stabilizing agent may, in at least one embodiment, inhibit degradation of a diagnostic marker for at least about 1 minute, at least about 5 minutes, at least about 10 minutes, at least about 15 minutes, at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 4 hours, or at least about 8 hours.
Additional Components for Stabilizing Agent Carriers:
According to at least one embodiment of the present disclosure, stabilizing agents may be incorporated into various carriers for their use with mammals. Such carriers may include various rinses, gums, lozenge, mouthwash, beverages, confectionary, washes, or other applicable vehicles to deliver a stabilizing agent to the site of a diagnostic marker.
Embodiments of a carrier, such as a rinse, according to the present disclosure may also comprise one or more additional component which may include an anti-caking agent, a chemical preservative, an emulsifying agent, a nutrient and dietary supplement, a sequestrant, a stabilizer, an additive, a synthetic and flavoring substance. Exemplary embodiments of these components are listed herein (See section entitled "Additional Components for Rinse"). Further, the one or more additional component may be a substance which has been labeled as Generally Recognized As Safe (GRAS) by the Food and Drug Administration of the United States of America.
At least one embodiment of a carrier of the present application may comprise a liquid. The liquid, in at least one embodiment, is a non-toxic liquid. Further, the liquid may comprise water, a beverage containing water, or glycerin. Moreover, in at least one embodiment, a stabilizing agent may be at least partially dissolved in the liquid.
It will be appreciated that the above list of excipients and/or additives is provided merely by way of example and that various other such components may be used in the formulation of the present invention.
The following components, in at least one embodiment of the rinse of the present disclosure, may include:
1. ANTI-CAKING AGENTS: aluminum calcium silicate, calcium silicate, magnesium silicate, sodium calcium aluminosilicate, and tricalcium silicate.
2. CHEMICAL PRESERVATIVES: ascorbic acid, ascorbyl palmitate, benzoic acid, butylated hydroxyanisole, butylated hydroxytoluene, calcium ascorbate, calcium propionate, calcium sorbate, caprylic acid, dilauryl thiodipropionate, erythorbic acid, gum guaiac, methylparaben, potassium bisulfite, potassium metabisulfite, potassium sorbate, propionic acid, propyl gallate, propylparaben, sodium ascorbate, sodium benzoate, sodium bisulfite, sodium metahisulfite, sodium propionate, sodium sorbate, sodium sulfite, sorbic acid, stannous chloride, sulfur dioxide, thiodipropionic acid, tocopherols.
3. EMULSIFYING AGENTS: cholic acid, desoxycholic acid, diacetyl tartaric acid esters of (M)mono- and diglycerides, glycocholic acid, mono- and diglycerides, monosodium phosphate derivatives of above, propylene glycol, ox bile extract, taurocholic acid.
4. NUTRIENTS AND DIETARY SUPPLEMENTS: alanine, arginine, ascorbic acid, aspartic acid, biotin, calcium carbonate, calcium citrate, calcium glycerophosphate, calcium oxide, calcium pantothenate, calcium phosphate, calcium pyrophosphate, calcium sulfate, carotene, choline bitartrate, choline chloride, copper gluconate, cuprous iodide, cysteine, cystine, ferric phosphate, ferric pyrophosphate, ferric sodium pyrophosphate, ferrous gluconate, ferrous lactate, ferrous sulfate, glycine, histidine, inositol, iron (reduced), isoleucine, leucine, linoleic acid, lysine, magnesium oxide, magnesium phosphate, magnesium sulfate, manganese chloride, manganese citrate, manganese gluconate, manganese glycerophosphate, manganese hypophosphite, manganese sulfate, manganous oxide, mannitol, methionine, methionine hydroxy analogue, niacin, niacinamide D- pantothenyl alcohol, phenylalanine, potassium chloride, potassium glycerophosphate, potassium iodide, proline, pyridoxine hydrochloride, riboflavin, riboflavin-5 -phosphate, serine, sodium pantothenate, sodium phosphate, sorbitol, thiamine hydrochloride, thiamine mononitrate, threonine, tocopherols, tocopherol acetate, tyrosine, valine, vitamin A, vitamin A acetate, vitamin A palmitate, vitamin B12, vitamin D2, vitamin D3, zinc sulfate, zinc gluconate, zinc chloride, zinc oxide, zinc stearate.
5. SEQUESTRANTS: calcium acetate, calcium chloride, calcium citrate, calcium diacetate, calcium gluconate, calcium hexametaphosphate, calcium phosphate, monobasic, calcium phytate, citric acid, dipotassium phosphate, disodium phosphate, isopropyl citrate, monoisopropyl citrate, potassium citrate, sodium acid phosphate, sodium citrate, sodium diacetate, sodium gluconate, sodium hexametaphosphate, sodium metaphosphate, sodium phosphate, sodium potassium tartrate, sodium pyrophosphate, sodium pyrophosphate, tetra, sodium tartrate, sodium thiosulfate, sodium tripolyphosphate, stecaryl citrate, tartaric acid.
6. STABILIZERS: acacia (gum arabic) , agar-agar, ammonium alginate, calcium alginate, carob bean gum, chondrus extract, ghatti gum, guar gum, potassium alginate, sodium alginate, sterculia (or karava) gum, tragacanth.
7. ADDITIVES: acetic acid, adipic acid, aluminum ammonium sulfate, aluminum potassium sulfate aluminum sodium sulfate, aluminum sulfate, ammonium bicarbonate, ammonium carbonate, ammonium hydroxide, ammonium phosphate, ammonium sulfate, bees wax, bentonite, butane, caffeine, calcium carbonate, calcium chloride, calcium citrate, calcium gluconate, calcium hydroxide, calcium lactate, calcium oxide, calcium phosphate, caramel, carbon dioxide, carnauba wax, citric acid, dextrans, ethyl formate, glutamic acid, glutamic acid hydrochloride, glycerin, glyceryl monostearate, helium, hydrochloric acid, hydrogen peroxide, lactic acid, lecithin, magnesium carbonate, magnesium hydroxide, magnesium oxide, magnesium stearate, malic acid, methylcellulose, monoammonium glutamate, monopotassium glutamate, nitrogen, nitrous oxide, papain, phosphoric acid, potassium acid tartrate, potassium bicarbonate, potassium carbonate , potassium citrate, potassium hydroxide, potassium sulfate, propane, propylene glycol, rennet, silica aerogel, sodium acetate, sodium acid pyrophosphate, sodium aluminum phosphate, sodium bicarbonate, sodium carbonate, sodium citrate, sodium carboxy-methylcellulose, sodium caseinate, sodium citrate, sodium hydroxide, sodium pectinate, sodium phosphate, sodium potassium tartrate, sodium sesquicarbonate, sodium tripolyphosphate, succinic acid, sulfuric acid, tartaric acid, triacetin, triethyl citrate. 8. SYNTHETIC FLAVORING SUBSTANCES: acetaldehyde, acetoin, aconitic acid, anethole, benzaldehyde, N-butyric acid, d- or 1-carvone cinnamaldehyde, citral, decanal, diacetyl, ethyl acetate, ethyl butyrate, ethyl vanillin, eugenol, 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 additional embodiment of a carrier, of the present disclosure, the carrier comprises an indicator compound capable of binding to and/or reacting with a target to produce an indicator signal. The target in an exemplary embodiment may be a diagnostic marker, such as the presence of glucose or glucose in excess of a defined threshold, the presence of one or more glycosylated protein related to diabetes or pre-diabetes, the presence of cancer (or pre-cancer) markers, and the presence of cardiac (or pre-cardiac) markers.
A. Indicators
The indicator compound in an exemplary embodiment of a carrier according to the present disclosure may be any compound, chemical, or biological component which may interact with a target or byproduct of a target. For example, an indicator compound may comprise an antibody, a reactive chemical compound, a labeled molecule, or any combination thereof. Antibodies used in an embodiment of the present disclosure may be monoclonal or polyclonal and derived from any species (e.g. human, rat, mouse, rabbit, pig). Further, indicator molecules may be aptamers, proteins, peptides, small organic molecules, natural compounds (e.g. steroids), non-peptide polymers, 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 isothiocyanate) Cy- Dyes o Cy2, Cy3, Cy3.5, Cy5, Cy5.5, Cy7 Fluorescent Proteins; o RPE(R- phycoerythrin), PerCp, APC(Allophycocyanin); other of phycobillin containing proteins, e.g. phycobiliprotein o Green fluorescent proteins (GFP);
• GFP and GFP derivated mutant proteins; BFP1CFP, YFP, DsRed, Tl , Dimer2, mPvFPl, MBanana, mOrange, dTomato, tdTomato, mTangerine, mStrawberry, mCherry Tandem dyes: o RPE-Cy5, RPE-Cy5.5, RPE-Cy7, RPE-AlexaFluor® tandem conjugates;
• RPE-Alexa610, RPE-TxRed o APC-Aleca600, APC-Alexa610, APC-Alexa750, APC-Cy5, APC-Cy5.5 Multi fluorochrome assemblies o Multiple fluorochromes attached to a polymer molecule, such as a peptide/protein, Dex, or poly-sacceride. o Any combination of the fluorescent dyes involving in generation of FRET;
• (Fluorescence resonance energy transfer) based techniques, Multiple fluorochromes associated or coupled to a polymeric molecule, or consisting of polymeric residues, lonophors; ion chelating fluorescent probes or Probes that change wavelength when binding a specific ion, such as calcium;
• Probes that change intensity when binding to a specific ion, such as calcium; and
• Combinations of fluorochromes on the same marker. The marker is not identified by a single fluorochrome but by a code of identification being a specific combination of fluorochromes, as well as inter related ratio's of intensities.
B. Target
The target of an exemplary indicator compound of the present disclosure may be any diagnostic marker or diagnostic condition for a disease state (or pre-disease state) of an individual. For example, the disease state may be diabetes, pre-diabetes, cardiac disease, 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, Kallikrein 3, Vascular endothelial growth factor A (VEGF), Calcitonin, Chromogranin A, Gastrin, SI 00 alpha chain, Somatostatin, Thyro globulin, V-erb-b2, cyckub-dependent kinase inhibitor 1 (p21), Breast Cancer Antigen 1 and 2 (BRCA1 and 2), MutL homolog 1 (MLH1), MutS homolog 2 (MSH2), MutS homolog 6 (MSH6), and postmeiotic segregation increased 1 and 2 (PMS1 and 2). Moreover, the target may be the presence or level of any diagnostic marker included herein.
C. Indicator Signal
The indictor signal in at least one embodiment of the present disclosure may comprise any detectable signal, including but not limited to, color change, fluorescence, and chemical/structural change of the target (and/or indicator compound) so as to be amenable to reacting with a secondary detection marker. Further, the detection of a signal may involve a secondary reactive molecule.
D. Glucose
In an embodiment of the present disclosure, glucose may be detected through an indicator rinse by a chemical or enzymatic method. Exemplary embodiments of this method may include alkaline copper reduction, alkaline ferricyanide reduction, glucose oxidase, and/or hexokinase.
In an exemplary embodiment of an indicator rinse of the present disclosure, the rinse may further comprise one or more additional component as described herein. These additional components may include stabilizing agents, as well as GRAS components (such as those listed herein).
E. Glycosylated Hemoglobin
In an embodiment of the present disclosure, glycated Hemoglobin Ale (HbAlc) may be detected through any known method for the detection of HbAlc or fragments thereof, such as high pressure liquid chromatography, immunoassays, and an indicator rinse by a chemical or enzymatic method. In an exemplary embodiment of an indicator rinse of the present disclosure, the rinse may further comprise one or more additional component as described herein. These additional components may include stabilizing agents, as well as GRAS components (such as those listed herein).
F. Cancer Antigen 19-9
In an embodiment of the present disclosure, cancer antigen 19-9 (CA 19-9) may be detected through any known method for the detection of CA 19-9 or fragments thereof, such as high pressure liquid chromatography, immunoassays, and an indicator rinse by a chemical or enzymatic method. In an exemplary embodiment of an indicator rinse of the present disclosure, the rinse may further comprise one or more additional component as described herein. These additional components may include stabilizing agents, as well as GRAS components (such as those listed herein).
III. Diagnostic Marker Detection
In an embodiment of the present disclosure, a diagnostic marker susceptible to degradation or alteration by a destructive component may be detected through any known method, including, but not limited to, high pressure liquid chromatography, immunoassays, and an indicator rinse by a chemical or enzymatic method. Further, the detection may be conducted in a micro well, deep well, microarray, microfluidic device, high throughput screen, or any applicable diagnostic device.
A. Rapid Diagnostic
Detection of a diagnostic marker may be conducted according to a rapid detection system of the present disclosure. Turning to Fig. 1A, a detection platform 100 may comprise a platform 102 having a plurality of test wells 104, where each well is capable of containing a plurality of test spots 106. The plurality of test wells 104 may include at least 2, at least 4, at least 8, at least 16, at least 32, at least 64, or at least 256 wells. The plurality of tests spots 106 may include at least 2, at least 4, at least 8, at least 16, at least 32, or at least 64 test spots 106. Further, each of the plurality of test spots 106 may comprise one or more diagnostic marker or control marker.
An embodiment of detection platform 100 may further be sized and shaped to allow for automated detection and analysis of a diagnostic or control marker. In at least one embodiment, the detection system 100 may comprise an identifier 108, such as a barcode or Radio-frequency identification (RFID) tag, to allow for the identification of the detection system 100 and data collected from the analysis of test spots 106.
Further, detection platform 100 may be comprised of any applicable material sufficient to house or define test wells 104. Exemplary embodiments of applicable materials for detection platform 100 may be a polymer, glass, metal, quartz, nylon, silica-based material, resin, or combination thereof. Further, embodiments of test wells 104 may include not only a depression defined or housed by platform 102, but may also include a defined area or region of the platform 102 (such as may be found on a microarray based platform).
Turning to Fig. IB, at least one embodiment of detection system 150 of the present disclosure is shown. An exemplary embodiment of detection system 150 comprises an embodiment of detection platform 100, a detection device 120 capable of determining a binding characteristic between a detection agent and a diagnostic marker on the detection platform 100, and a computer processor 130 coupled to a computer database 135 and to the detection device 120. In at least embodiment, the computer processor 130 controlling the detection device 120 is able to (1) determine the binding characteristic of a detection agent to a diagnostic agent, (2) compare the binding characteristic among each of a plurality of samples tested, (3) generate a binding report using he compared binding characteristics, and deliver the binding report to a recipient or external computer (not shown) in communication with the computer processor 130.
A binding characteristic as used herein may include any measurement of the attraction (or repulsion) of two molecules (such as a detection agent and a diagnostic marker).
Detection system 100 may also comprise an embodiment of a bead-based system of the present disclosure having at least one bead type. For example, a dual bead system may be used wherein at least one bead type is magnetic or paramagnetic. These beads may also be bound to a carrier protein with a conjugated hapten. Further, the beads may have an analyte capture monoclonal antibody reagent bound to it. Additionally, the beads, in at least one embodiment, do not retain their magnetic properties when removed from a magnetic field. In at least one embodiment, the beads may be Europium micro-particles. The sizes of the Europium micro-particles may be varied as needed to accommodate the method of detection used.
At least one embodiment of a bead based system may comprise a latex particle (See Figs. 2-4). The latex particle according to at least one embodiment may be a Carboxylate modified latex (CML) bead. Further, an exemplary embodiment of a bead may include bovine serum albumin (BSA) and/or human serum albumin (HSA). Additionally, an embodiment of a bead based system of the present disclosure may include a linker coupled to HSA and/or BAS. The linker according to an embodiment, may comprise a maleimide compound. At least one exemplary embodiment of a maleimide compound may be Succinimidyl 4-[N-Maleimidomethyl] Cyclohexane-1 -Carboxylate (SMCC) or N- Hydroxysuccinimide-activated hexa(ethylene glycol) undecane Thiol (NHS). Moreover, at least one exemplary embodiment of a bead based system may further comprise an antibody linked to maleimide on the bead. Accordingly, at least one embodiment of the bead based system comprises a latex particle coupled to BAS or HSA, maleimide coupled to BAS or HSA, and an antibody coupled to maleimide.
The latex particle, according to at least one embodiment may be in a size range from about 0.02μιη to about 7.0μιη. Turning to Fig. 5, at least one at least one method 500 of coupling an antibody to a bead comprises the step 502 of modifying an embodiment of a bead (such as a latex bead/particle) with BSA or HAS, wherein the BSA or HSA may further comprise a maleimide containing molecule, such as SMCC or NHS. Additionally, an embodiment of the method 500 may further comprise the step 504 of attaching a diagnostic marker binding agent (such as an antibody) to the bead. Step 504 of attaching the diagnostic marker binding agent to the bead may be accomplished through binding the diagnostic marker binding agent to maleimide. Further, method 500 may also comprise the step 506 of attaching one or more stabilizing agent to an embodiment of a bead. Additionally, while an embodiment of method 500 may use latex beads for attachment of a maleimide containing molecule and/or a stabilizing agent, alternate embodiments of the bead may comprise silicon, a polymer, or other applicable materials.
According to at least one embodiment of the present disclosure, one or more stabilizing agent may be attached or adsorbed to any one of an embodiment of a detection platform, test strip, microchip, or micro fluidic device.
B. Test Strip
Turning to Figs. 6A and B, exemplary embodiments of a diagnostic testing device 600 are described. An exemplary embodiment of diagnostic testing device 600 comprises housing structure 602 which comprises a collection chamber 604 capable of receiving body fluid, at least one membrane strip 606 in fluid communication with collection chamber 604, an immunoassay-based fingerprint acquisition pad 608 in fluid communication with the collection chamber 604, and a plurality of reaction zones 610 which may allow the visual display of the presence of a predetermined diagnostic marker. For example, reaction zone 610 may in an exemplary embodiment be capable of displaying a characteristic of the predetermined diagnostic marker, such as the presence of, the concentration of, or a concentration above or below a predetermined value for the predetermined diagnostic agent.
In an exemplary embodiment of diagnostic testing device 600, diagnostic testing device 600 further comprises a fluid collector 612 which is operable to collect body fluid from a subject, and wherein the fluid collector is capable of supply collected body fluid to collection chamber 604.
Exemplary embodiments of test strip 606 may comprise any material capable of adsorbing or attaching a stabilizing agent and may bind a diagnostic marker. Further, exemplary embodiments of test strip 606 may comprise compositions comprising a polyvinyl chloride-silica combination, nitrocellulose, or any suitable synthetic, resinous material. At least one embodiment, housing 602 is capable of reducing the exposure of the test strip 606 at sites other than the collection chamber 604 to foreign material. Test strip 606 in at least one embodiment may be capable of interfacing with an analysis device 615. The analysis device 615 in an exemplary embodiment may be capable of measuring the level of the predetermined diagnostic marker in the sample.
Turning to Fig. 6C, an exemplary embodiment of a microarray system 620 is described. An exemplary embodiment of microarray system 620 may comprise a microarray product 622 having a microarray identifier 624, and a plurality of microarray sites 626, a control microarray product 628 located on at least one microarray site 626 and bound to microarray product 622, and operably connected to a computer processor 130 for providing information regarding the identification and concentration of markers on the microarray product 622 based on the microarray identifier 624. Additionally, microarray product 622 may further comprise, in at least one embodiment, an embodiment of a stabilization agent 630 located on at least one microarray site 626 and bound to microarray product 622. Further, microarray product 622 may comprise one or more diagnostic marker 632 located on at least one microarray site 626 and bound to microarray product 622. Moreover, microarray system 620, in at least one embodiment, may further comprise a microarray detector 634 operable to detect a signal from at least one of the control microarray product 628 and/or diagnostic marker 632.
A microchip or microarrays may refer to an array of distinct polynucleotides affixed to a substrate, such as glass, plastic, paper, nylon or other type of membrane, filter, chip, or any other suitable solid support. The polynucleotides can be synthesized directly on the substrate, or synthesized separate from the substrate and then affixed to the substrate. In an embodiment, the microarray may be prepared and used at least according to the methods described in U.S. Pat. No. 5,837,832, Chee et al, PCT application W095/11995 (Chee et al), Lockhart, D. J. et al. (1996; Nat. Biotech. 14: 1675-1680) and Schena, M. et al. (1996; Proc. Natl. Acad. Sci. 93: 10614-10619), all of which are incorporated herein in their entirety by reference.
Turning to Fig. 6D, an exemplary embodiment of microfluidic device 640 is described. An embodiment of microfluidic device 640 comprises a sample reservoir 642 for receiving a body fluid (or other fluid sample) through a sample input 644, where the sample reservoir 642 is fluidly connected to a detector array 646. Additionally, in at least one embodiment of microfluidic device 640, detector array 646 comprises a reaction site 647 and a results display 648. Moreover, an exemplary embodiment of microfluidic device 640 may further comprise a filter device 650 capable of separating at least one unwanted component from a fluid sample and fluidly coupled between sample reservoir 642 and detector array 646. Detector array 646 may also, in at least one embodiment comprise a control reagent display 652, where at least one control reagent may be visually detected.
Further, microfluidic device 640 may also, in at least one embodiment, be able to couple to a processor 655. Processor 655 may be able to compare at least one reading, such as from results display 648 or control reagent display 652 from an embodiment of microfluidic device 640 to at least one additional stored reading on a computer database 657 of the processor 655. An embodiment of microfluidic system 660 of the present disclosure, may comprise one or more microfluidic device 640 operationally coupled to a processor 655.
Embodiments of microfluidic devices 640, which may also be referred to as "lab-on-a- chip" systems, biomedical micro-electro-mechanical systems (bioMEMs), or multicomponent integrated systems, are exemplary kits/systems for analyzing polynucleotide regions. Such systems may miniaturize and compartmentalize processes such as probe/target hybridization, nucleic acid amplification, and capillary electrophoresis reactions in a single functional device. Such microfluidic devices typically utilize detection reagents in at least one aspect of the system, and such detection reagents may be used to detect one or more polynucleotide region of the present disclosure. Exemplary microfluidic devices 640 may comprise a pattern of microchannels designed onto a glass, silicon, quartz, or plastic wafer included on a microchip. The movements of the samples may be controlled by electric, electroosmotic or hydrostatic forces applied across different areas of the microchip to create functional microscopic valves and pumps with no moving parts. Varying the voltage can be used as a means to control the liquid flow at intersections between the micro-machined channels and to change the liquid flow rate for pumping across different sections of the microchip.
In at least one embodiment of a microarray or microfludic device 640, the microarray or microfluidic device may comprise one or more stabilizing agent capable of reducing the degradation of a predefined diagnostic marker. For example, a stabilizing agent may be incorporated onto the surface of the microarray device, and/or on at least one part of the microfluidic device.
Turning to Fig. 6E, at least one embodiment of a sample collection device 665 of the present disclosure is shown. At least one embodiment of sample collection device 665 comprises an intake tube (or first tube) 666 having a first end and a second end, where the second end is coupled to a suction device 667. Suction device in at least one embodiment is also coupled to a dispensing tube 668 (or second tube) having a first and second end at the first end. In at least one embodiment of sample collection device 665, intake tube 666 comprises a filter 669 capable of removing at least one component of a body fluid before entering dispensing tube 668. Further, at least one embodiment of sample collection device 665 may also comprise securing devices 670 to fluidly seal either, or both of, the first end of the intake tube 666 and the second end of the dispensing tube 668. Moreover, an embodiment of sample collection device 665 may also comprise an effective amount of an embodiment of a stabilizing agent of the present disclosure.
In at least one embodiment of sample collection device 665, device 665 may be operated by placing device 665 in contact with a fluid source, using the suction device 667 to provide negative pressure so as to pull a portion of the fluid source into sample collection device 665. Optionally, the portion of the fluid source may pass through filter 669 where at least one component of the body fluid may be removed. Additionally, the process of intake of the portion of fluid source may also initiate the contact of the of the portion of the fluid source with a stabilization compound housed within the sample collecting device. The stabilized portion of the body fluid may then be dispensed in a controlled fashion through the dispensing tube 668 by positive pressure provided by suction device 667. In at least one embodiment of sample collection device 665, the dispensing tube 668 is structured so as to deliver a predetermined amount of liquid per drop. The predetermined amount of liquid per drop may be ΙΟμΙ, 30μ1, 50μ1, or ΙΟΟμΙ in at least one embodiment of sample device 665.
Turning to Fig. 6F, at least one embodiment of fluid collection device 672 is shown.
In at least one embodiment, sample collection device 672 comprises a housing 673 having a top and bottom opening (not shown), a tubular compartment 675 within housing 673 and in fluid communication with both top and bottom openings, a swab portion 674 coupled to the bottom opening and in fluid communication with tubular compartment 675, and a stabilizing agent contained within the tubular compartment 675 . At least one embodiment of fluid collection device 672 may also comprise a sealing mechanism 680 capable of fluidly sealing the tubular compartment 675 and one or both of the top and bottom openings.
In at least one embodiment of fluid collection device 672, housing 673 may comprise a sample compartment 675 coupled to the top and bottom openings, and a stabilization compartment 676 containing an embodiment of stabilization agent 677. In at least one embodiment, compartments 675 and 676 are connected by mixing tube 678, but fluid communication is prevented by removable barrier 679. Barrier 679 may be operable to allow fluid communication between compartments 675 and 676 upon mechanical or electrical activation by a user. Optionally, an embodiment of a stabilization agent 677 may also be provided in fluid collection device 672 in at least one of swab portion 674 and sample compartment 675.
Turning to Fig. 6G, at least one embodiment of a transport system 682 is shown. In at least one embodiment of the present disclosure, transport system 682 comprises an inner container 684 contained within outer container 685. Inner container 684, in an exemplary embodiment, has an inner compartment 686 and an opening 687. Inner compartment 686 may be sized and shaped to be capable of receipt of a sample through opening 687. Additionally, inner compartment 686 may comprise a solid support 689, where solid support 689 comprises or has attached an embodiment of a stabilizing agent 690. Inner container 686 may be reversibly sealed to prevent liquid release from the inner compartment 686. Further, transport system 682 may further comprise an absorbent material 691 contained within outer container 685 and sufficient to absorb the liquid contents of inner container 684 in the event of an unexpected release of the liquids contained therein. In at least one embodiment, transport system 682 is structured in such a manner, and stabilizing agent 690 contained therein has the properties, to prevent the degradation of a diagnostic agent in a liquid sample stored within the inner container 684 for a period of at least twenty-four hours.
Turning to Fig. 6H, at least one embodiment of a stabilizing system 692 is shown. In at least one embodiment of the present disclosure, stabilizing system 692 comprises a sample container 694 having an opening 695 and at least one rigid chamber 696 for containing a fluid sample. Additionally, stabilizing system 692 may also contain an effective amount of an embodiment of stabilizing agent 697 that may be adsorbed to, or part of, solid support 698. Stabilizing system 692 may be reversibly sealed with securing device 699 that may seal the opening 695.
IV. Methods of Biomarker Stabilization
In at least one embodiment of a method for stabilizing biomarkers, as described in
Fig. 7, the method 700 comprises the step 702 of mixing a body fluid with an embodiment of a stabilizing agent. Optionally, the step of mixing a body fluid with a stabilizing agent may comprise the step 704 of introducing the stabilizing agent into the patient, such as through the oral cavity, so that the stabilizing agent mixes with the bodily fluid, and the step 706 of retrieving a body fluid comprising a stabilized diagnostic marker. In an additional exemplary embodiment of a method of stabilizing a diagnostic marker, the method comprises the step 708 of isolating a body fluid having a diagnostic marker, the step 710 of treating the body fluid with an embodiment of a carrier, such as a rinse, gum, or beverage, as described herein, and the step 712 of analyzing the diagnostic marker. Isolation of the body fluid may occur through any customary mechanism, including, but not limited to, rinsing, swabbing, suction, collection, and lavage. Further, the rinse in an exemplary embodiment of a method 700 of the present disclosure, may be ingested to stabilize a diagnostic marker present in the bodily fluid, such as in urine, semen, anal secretions, and vaginal secretions.
The step 702 of mixing a body fluid with a carrier may occur prior to, or after isolation step 708 of the body fluid. As an exemplary embodiment, a rinse may be used to rinse the mouth. Afterwards, the rinse containing the treated body fluid may be collected by spitting, suction, or other means. Alternately, expirated saliva may be mixed with a rinse to treat the saliva ex vivo. Similar treatment of body fluids may be performed with any type of body fluid.
In analyzing the treated body fluid, the diagnostic marker may be analyzed through any known means. Optionally, the analysis of the treated body fluid may include the separation of solid materials from soluble materials through means such as filtration, or by centrifugation. Analysis of the treated body fluid may use any appropriate technique, such as western blot analysis, Enzyme-Linked Immunosorbent Assay (ELISA), protein activity assays, reverse transcription polymerase chain reaction (RT-PCR), microarray, high pressure liquid chromatography, or any comparable assay to determine a characteristic of the diagnostic marker. Such an analysis, in an exemplary embodiment, may be of a modified product, a cleavage product, or cleavage pattern, of a diagnostic marker.
In an exemplary embodiment of the analyzing step 712, the treated body fluid may be placed in contact with an embodiment of a test strip, such as a nitrocellulose strip, prior to detection with an appropriate probe specific for the diagnostic marker. The nitrocellulose strip may be designed to trap or filter particulates in the body fluid. This may reduce or eliminate potential contaminants or other substances in the oral fluid that would otherwise reduce the signal to noise ration during the detection step. A probe (such as an antibody) with affinity to the diagnostic marker may be affixed or immobilized to a specific location on the nitrocellulose membrane for detection of the analyte. Utilizing standard principles of immunoassay detection (or nucleotide detection), the signal generated may be visible to the eye or detectable by an instrument.
In an exemplary embodiment of a method of analyzing a stabilized diagnostic marker of the present disclosure as depicted in Fig. 8, the method 800 comprises the step 802 of operating a system for the detection of a diagnostic marker in a body fluid, where the system comprises a diagnostic device having a plurality of test wells each capable of containing at least one diagnostic marker binding agent, and a detection device capable of interacting with diagnostic device, wherein the detection device is capable of detecting an interaction between the at least one diagnostic marker binding agent and a diagnostic marker. Following step 802, the sample of bodily fluid may be diluted in step 803 with a reagent which may contain an embodiment of stabilizing agent as described herein. At least one embodiment of method 800 further comprises the steps of: contacting 804 a sample of a bodily fluid with at least one diagnostic marker binding agent, incubating 806 the contacted sample in the test well at a first temperature for a pre-determined period of time. The step 804 of contacting of the sample of bodily fluid with at least one diagnostic marker binding agent may be accomplished through a magnetic field applied to the test well.
In at least one embodiment of method 800, the first temperature may be at room or at body temperature (such as about 22°C or about 37°C). The pre-determined period of time may, for example, be between five and fifteen minutes. The incubated and contacted sample may then be removed in step 808 from the plurality of test wells, and the test wells analyzed in step 810 to detect a binding event with the detection device. Alternately, a stabilizing agent may be incubated with the diagnostic marker binding agent (also referred to as a detection agent) prior to introduction of a bodily fluid. In at least one embodiment of the method, each diagnostic marker binding agent may have a different monoclonal antibody designed to bind a specific hapten molecule.
Following the detection of a characteristic of a treated diagnostic marker (the results of which may be termed a "profile" such as a "cancer profile"), the characteristic may in step 812 be compared to a standard value to determine the presence or absence of a disease state. The characteristic determined may be an activity level, the concentration of the diagnostic marker, or a particular modification, such as glycosylation or methylation.
Optionally, an embodiment of the method 800 may further comprise the step 814 of customizing a detection method through screening of applicable pre-clinical samples through a database of stabilizing agents/cocktails. Further, an embodiment of method 800 of diagnostic marker detection may further comprise the step 816 of comparing a level or characteristic (such as a predictable degradation products) of a diagnostic agent with a library of known characteristics. Moreover, an exemplary embodiment of method 800 may additionally comprise the step 818 of determining a diagnosis of a disease state using the compared characteristic. In at least one embodiment, a method of diagnostic marker detection may be automated, and capable of detecting at least about ten thousand samples in a twenty-four hour period. Method 800 in at least one embodiment, may be at least partially completed by a computer processor. Additionally, the processor may be in communication with at least one more processor and/or a computer database. For instance, in at least one embodiment of method 800, most of the steps of the method may be completed with or monitored by a computer processor. Moreover, method 800, in at least one embodiment, may use a computer processor to perform additional step 820 of generating a report by using a comparison of determined characteristics (such as a binding report where a comparison is performed of binding characteristics), and step 822 of delivering the report to a recipient (such as a user, or secondary processor).
A method of biomarker stabilization may, in an exemplary embodiment, may be used to diagnose a particular disease state or condition of a patient. For example, a disease state or condition as used with this method may include one or more of Acid-Base Disorders, Acidosis and Alkalosis, Acidosis/Alkalosis, Acute inflammatory demyelinating polyneuropathy, Acute myocardial infarct, Addison's Disease, Adrenal Insufficiency, Adrenal Insufficiency & Addison's Disease, Alcohol dependence, Alcoholism, Allergies, Alzheimer's Disease, Anemia, Angina Pectoris, Anthrax, Arthritis, Asthma, Atypical Pneumonia, Autoimmune Disorders, Autoimmune thyroiditis, Avian flu, Benign Prostatic Hyperplasia, Benign Prostatic Hypertrophy, Bioterrorism Agents, Bleeding Disorders, Bone Marrow Disorders, Breast Cancer, Cardiovascular Disease, Celiac Disease, Cervical Cancer, Chlamydia, Chronic Fatigue and Immune Dysfunction Syndrome, Chronic Fatigue Syndrome, Chronic thyroiditis, Colon Cancer, Community-Acquired Pneumonia, Congestive Heart Failure, Conn's Syndrome, Cushing's Syndrome, Cystic Fibrosis, Degenerative Joint Disease, Diabetes, Diarrhea, Diffuse thyrotoxic goiter, Diseases of the Pancreas, Disseminated lupus erythematosus, Double pneumonia, Down Syndrome, Encephalitis, Endocrine Syndromes, Endocrine System and Syndromes, Epilepsy, Fibromyalgia, Flu, Folate Deficiency, Fungal Infections, Genital Herpes, Gonorrhea, Gout, Gouty arthritis, Graves' Disease, Guillain-Barre Syndrome, Influenza H1N1, Hashimoto's Thyroiditis, Healthcare-Associated Pneumonia, Heart Attack, Heart Attack and Acute Coronary Syndrome, Heart Disease, Hemochromatosis, Hepatitis, Herpes, Herpes Zoster, High blood pressure, Hospital-Acquired Pneumonia, Human Immunodeficiency Virus, Human Papillomavirus, Hypercoagulable Disorders, Hypersensitivity, Hypertension, Hyperthyroidism, Hypothyroidism, Infectious Arthritis, Infectious polyneuritis, Infertility, Inflammatory Bowel Disease, Influenza, Influenza A, Influenza B, Insulin Resistance, Jaundice, Juvenile Rheumatoid Arthritis, Kidney and Urinary Tract Function, Disorders, and Diseases, Kidney Disease, Landry's ascending paralysis, Lead Poisoning, Leukemia, Liver Disease, Lobar pneumonia, Lower Respiratory Tract Infection, Lung Diseases, Lupus, Lupus erythematosus, Lyme Disease, Lymphoma, Malnutrition, Meningitis, Meningitis and Encephalitis, Menopause, Metabolic Syndrome, Metabolic Syndrome / Syndrome X, Multiple Myeloma, Multiple Sclerosis, Myeloproliferative Disorders, Myocardial infarct, Neural Tube Defects, Nontuberculous Mycobacteria, Osteoarthritis, Osteoporosis, Ovarian Cancer, Pancreatic Cancer, Pancreatic Diseases, Pancreatic Insufficiency, Pancreatitis, Pelvic Inflammatory Disease, Peptic Ulcer, Pituitary Disorders, Pneumonia, Polycystic ovarian syndrome, Pregnancy, Primary hyperaldosteronism, Prostate Cancer, Proteinuria, Rheumatoid Arthritis, Septic Arthritis, Sexually Transmitted Diseases, Sexually transmitted infections, Shingles, Sickle Cell Anemia, Sickle Cell Disease, 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 B12 and Folate Deficiency, Vitamin B12 Deficiency, Vitamin K Deficiency, Walking pneumonia, West Nile Virus, Wilson's Disease, Wound and Skin Infections.
V. Methods to Determine Efficacy and Specificity of a Therapeutic
According to Fig. 9, in at least one embodiment of the present disclosure, a method 900 of improving biomarker availability is described and shown. Method 900 comprises the step 902 of contacting a diagnostic marker from a patient with a stabilization agent, the step 904 of introducing a therapeutic agent to the diagnostic marker and the stabilization agent, and the step 906 of analyzing the interaction of the therapeutic agent with the diagnostic marker to determine a therapeutic profile, such as the binding affinity of the agent to the diagnostic marker. Further, method 900 may also comprise the step 908 of comparing the therapeutic profile with at least one other therapeutic profile to determine a stabilizing impact from the stabilizing agent on the diagnostic marker. Step 908 of comparing a plurality of therapeutic profiles may be performed using a computer processor and may further utilize a computer database of previously documented therapeutic profiles. Moreover, in an exemplary embodiment of method 900, the step 902 of contacting a diagnostic marker with a stabilization agent may be repeated with a plurality of different stabilization agents and/or therapeutic agents. Additionally, the therapeutic profile may be used to determine a therapeutic course to treat the patient.
In Fig. 10, a method of identifying diagnostic markers of a disease state is shown. Method 1000, according to at least one embodiment, comprises the step 1010 of contacting a bodily fluid from a subject having a disease state with a stabilizing agent, and the step 1020 of analyzing the stabilized bodily fluid to determine at least one property of a diagnostic agent, to create a disease state profile. An exemplary embodiment of method 1000 may further comprise the step 1030 of comparing the disease state profile with a profile generated in a like manner from a subject not having the disease state, and the step 1040 of generating a disease state identifier panel from the compared disease state profile. In at least one embodiment of method 1000, the disease state is cancer. Exemplary embodiments of cancer may include at least bladder cancer, lung cancer, breast cancer, melanoma, colon and rectal cancer, non-hodgkin lymphoma, endometrial cancer, pancreatic cancer, kidney (renal cell) cancer, prostate cancer, leukemia, and thyroid cancer. In at least one embodiment of method 1000, the subject is currently receiving treatment with an original treatment agent, and the disease state profile may be compared with a disease profile of the same subject while receiving a different treatment agent, a different concentration of the same treatment agent, or at an earlier point of time.
In Fig. 11, an exemplary embodiment of a method for screening biomarkers is described. Method 1100 comprises the steps of (a) contacting 1110 a cell from a subject having a disease state with a stabilizing agent, (b) incubating 1120 the stabilized cell with a therapeutic compound, and (c) analyzing 1130 the incubated cell for at least one diagnostic marker to create a marker profile. Optionally, method 1100 may further comprise the step 1140 of comparing the marker profile with at least one additional marker profile to determine the efficacy of the therapeutic compound.
In at least one method of stabilizing diagnostic markers of the present disclosure, as shown in Fig. 12, the method 1200 comprises the step 1210 of measuring the level of diagnostic marker in a subject with a disease state, the step 1220 of treating the subject with a therapeutic agent, and the step 1230 of determining the post-treatment level of diagnostic marker. Wherein the step 1210 of measuring the level of diagnostic marker includes the step of incubation of the diagnostic marker with a stabilization agent.
In at least one embodiment of method 900, 1000, 1100, or 1200, the method may be used with a device as described herein, such as an embodiment of diagnostic system 100, test strip 600, a microarray 640, or microfluidic device 660, any of which may also be performed in conjunction with a computer processor.
VI. Computational analysis
In at least one embodiment of the systems, methods, and devices of the present disclosure, a computer processor and a database may be used in or during the system, method, or device. An exemplary embodiment of a system framework comprising at least one computer processor and at least one database, as may be used in the embodiments of the present disclosure is shown in Fig. 13.
In at least one embodiment, a result from a diagnostic method may be compared using a computer processor to at least one additional result from the diagnostic test, and/or to a result stored on a database. Such a comparison, in at least one embodiment of the present disclosure, may reveal the stabilizing agent with a greater effect on the binding characteristic, may allow for the diagnosis of a disease state or health/lifestyle characteristic. Further, such a comparison, in at least one embodiment, may be used to develop, confirm, or modify a therapeutic treatment of a patient having a disease state or health/lifestyle characteristic.
As shown in exemplary system framework 1300 shown in Fig. 13, one or more user computers 1302 may be operably connected to a system server 1304. A user computer 1302 may be a computer, computing device, or system of a type known in the art, such as a personal computer, mainframe computer, workstation, notebook computer, laptop computer, hand-held computer, wireless mobile telephone, personal digital assistant device, and the like.
One or more administrator computers 1306 may also be operably connected to system server 1304 including through a network 1308 such as the Internet. Administrator computers 1306, similar to user computers, may be computers, computing devices, or systems of a type known in the art, such as personal computers, mainframe computers, workstations, notebook computers, laptop computers, hand-held computers, wireless mobile telephones, personal digital assistant devices, and the like. In addition, user computers and administrator computers may each comprise such software (operational and application), hardware, and componentry as would occur to one of skill of the art, such as, for example, one or more microprocessors, memory, input/output devices, device controllers, and the like. User computers and administrator computers may also comprise one or more data entry means (not shown in Fig. 13) operable by a user of client computer and/or an administrator computer, such as, for example, a keyboard, keypad, pointing device, mouse, touchpad, touchscreen, microphone, and/or other data entry means known in the art. User computers and administrator computers also may comprise an audio display means (not shown in Fig. 13) such as one or more loudspeakers and/or other means known in the art for emitting an audibly perceptible output. The configuration of user computers and administrator computers in a particular implementation of one or more systems of the present disclosure is left to the discretion of the practitioner.
System server 1304 may comprise one or more server computers, computing devices, or systems of a type known in the art. System server 1304 may comprise server memory. System server 1304 may comprise one or more components of solid-state electronic memory, such as random access memory. System server 1304 may also comprise an electromagnetic memory such as one or more hard disk drives and/or one or more floppy disk drives or magnetic tape drives, and may comprise an optical memory such as a Compact Disk Read Only Memory (CD-ROM) drive. System server 1304 may further comprise such software (operational and application), hardware, and componentry, as would occur to one of skill of the art, such as, for example, microprocessors, input/output devices, device controllers, video display means, and the like.
System server 1304 may comprise one or more host servers, computing devices, or computing systems configured and programmed to carry out the functions allocated to system server 1304. System server 1304 may be operated by, or under the control of, a "system operator," which may be an individual or a business entity. For purposes of clarity, System server 1304 is shown in Fig. 13 and referred to herein as a single server. System server 1304 need not, however, be a single server. System server 1304 may comprise a plurality of servers or other computing devices or systems connected by hardware and software that collectively are operable to perform the functions allocated to the various systems of present disclosure. Specifically, system server 1304 may be operable to be a web server, configured and programmed to carry out the functions allocated to a system server according to the present disclosure. Further, although user computers 1302 and administrator computers 1306 may be connected directly to system server 1304, these computers may be connected to system server 1304 through any suitable network, such as network 1308. Further, in one embodiment, the users need not be provided access to system server 1304, but instead the content posts from users are made by the user(s) and saved to one or more particular locations and the posts are accessed or harvested by the administrator or system automatically.
System server 1304 may be operably connected to the various user computers 1302 and/or an administrator computers 1306 by network 1308, which in an embodiment of the present disclosure comprises the Internet, a global computer network. However, network 1308 need not comprise the Internet. Network 1308 may comprise any means for electronically interconnecting system server 1304 and a user computer 1302 and/or an administrator computer 1306. Thus, it will be appreciated by those of ordinary skill in the art that the network 1308 may comprise the Internet, the commercial telephone network, one or more local area networks, one or more wide area networks, one or more wireless communications networks, coaxial cable, fiber optic cable, twisted-pair cable, the equivalents of any of the foregoing, or the combination of any two or more of the foregoing. In an embodiment where system server 1304 and user computer 1302 and/or an administrator computer 1306 comprise a single computing device operable to perform the functions delegated to both system server 1304 and user computer 1302 and/or an administrator computer 1306 according to the present disclosure, network 1308 comprises the hardware and software means interconnecting system server 1304 and user computer 1302 and/or an administrator computer 1306 within the single computing device. Network 1308 may comprise packet switched facilities, such as the Internet, circuit switched facilities, such as the public switched telephone network, radio based facilities, such as a wireless network, etc.
The various systems, methods, schema, ontologies, and architectures of the present disclosure may be used for purposes outside of the medical transcription field as referenced in the various examples cited herein. For example, the system for analyzing verbal records may comprise various components and relationships suitable for use in any number of areas where various experiences are utilized and processed, with feedback being fed back into system componentry to improve overall system outcomes. In addition, various components described herein may share a name (or a portion thereof) but have duplicative reference numbers, and therefore the descriptions for the various components should read in view of one another.
In addition, and regarding the various systems of the present disclosure, such systems may be operable, as desired by a user of such systems, to generate visual, electronic (video, audio, database, transcript, etc.), and/or printed reports, outputs, outcomes, and the like. Such exemplary outputs may be used for any number of purposes, and may be useful generally to "report" results, data, and/or knowledge contained within and generated from such systems. Furthermore, the disclosure of the present application further encompasses uses of the various methods, systems, architectures, etc., to perform various tasks in connection therewith.
EXAMPLES:
Examples
1. a- Amylase Inhibition Saliva Screening Method
At least one assay used for the detection of a-amylases activity, as used herein includes the use of a chromagenic substrate, 2-chloro-p-nitrophenol linked to maltotriose. Enzymatic activity of a-amylase on the substrate yields 2-chloro-p-nitrophenol, which can be measured spectrophotometrically at 405nm. The amount of a-amylase activity present in the experimental sample is directly proportional to the increase in absorbance at 405nm. For the assay, test compounds are mixed with saliva, prior to the addition of a- amylase substrate. The test compounds, such as GRAS materials, are prepared at a concentration of 5,000 ppm in distilled water. Following this preparation, 300μ1 of a- amylase substrate (Salimetrics a-amylase Assay Kit) that has been pre-heated to 37°C is added to 20μ1 of the test compound. To initiate the assay, ΙΟμΙ of a dilute (3.5%) or full strength (100%) saliva solution is added to the mixture. Following the initiation, each mixture is measured at 1 minute intervals using a spectrophotometer (such as Molecular Devices M5 reader) at 405nm. Temperatures of the mixtures are kept constant at 37°C during the assay.
2. a- Amylase Inhibitor Assays.
A series of GRAS compounds were tested using the a-amylase assay protocol described here. The tested compounds are included in Table I. From the screen of GRAS compounds, five compounds (CDI-030,034, 036,037 and 042) were shown to have a- Amylase Inhibitor Activity in dilute (3.5%) saliva (See Fig. 14). Further details of these five compounds may also be seen in Table 4.
Figure imgf000052_0001
Saliva Amjlase
Time CDI-30 CDI-34 CDI-36 CDI-37 CDI-42 Control Control
0 min 1.60 0.40 3.63 4.00 0.60 4.00 0.03
2.03 0.36 3.20 3.85 1.13 4.00 0.03
2.02 0.40 3.46 4.00 0.90 4.00 0.03
1.87 0.45 4.00 4.00 0.84 4.00 0.10
18.87 8.13 8.42 85 18.3S 8.88 1.84
188 8.48 3.8? 3.SS 0.82 4.80 0.89
38.88 18.04 89.28 08.88 £2.83 188.88 2.34
% inhibition 53.84 §8.38 Ιβ.ίί 8.S2 0..88 3T.S0
5 min 4.00 0.43 4.00 4.00 1.65 4.00 0.03
4.00 0.50 4.00 4.00 2.15 4.00 0.03
4.00 0.50 4.00 4.00 1.80 4.00 0.03
4.00 0.52 4.00 4.00 1.81 4.00 0.10
8.80 8.88 8.88 11.31 0..88 2.?9
4.88 4.88 4.88 188 4.88 0.88
188.80 12.61 188.80 tos.88 48.24 198.80 £.36
S 88 S 88 4? ίΐίΐ S3.78 S 88 97.84
15 min 4.00 0.53 4.00 4.00 2.37 4.00 0.03
4.00 0.61 4.00 4.00 2.38 4.00 0.10
4.00 0.54 4.00 4.00 2.25 4.00 0.03
4.00 0.56 4.00 4.00 2.17 4.00 0.10
K ¥ S 88 S 88 J? ίΐίΐ 4.33 Si 88 1.88
Ave 4.8s) 8.5s§ 4.8s) 4.88 2.29 4.8s) 8.8S
X S¾d 588.88 14.82 188.88 168.08 S7.35 190.80 2.37
8.88 85.08 8.88 8.00 42.85 8.88 S?.SJ4 JJ 3t*
Table 4
Among these five compounds, CDI-034 and CDI-42 were shown to inhibit a-Amylase in 100% saliva samples (See Fig. 15).
3. Lysozyme Inhibition Saliva Screening Method
At least one assay used for the detection of a-amylases activity, as used herein includes the use of a Micrococcus lysodeikticus labeled with fluorescein. The assay measures lysozyme activity on Micrococcus lysodeikticus cell walls, which are labeled to such a degree that the fluorescence is quenched. Lysozyme activity can relieve this quenching; yielding increased fluorescence that is proportional to lysozyme activity.
For the assay, test compounds are mixed with saliva, prior to the addition of lysozyme substrate. The test compounds, such as GRAS materials, are prepared at a concentration of 5,000 ppm in distilled water. Following this preparation, 50μ1 of lysozyme substrate at lmg/ml (Molecular Probes EnzChek Lysozyme Assay Kit) that has been pre-heated to 37°C is added to 50μ1 of the test compound. To initiate the assay, 50μ1 of a dilute (3.5%) or full strength (100%) saliva solution is added to the mixture. Following the initiation, each mixture is measured at 1 minute intervals using a spectrophotometer (such as Molecular Devices M5 reader) for absorption at 494 nm and fluorescence emission at 518 nm. Temperatures of the mixtures are kept constant at 37°C during the assay.
4. Lysozyme Inhibitor Assays.
A series of GRAS compounds were tested using the Lysozyme assay protocol described above. The tested compounds are included in Table I. From the screen of GRAS compounds, four compounds including Fixanal® Buffer 6.0 (Sigma-Aldrich Co., CDI-030), bentonite (CDI-37), benzoic acid (CDI-40), and acetic acid (CDI-047) were shown to have Lysozyme Inhibitor Activity in dilute (3.5%) saliva (See Fig. 16). Further details of these five compounds may also be seen in Table 5.
Figure imgf000053_0001
Table 5
Among these four compounds, benzoic acid (CDI-40), and acetic acid (CDI-047) were shown to inhibit Lysozyme in 100% saliva samples (data not shown).
5. Galactose Oxidase Inhibition Saliva Screening Method
At least one assay used for the detection of galactose oxidase activity, as used herein, includes the use of a chromagenic or fluorogenic substrate, such as with Amplex® Red
(Molecular Probes). In an embodiment of the assay used herein, galactose oxidase catalyzes the oxidation of galactose at the C6 position and generates hydrogen peroxide (H202). The
H202 then, in the presence of horseradish peroxidase (HRP), reacts with 1 : 1 stoichiometry with Amplex® Red reagent to generate the red-fluorescent oxidation product, resorufin. Resorufin has absorption and fluorescence emission maxima of approximately 571 nm and 585 nm, respectively, and because the extinction coefficient is high (54,000 cm-lM-1), the assay can be performed either fluorometrically or spectrophotometrically.
For the assay, test compounds are mixed with Amplex Red, reaction buffer, horseradish peroxidase (HRP) at lOOU/ml, and galactose stock solution (according to manufacturers recommended protocol, Molecular Probes A22179), prior to the addition of saliva. The test compounds, such as GRAS materials, are prepared under various concentration for testing. Following this preparation, 50μ1 of the test compound solution (with Amplex Red, reaction buffer, and HRP) is added to 50μ1 of saliva. Following the initiation, each mixture is measured at 1 minute intervals using a spectrophotometer (such as Molecular Devices M5 reader) using excitation in the range of 530-560nm and emission detection at about 590nm or absorbance at 560nm.. Temperatures of the mixtures are kept constant at 37°C during the assay.
6. Galactose Oxidase Inhibitor Assays.
A series of GRAS compounds were tested using the Galactose Oxidase assay protocol described herein. The tested compounds are included in Table I. From the screen of GRAS compounds, four compounds (CDI-039, 040, 042, and 047) were shown to have Galactose Oxidase Inhibitor Activity in dilute (3.5%) saliva {See Fig. 17). Fig. 17 includes the following symbols which are: open circle = Aluminum potassium sulfate dodecahydrate, open square = 3-tert-butyl-hydroxyanisole, open triangle = benzoic acid, open diamond = acetic acid, filled circle=fresh saliva, and filled square = water (control blank).
7. Stabilization of Glycosylated Hemoglobin
At least one procedure used for the stabilization and detection of glycosylated hemoglobin (HbAlc), as described herein, includes the incubation of HbAlc with a stabilizing agent prior to, or concurrently with, detection. Analysis of samples was conducted on a HPLC 1100 series (Agilent Technologies) with a 2.1X150 (3.5μ) CI 8 reverse phase column. The mobile phase used was acetonitrile with 6.5mM Ammonium Carbonate. A HbAlc standard was prepared by dissolving HbAlc (Sigma Aldrich, 405 RM) with 1.0ml of deionized water. The samples analyzed by HPLC included pure HbAlc (Fig. 18), 100ml HbAlc mixed with 100ml of fresh saliva (Fig. 19), and 200ml HbAlc mixed with 100ml of fresh saliva and 100ml of a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1 : 1 : 1 ratio) (Fig. 20). While the pure HbAlc sample was analyzed fresh, samples of HbAlc with saliva were incubated overnight at room temperature prior to analysis. For each sample analyzed, the data was displayed through a three dimensional plot (Figs 18 A, 19A, and 20A), a UV image (Figs. 18B, 19B, and 20B), and a chromatogram (Figs. 18C, 19C, and 20C). Additionally, a comparison of the analysis HbAlc in the presence of saliva with and without stabilizing agent is shown in Figs. 21 (chromatography), 22 (UV image), and 23 (three dimensional plot).
8. HbAlc protection assays
A series of GRAS compounds were tested, as shown in Figs. 18-20, for the ability to protect HbAlc from degradation by components in saliva. The tested compounds are included in Table I. As described above, a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1 :1 : 1 ratio) minimized the degradation of HbAl c, and yielded a defined pattern for HbAl c.
9. Stabilization of Cancer Antigen 19-9
At least one procedure used for the stabilization and detection of Cancer Antigen 19-9 (CA 19-9) as described herein, includes the incubation of CA 19-9 with a stabilizing agent prior to, or concurrently with, detection. Analysis of samples was conducted on a HPLC 1100 series (Agilent Technologies) with a 2.1X150 (3.5μ) CI 8 reverse phase column. The mobile phase used was acetonitrile with 6.5mM Ammonium Carbonate. A CA 19-9 standard was prepared by dissolving CA 19-9 with 1.0ml of deionized water. The samples analyzed by HPLC included pure CA 19-9 (Fig. 24), 100ml CA 19-9 mixed with 100ml of fresh saliva (Fig. 25), and 200ml CA 19-9 mixed with 100ml of fresh saliva and 100ml of a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1 : 1 : 1 ratio) (Fig. 26). While the pure CA 19-9 sample was analyzed fresh, samples of CA 19-9 with saliva were incubated overnight at room temperature prior to analysis. For each sample analyzed, the data was displayed through a three dimensional plot (Figs 24A, 25A, and 26A) and a UV image (Figs. 24B, 25B, and 26B). Additionally, a comparison of the analysis CA 19-9 in the presence of saliva with and without stabilizing agent is shown in Figs. 27 (three dimensional plot) and 28 (UV image).
10. CA 19-9 protection assays
A series of GRAS compounds were tested, as shown in Figs. 24-26, for the ability to protect CA 19-9 from degradation by components in saliva. The tested compounds are included in Table I. As described above, a stabilizing compound comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1 :1 : 1 ratio) minimized the degradation of CA 19-9 in at least one embodiment. 11. Peroxidase Assay
To determine the effect of various GRAS compounds as inhibitors of peroxidase activity, a standard assay using Amplex® Red (Life Technologies), which releases a fluorescent oxidation product. This oxidation product, named resorufin, has an excitation and emission maxima of approximately 571 nm and 585 nm, respectively, and because the extinction coefficient is high (58,000 ± 5,000 cm-lM-1), it allows for the use with fluorometric or spectrophotometric assays.
The test compounds, such as GRAS materials or cocktails of same, were prepared at a concentration of 500ppm in distilled water. For the assay, 50 μΐ samples of test compounds were mixed with 20-30 μΐ of saliva (either "pooled" or "unpooled") prior to the addition of Amplex® Red. Following the mixture of the test compounds with saliva, 50μ1 of Amplex® Red/HRP working solution (See Protocol for Invitrogen Assay #A22188, which is incorporated herein in its entirety), which has a concentration of 100 μΜ of Amplex® Red, is added to the test compound/saliva mixture. Following the initiation of the reaction, each mixture is measured at 1 minute intervals using a spectrophotmoter (such as a Molecular Devices M5 reader) for excitation in the range of 530-560 nm and fluorescence emission detection at approximately 590nl, or for absorption at approximately 560nm. Reactions were allowed to run for 30 minutes during these assays.
12. Peroxidase protection assays
A series of GRAS compounds and cocktails of GRAS compounds were tested, as shown in Figs. 29-33, for the ability to inhibit degradation of a peroxidase-sensitive marker by components in saliva. Various embodiments of the tested compounds are included in Table I. As described above, a stabilizing compound/cocktail comprising aluminum sulfate hydrate, benzoic acid, and aluminum potassium sulfate dodecahydrate (each present in a 1 : 1 : 1 ratio) minimized the degradation of a peroxidase sensitive marker in at least one embodiment (Fig. 29, open circle = blank control, open square = stabilizing cocktail). Further, inhibition assays were also conducted with caffeine (Fig. 30, CDI #33; Key: open circle = pooled saliva, open square=unpooled saliva, open triangle = caffeine, open triangle = caffeine (repeat)), 3-tert-butyl-hydroxyanisole (Fig. 31, CDI #39; Key: open circle = pooled saliva, open square=unpooled saliva, open triangle = 3-tert-butyl-hydroxyanisole, open triangle = 3-tert-butyl-hydroxyanisole (repeat)), benzoic acid (Fig. 32, CDI #16; Key: open circle = pooled saliva, open square=unpooled saliva, open triangle = benzoic acid, open triangle = benzoic acid (repeat)), and aluminum potassium sulfate dodecahydrate (Fig. 33, CDI #42; Key: open circle = pooled saliva, open square = unpooled saliva, open triangle = aluminum potassium sulfate dodecahydrate, open triangle = aluminum potassium sulfate dodecahydrate (repeat)), each of which showed significant peroxidase inhibition capabilities.
13. Diagnostic Devices
In an example of a diagnostic device, as shown in Fig. 34, a test slide with 28 test wells are spotted with 12 to 18 sets of diagnostic markers. These reagents may include diagnostic markers, or controls for the diagnosis. The diagnostic device, in this example, has four diagnostic markers spotted in duplicate, along with two positive and two negative control marker. Each marker is spotted with a competitor, such as anti-haptin, which binds the capture antigen particle to the marker. The device may have four different hatpin/anti- haptin systems to serve as the generic capture system. A unique haptin may be conjugated to a fully paramagnetic particle (PmMp). The PmMp is also coated with a capture monoclonal antibody (MAb) specific for the marker. A second particle (polystyrene microparticle with Europium) is conjugated with the second or detection MAb. In the diagnostic device shown in Fig. 34, the hash marks on the side of the diagnostic device (slide) are indentations molded into the plastic, which may be used as cog grooves. Additionally the slide is designed to fit into a processor that may have more than one processing station, as shown in Fig. 36.
In performing a method of detecting a diagnostic marker using an embodiment of the diagnostic device, such as that in Fig. 34 and 36, the user adds a pre-determined amount of specimen (such as 120μ1) to a specimen diluent (such as 40 to 120 μΐ), and adds the mixture to each test well. Following addition of the diluted specimen, the test slide is incubated at 37°C for about 5 to about 15 minutes with agitation. Following the incubation period, the test wells move over a magnetic source that brings the PmMp to the bottom of the well, so that there is maximal binding of the PmMp with the anti-haptin bound in the test well. Next, specimen and assay reagents are removed, wash buffer is added and aspirated off. Lastly, each well is scanned using a visualization device, such as a CCD camera, and then the fluroescent signal (or other signal) of each marker is determined.
14. Fluid Collection Device.
An example of a collection device for the collection of an bodily fluids is depicted in Fig. 35. In this example, the collection device is capable of collecting at least 500μ1 of bodily fluid, such as saliva. The cap is first removed from the tube. Squeezing the bulb provides suction adequate to pull an amount of saliva into the bulb. In some cases, a filter may be included in the tube to remove unwanted materials (such as particulate and mucous). The filter may be of any known filter material, such as rayon, that is appropriate for the purposes described herein. Following the obtaining of the bodily fluid in the collection device, a cap is placed on the tube, and a second cap is removed from a dispensing tube. The dispensing tube is in fluid communication with the bulb and is sized and shaped to allow for drops of a predetermined size to be formed when the bulb is squeezed. For example, the dispensing tube may allow drops of 30μ1 to be formed when the bulb is squeezed.
While various embodiments of the systems, methods, and compositions of the present disclosure have been described in considerable detail herein, the embodiments are merely offered by way of non-limiting examples of the disclosure described herein. It will therefore be understood that various changes and modifications may be made, and equivalents may be substituted for elements thereof, without departing from the scope of the disclosure. Indeed, this disclosure is not intended to be exhaustive or to limit the scope of the 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 stabilizing system, comprising:
a stabilizing agent useful to completely or substantially prevent degradation or inactivation of a diagnostic marker present within a mammalian body, the diagnostic marker indicative of a mammalian condition; and
a detection agent capable of detecting the diagnostic marker.
2. The stabilizing system of claim 1, further comprising:
a detection platform comprising a plurality of detection sites each capable of receiving the diagnostic marker, the stabilizing agent, and the detection agent;
a computer database capable of receiving a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of the diagnostic marker to the detection agent;
a processor operably coupled to the computer database and the detection platform, the processor having and executing a software program operational to:
determine a binding characteristic between the detection agent and the stabilized diagnostic agent in each of the plurality of detection sites;
compare the binding characteristic among each of the plurality of detection sites, wherein the comparison of binding characteristics is capable of determining the stabilizing agent with the greatest effect on the binding characteristic between the detection agent and the diagnostic agent;
generate a binding record using the compared binding characteristics; and
deliver the binding record to a recipient.
3. The system of claim 2, wherein the software program is further operational to compare the binding characteristic with at least one of a plurality of stored binding characteristics in the computer database.
4. The stabilizing system of claim 1, further comprising:
a detection platform comprising:
a plurality of detection sites each capable of receiving the diagnostic marker, the stabilizing agent, and the detection agent;
a detection device capable of determining a binding characteristic between the detection agent and the diagnostic marker in the detection sites; a computer database capable of receiving a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of the diagnostic marker to the detection agent;
a processor operably coupled to the computer database and the detection device, the processor having and executing a software program operational to:
determine the binding characteristic of the detection agent to the diagnostic marker in each of the plurality of detection sites;
compare the binding characteristic among each of the plurality of detection sites to order the level of effect of the stabilizing agents assayed on the binding characteristic between the detection agent and the diagnostic agent;
generate a binding report using the compared binding characteristics; and
deliver the binding report to a recipient.
5. The stabilizing system of claim 1, further comprising:
a detection platform comprising a plurality of detection sites each capable of receiving at the stabilizing agent, the detection agent, and a body fluid from a patient, the body fluid comprising the diagnostic marker;
a computer database capable of storing a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of the diagnostic marker to the detection agent; and
a processor operably coupled to the computer database and the detection platform, the processor having and executing a software program operational to:
determine a binding characteristic of the detection agent and a stabilized diagnostic marker in each of the plurality of detection sites;
compare the binding characteristic among each of the plurality of detection sites, wherein the comparison of binding characteristics is capable of determining the stabilizing agent with the greatest effect on the binding characteristic between the detection agent and the diagnostic marker;
generate a binding record using the compared binding characteristics; and
deliver the binding record to a recipient.
6. The stabilizing system of claim 5, wherein the binding record may be used by to assist in determining a therapeutic course for the patient.
7. The stabilizing system of claim 1, further comprising: a fluid collector to collect a body fluid; and
a housing, to test and retain the body fluid, the housing comprising:
a collection chamber, having an open end, to receive the fluid collector and contain the stabilization agent;
at least one membrane test strip, in fluid communication with the collection chamber, to indicate the presence or absence of the diagnostic marker; and
an immunoassay-based fingerprint acquisition pad in fluid
communication with the collection chamber, the acquisition pad having at least one control zone including a control reagent to identify a donor of the body fluid, the reagent including a member of a predetermined ligand/receptor binding pair; and
a plurality of reaction zones, each of which includes the detection agent to determine the presence of the diagnostic marker in the fluid sample, the detection agent including a member of a predetermined ligand/receptor binding pair.
8. The stabilizing system of claim 1, further comprising:
a detection platform comprising a plurality of detection sites each capable of receiving the diagnostic marker, the stabilizing agent, and the detection agent;
a computer database capable of receiving a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of the diagnostic marker to the detection agent;
a processor operably coupled to the computer database and the detection platform, the processor having and executing a software program operational to:
determine a binding characteristic of the detection agent and a stabilized detection agent in each of the plurality of detection sites;
compare the binding characteristic among each of the plurality of detection sites, wherein the comparison of binding characteristics is capable of determining the stabilizing agent with the greatest effect on the binding characteristic between the detection agent and the diagnostic agent;
generate a binding record using the compared binding characteristics; and
deliver the binding record to a recipient.
9. The stabilizing system of claim 1, further comprising:
a detection platform comprising a plurality of detection wells housed in a solid matrix; the detection wells containing a plurality of the diagnostic markers and at least one competitor molecule, wherein the plurality of the diagnostic markers and at least one control molecule are each bound to a paramagnetic particle;
a detection system capable of receiving the detection platform, exerting a magnetic field upon a surface of the detection platform, and determining at least one characteristic of the diagnostic marker.
10. The system of claim 9, wherein the solid matrix is comprised of material selected for the group consisting of quartz, glass, metal, silica-based materials, resins, polymers, or combinations thereof.
11. The system of claim 9, wherein the magnetic field is able to force the paramagnetic particle towards the surface of the detection platform.
12. The system of claim 9, wherein the system further comprises a second paramagnetic particle linked to a detection antibody.
13. The system of claim 9, wherein the second paramagnetic particle is comprised of Europium.
14. The system of claim 9, wherein the second paramagnetic particle is coupled to a second detection agent.
15. The stabilizing system of claim 1, further comprising:
a diagnostic device comprising a plurality of test wells, wherein each test well is capable of containing the detection agent; and
a detection device in communication with the diagnostic device, wherein the detection device is capable of detecting an interaction between the at least one detection agent and the diagnostic marker in at least one of the plurality of test wells;
wherein the stabilizing agent is contained within at least one of the plurality of test wells.
16. The stabilizing system of claim 15, wherein the diagnostic device is comprised of a material selected for the group consisting of quartz, glass, metal, silica-based materials, resins, polymers, or combinations thereof.
17. The stabilizing system of claim 15, wherein the detection device is capable of detecting a colorimetric or fluorometric signal from at least one of the plurality of test wells.
18. The stabilizing system of claim 1, further comprising:
a microarray product comprising at least 100 diagnostic markers/cm , a microarray identifier, and the stabilizing agent; a control microarray product comprising a first specific binding pair member that binds to a first detectable label; and
a processor for providing information regarding the identification and concentration of markers on the microarray based on the identity of the array provided by the microarray identifier.
19. The stabilizing system of claim 18, wherein the microarray product is comprised of a compound selected from the group consisting of gel, nitrocellulose, nylon, quartz, glass, metal, silica based materials, silica, resins, polymers, or combinations thereof.
20. The stabilizing system of claim 18, wherein the diagnostic marker comprises a DNA fragment having a length of about 10 base pairs to about 50 base pairs of DNA.
21. The stabilizing system of claim 18, wherein the diagnostic marker comprises an R A fragment having a length of about 10 base pairs to about 50 base pairs of R A.
22. The stabilizing system of claim 18, wherein the diagnostic marker comprises a peptide fragment having a length of at least about 50 amino acids.
23. The stabilizing system of claim 1, further comprising:
a latex particle having a carboxylate group, the latex particle functionally coupled to the stabilizing agent.
24. The stabilizing system of claim 23, further comprising at least one of human serum albumin (HSA) and bovine serum albumin (BSA).
25. The stabilizing system of claim 23, further comprising a linker group coupled to latex particle.
26. The stabilizing system of claim 25, wherein the linker group comprises a maleimide compound.
27. The stabilizing system of claim 26, wherein the maleimide compound is selected from the group consisting of Succinimidyl 4-[N-Maleimidomethyl] Cyclohexane-1- Carboxylate (SMCC) and N-Hydroxysuccinimide-activated hexa(ethylene glycol) undecane Thiol (NHS).
28. The stabilizing system of claim 26, wherein the linker group is coupled to the HSA or BSA.
29. The stabilizing system of claim 23, further comprising an antibody functionally linked to the latex particle.
30. The stabilizing system of claim 23, wherein the latex particle has a diameter of about 0.02μιη to about 7.0μιη.
31. The stabilizing system of claim 1, wherein the stabilizing agent is selected from the group consisting of a protease inhibitor, a DNase inhibitor, and a RNase inhibitor.
32. The stabilizing system of claim 1, wherein the diagnostic marker is selected from the group consisting of a protein, a glycoprotein, a nucleic acid, an enzyme, an enzyme inhibitor, and a metabolite.
33. The stabilizing system of claim 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.
34. The stabilizing system of claim 1, wherein the condition is selected from a group consisting of insulin resistance, glucose intolerance, cancer, cardiovascular disease, a bacterial infection, a viral infection, a fungal infection, a parasite infection, a food allergy, a non-food allergy, drug use, fertility, pregnancy, circulating calcium,
35. The stabilizing system of claim 1, wherein the degradation or inactivation of the diagnostic marker for a condition occurs by a destructive agent.
36. The stabilizing system of claim 35, wherein the destructive agent is galactose oxidase.
37. The stabilizing system of claim 1, wherein the body fluid is selected from the group consisting of saliva, a mucous secretion, tears, sweat, semen, urine, a vaginal secretion, exhalate, blood, serum, and an anal secretion.
38. The stabilizing system of claim 1, wherein the stabilizing agent is selected from the group consisting of Fixanal® Buffer 6.0 (Sigma-Aldrich Co.), acetic acid, aluminum hydroxide bentonite, aluminum sulfate hydrate, aluminum potassium sulfate dodecahydrate, benzoic acid, caffeine, and 3-tert-butyl-hydroxyanisole, or a combination thereof.
39. The stabilizing system of claim 1, wherein the stabilizing agent comprises a plurality of stabilizing agents each present in approximately the same concentration.
40. The stabilizing system of claim 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 the 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%, and at least about 99%.
41. The stabilizing system of claim 1, wherein the stabilizing agent has a concentration selected from the group consisting of about 200 parts per million (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.
42. The stabilizing system of claim 1, wherein the stabilizing agent is able to inhibit the degradation or inactivation of the diagnostic marker for an inhibitory period selected from the group consisting of at least one minute, at least about five minutes, at least about ten minutes, at least about fifteen minutes, at least about thirty minutes, at least about one hour, at least about two hours, at least about four hours, and at least about eight hours.
43. The stabilizing system of claim 1, wherein the diagnostic marker is selected from the group consisting a-hydroxybutyrate, 1-linoleoyl-GPC, palmitate, Glycine, and 3- methyl-2-oxybutyrate, glycosylated hemoglobin (HbAlc), IgE, and IgG.
44. The stabilizing system of claim 1, wherein the diagnostic marker is selected from the group consisting of Aldose reductase, Angiogenin, Annexin Al, B-cell activating factor (BAFF), B-cell lymphoma 2 (BCL2)-like 2, Beta Human chorionic gonadotropin, Cal5-3, Calcyclin, Calvasculin, Cancer Antigen CA 19-9, Cancer Antigen CA 15-3, Cathepsin D, Caveolin-1, Chromogranin A, Alpha-crystallin B chain (CRYAB), Endostatin, Eotaxin-2, Epithelial cell adhesion molecule (EpCAM), Ezrin, fatty acid binding protein 4 (FABP4), Galectin-3, γ-glutamylcysteine ligase regulatory chain (GCLR), Gelsolin, Glucose 6-phosphate (G6P), Glycoprotein 130 (gpl30), Glutathione S-transferase Mu 1 (GSTM1), Hepsin, High-mobility group protein Bl (HMGB-1), Insulin- like growth factor binding protein 1 (IGFBP-1), Insulin-like growth factor binding protein 4 (IGFBP-4), Insulin-like growth factor binding protein 5 (IGFBP-5), Insulin-like growth factor binding protein 6 (IGFBP-6), LGL, latency associated peptide (LAP), macrophage stimulating protein (MSP), MHC class I polypeptide-related sequence A (MICA), Nucleoside diphosphate kinase B (NME2), Neuron-specific Enolase (NSE), Osteopontin, Osteoprotegerin, Pepsinogen, Peroxiredoxin, Phosphoserine aminotransferase (PSAT1), Prostate Specific Antigen, Receptor tyrosine-protein kinase erbB-3 (ErbB3), Serpin B3, Vascular smooth muscle cell growth factor R2 (VSGF R2/KDR), Vascular endothelial growth factor R3 (VEGF R3/Flt-4), Thyro globulin, Tyrosine kinase with immunoglobulin-like and EGF-like domains 2 (TIE-2), Tissue plasminogen activator (tPA), Transforming growth factor beta (TGF-βΙ), Tumor necrosis factor receptor 1 (TNF-R1), urokinase-type Plasminogen Activator (uPA), urokinase-type Plasminogen Activator Receptor (uPAR), Brcal, Brcall, kallikreins, e- cadherin, Hox peptide, and Engrailed-2.
45. The stabilizing system of claim 1, wherein the diagnostic marker is selected from the group consisting of creatine kinase, troponin I and T, LD, Myoglobin, Alanine aminotransferase (ALT), Aspartate transaminase(AST), ALT/AST ratio, Heart-type fatty acid binding protein (H-FABP), and Glycogen phosphorylase B.
46. The stabilizing system of claim 1, wherein the diagnostic marker is selected from the group consisting of a bacterial protein, a bacterial outer membrane protein, a bacterial secreted protein, a bacteria-specific DNA sequence, a bacteria-specific RNA sequence, a bacterial metabolite, a host-reaction product to a bacteria, a viral protein, a viral outer membrane protein, a viral secreted protein, a viral-specific DNA sequence, a viral- specific RNA sequence, a viral metabolite, and a host-reaction product to a virus, a fungal protein, a fungal outer membrane protein, a fungal secreted protein, a fungal-specific DNA sequence, a fungal-specific RNA sequence, a fungal metabolite, and a host-reaction product to a fungus, a parasite protein, a parasite outer membrane protein, a parasite secreted protein, a parasite-specific DNA sequence, a parasite-specific RNA sequence, a parasite metabolite, and a host-reaction product to a parasite.
47. The stabilizing system of claim 1, wherein the diagnostic marker is indicative of a drug selected from the group consisting alcohol, cocaine, marijuana, opiates, amphetamine, methamphetamine, amphetamines, phencyclidine, benzodiazepines, barbiturates, methadone, tricyclic antidepressants, heroin, steroids, niacin, xanan, vicodin, oxycontin, adderall, morphine, and nicotine.
48. The stabilizing system of claim 1, wherein the diagnostic marker is selected from the group consisting of lutenizing hormone, progesterone, human chorionic gonadotropin (hCG), early pregnancy factor (EPF), calcium, vitamin D, phosphorus, and magnesium.
49. A method of stabilizing the diagnostic marker, the method comprising the step of:
mixing a mammalian body fluid comprising a diagnostic marker indicative of a condition with an effective amount of a stabilizing agent; and
detecting the stabilized marker with a detection agent capable of detecting the diagnostic marker;
wherein the stabilizing agent completely or substantially prevents the cleavage, degradation, or inactivation of the diagnostic marker.
50. The method of claim 49 wherein the step of mixing a body fluid with the stabilizing agent comprises introducing the stabilizing agent into a mouth/oral cavity of a patient so that the stabilizing agent mixes with saliva.
51. The method of claim 49, 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.
52. The method of claim 49, wherein the body fluid is selected from the group consisting of saliva, a mucous secretion, tears, sweat, semen, urine, a vaginal secretion, exhalate, blood, serum, and an anal secretion.
53. The method of claim 49, wherein the stabilizing agent is selected from the group consisting of Fixanal® Buffer 6.0 (Sigma-Aldrich Co.), acetic acid, aluminum hydroxide bentonite, aluminum sulfate hydrate, aluminum potassium sulfate dodecahydrate, benzoic acid, caffeine, and 3-tert-butyl-hydroxyanisole, or a combination thereof.
54. The method of claim 49, wherein the stabilizing agent is capable of inhibiting degradation of the diagnostic marker to an inhibitory degree, wherein the inhibitory degree is selected from the 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%, and at least about 99%.
55. The method of claim 49, wherein the stabilizing agent has a concentration selected from the group consisting of about 200 parts per million (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.
56. The method of claim 49, wherein the diagnostic marker is selected from the group consisting of a-hydroxybutyrate, 1-linoleoyl-GPC, palmitate, Glycine, and 3-methyl-2- oxybutyrate.
57. The method of claim 49, wherein the diagnostic marker is selected from the group consisting a-hydroxybutyrate, 1-linoleoyl-GPC, palmitate, Glycine, and 3-methyl-2- oxybutyrate, glycosylated hemoglobin (Fib Ale), IgE, and IgG.
58. The method of claim 49, wherein the diagnostic marker is selected from the group consisting of Aldose reductase, Angiogenin, Annexin Al, B-cell activating factor (BAFF), B-cell lymphoma 2 (BCL2)-like 2, Beta Human chorionic gonadotropin, Cal5-3, Calcyclin, Calvasculin, Cancer Antigen CA 19-9, Cancer Antigen CA 15-3, Cathepsin D, Caveolin-1, Chromogranin A, Alpha-crystallin B chain (CRYAB), Endostatin, Eotaxin-2, Epithelial cell adhesion molecule (EpCAM), Ezrin, fatty acid binding protein 4 (FABP4), Galectin-3, γ-glutamylcysteine ligase regulatory chain (GCLR), Gelsolin, Glucose 6- phosphate (G6P), Glycoprotein 130 (gpl30), Glutathione S-transferase Mu 1 (GSTM1), Hepsin, High-mobility group protein Bl (HMGB-1), Insulin- like growth factor binding protein 1 (IGFBP-1), Insulin-like growth factor binding protein 4 (IGFBP-4), Insulin-like growth factor binding protein 5 (IGFBP-5), Insulin-like growth factor binding protein 6 (IGFBP-6), LGL, latency associated peptide (LAP), macrophage stimulating protein (MSP), MHC class I polypeptide-related sequence A (MICA), Nucleoside diphosphate kinase B (NME2), Neuron-specific Enolase (NSE), Osteopontin, Osteoprotegerin, Pepsinogen, Peroxiredoxin, Phosphoserine aminotransferase (PSAT1), Prostate Specific Antigen, Receptor tyrosine-protein kinase erbB-3 (ErbB3), Serpin B3, Vascular smooth muscle cell growth factor R2 (VSGF R2/KDR), Vascular endothelial growth factor R3 (VEGF R3/Flt-4), Thyro globulin, Tyrosine kinase with immunoglobulin-like and EGF-like domains 2 (TIE -2), Tissue plasminogen activator (tPA), Transforming growth factor beta (TGF-βΙ), Tumor necrosis factor receptor 1 (TNF-R1), urokinase-type Plasminogen Activator (uPA), urokinase-type Plasminogen Activator Receptor (uPAR), Brcal, Brcall, kallikreins, e- cadherin, Hox peptide, and Engrailed-2.
59. The method of claim 49, wherein the diagnostic marker is selected from the group consisting of creatine kinase, troponin I and T, LD, Myoglobin, Alanine aminotransferase (ALT), Aspartate transaminase(AST), ALT/AST ratio, Heart-type fatty acid binding protein (H-FABP), and Glycogen phosphorylase B.
60. The method of claim 49, wherein the diagnostic marker is selected from the group consisting of a bacterial protein, a bacterial outer membrane protein, a bacterial secreted protein, a bacteria-specific DNA sequence, a bacteria-specific RNA sequence, a bacterial metabolite, a host-reaction product to a bacteria, a viral protein, a viral outer membrane protein, a viral secreted protein, a viral-specific DNA sequence, a viral-specific RNA sequence, a viral metabolite, and a host-reaction product to a virus, a fungal protein, a fungal outer membrane protein, a fungal secreted protein, a fungal-specific DNA sequence, a fungal-specific RNA sequence, a fungal metabolite, and a host-reaction product to a fungus, a parasite protein, a parasite outer membrane protein, a parasite secreted protein, a parasite- specific DNA sequence, a parasite-specific RNA sequence, a parasite metabolite, and a host- reaction product to a parasite.
61. The method of claim 49, wherein the diagnostic marker is indicative of a drug selected from the group consisting alcohol, cocaine, marijuana, opiates, amphetamine, methamphetamine, amphetamines, phencyclidine, benzodiazepines, barbiturates, methadone, tricyclic antidepressants, heroin, steroids, niacin, xanan, vicodin, oxycontin, adderall, morphine, and nicotine.
62. The method of claim 49, wherein the diagnostic marker is selected from the group consisting of lutenizing hormone, progesterone, human chorionic gonadotropin (hCG), early pregnancy factor (EPF), calcium, vitamin D, phosphorus, and magnesium.
63. The method of claim 49, 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.
64. The method of claim 49, wherein the condition is selected from a group consisting of insulin resistance, glucose intolerance, cancer, cardiovascular disease, a bacterial infection, a viral infection, a fungal infection, a parasite infection, a food allergy, a non-food allergy, drug use, fertility, pregnancy, and a level of circulating calcium.
65. The method of claim 49, wherein the degradation or inactivation of the diagnostic marker for a condition occurs by a destructive agent.
66. The method of claim 65, wherein the destructive agent is selected from a group consisting of galactose oxidase.
67. A computer-implemented method of improving diagnostic marker availability, the method comprising the steps of:
introducing a predetermined diagnostic marker for cancer into a plurality of detection sites of a detection platform;
introducing a stabilization agent into each of the plurality of detection sites containing the predetermined diagnostic marker for cancer, wherein the stabilization agent in each of the plurality of detection sites is capable of completely or substantially preventing the degradation or inactivation of the diagnostic marker;
introducing a detection agent into each of the plurality of detection sites having a stabilized diagnostic agent;
determining a binding characteristic of the detection agent and the stabilized diagnostic agent in each of the plurality of detection sites with a processor; and
computationally comparing the binding characteristic among each of the plurality of detection sites with the processor, wherein the comparison of binding characteristics is capable of determining the stabilizing agent with the greater effect on the binding characteristic between the detection agent and the diagnostic agent.
68. The method of claim 67, wherein the detection platform is selected from the group consisting of a microtitre plate, a microarray, and a multi-well plate.
69. The method of claim 67, further comprising the step of computationally comparing the binding characteristics to at least one stored binding characteristic contained in the computer database in communication with the processor.
70. A method of identifying diagnostic markers of cancer, the method comprising the steps of:
contacting a body fluid of a diseased subject having a cancer with a stabilizing agent capable of completely or substantially preventing the degradation or inactivation of a diagnostic marker indicative of the cancer, the body fluid comprising the diagnostic agent;
analyzing the stabilized body fluid to determine at last one property of the diagnostic marker to create a cancer profile;
comparing the cancer profile with a control profile of a healthy subject not having the cancer, wherein the control profile is created in a like manner to the cancer profile; and
generating a cancer identifier panel from the compared cancer profile, wherein the cancer identifier panel contains at least one cancer diagnostic marker indicative of the cancer.
71. The method of claim 70, wherein the stabilizing agent prevents the degradation or inactivation of the diagnostic marker for at least a twenty four hours period following interaction with the diagnostic agent.
72. A method for analyzing stabilization agents, the method comprising the steps of:
introducing a diagnostic marker into each of a plurality of detection sites of a detection platform, the detection platform comprising:
the plurality of detection sites each capable of receiving a diagnostic marker, a stabilization agent, and a detection agent;
a detection device capable of determining a binding characteristic between the detection agent and the diagnostic marker in the detection sites;
a computer database capable of receiving a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of a diagnostic marker to a detection agent;
a processor operably coupled to the computer database and the detection device, the processor having and executing a software program operational to:
determine the binding characteristic of the detection agent to the diagnostic marker in each of the plurality of detection sites; compare the binding characteristic among each of the plurality of detection sites to order the level of effect of the stabilizing agents assayed on the binding characteristic between the detection agent and the diagnostic agent;
generate a binding record using the compared binding characteristics; and
deliver the binding record to a recipient;
introducing the stabilization agent to each of the plurality of detection sites containing the diagnostic marker;
combining the detection agent with the stabilization agent and diagnostic marker in the detection sites;
comparing the binding characteristic with the processor among each of the plurality of detection sites to order the level of effect of the stabilizing agents assayed on the binding characteristic between the detection agent and the diagnostic agent;
generating a binding report using the compared binding characteristics; and delivering the binding report to a recipient.
73. A method of determining the efficacy of a therapeutic compound, the method comprising the steps of:
treating a subject having a disease state with an effective amount of therapeutic compound;
harvesting at least one cell from the subject following a treatment with the therapeutic compound;
bringing the at least one cell into contact with a stabilizing compound capable of completely or substantially preventing the degradation or inactivation of a diagnostic marker for the disease state;
analyzing the at least one stabilized cell for the at least one diagnostic marker; creating a marker profile from at least one result determined from analyzing the at least one stabilized cell; and
comparing the marker profile with at least one previous marker profile to determine efficacy of the therapeutic compound on the disease state.
74. The method of claim 73, wherein the at least one cell is a tumor cell.
75. The method of claim 73, wherein the step of harvesting the at least one cell comprises the step of collecting a body fluid from the subject, wherein the body fluid comprises the at least one cell.
76. A computer-implemented method of determining the therapeutic potential of a therapeutic agent, comprising the steps of:
harvesting a body fluid from a patient having a disease state;
introducing the body fluid to a detection platform comprising:
a plurality of detection sites each capable of receiving a stabilization agent, a detection agent, and a body fluid from a patient comprising a diagnostic marker;
a computer database capable of storing a plurality of binding characteristics, the plurality of binding characteristics comprising at least one binding property of a diagnostic marker to a detection agent;
a processor operably coupled to the computer database and the detection platform, the processor having and executing a software program operational to:
determine a binding characteristic of the detection agent and a stabilized diagnostic marker in each of the plurality of detection sites;
compare the binding characteristic among each of the plurality of detection sites, wherein the comparison of binding characteristics is capable of
determining the stabilizing agent with the greatest effect on the binding characteristic between the detection agent and the diagnostic marker;
generate a binding record using the compared binding characteristics; and
deliver the binding record to a recipient;
contacting the body fluid with a stabilizing agent capable of completely or substantially preventing the degradation or inactivation of a diagnostic marker for the disease state;
analyzing the at least one stabilized body fluid for the at least one diagnostic marker to create a marker profile;
comparing the marker profile with a marker profile library stored in the computer database to determine a therapeutic potential of the therapeutic compound on the disease state;
generating a binding record using the compared binding characteristics; and delivering the binding record to a recipient.
77. A method of analyzing biomarker levels, the method comprising the steps of: combining a body fluid comprising a diagnostic marker with a stabilization agent for an incubation period, the stabilization agent capable of completely or substantially preventing the degradation or inactivation of the diagnostic marker; introducing the stabilized body fluid into a detection platform; combining a detection agent with the stabilized body fluid;
determining an interaction characteristic of the detection agent and the diagnostic marker of the stabilized body fluid using a detection device in communication with a processor;
comparing the interaction characteristic with a plurality of indexed interaction records contained on a database in communication with the processor to determine an identity and concentration of the detection marker;
generating a report of the identity and concentration of the detection marker in the body fluid; and
delivering the report to a recipient.
78. The method of claim 77, wherein the detection platform is selected from the group consisting of a microarray, a multi-well plate, a high-performance liquid chromato graph, and a mass spectrometer.
79. A micro fluidic device for analyzing the presence or absence of the diagnostic marker to be detected in a body fluid, the micro fluidic device comprising:
a container, the container housing:
a sample reservoir having a sample input port in fluid connection with at least one detector array, wherein the detector array comprises at least one detector comprising a reaction chamber comprising an immobilized capture molecule;
the stabilizing agent contained within the sample reservoir, the stabilizing agent useful to completely or substantially prevent degradation or inactivation of the diagnostic marker; and
a reagent contained within the detector array, the reagent capable of undergoing a colorimetric reaction or displaying an optically detectable signal and capable of reacting with the diagnostic marker.
80. The microfluidic device of claim 79, further comprising filter capable of separating at least one unwanted component from a body fluid.
81. The microfluidic device of claim 79, further comprising a results display, wherein a result from the detector array is visually detectable.
82. The microfluidic device of claim 79, wherein the results display comprises a plurality of displays capable to visualizing a plurality of detected diagnostic markers.
83. The microfluidic device of claim 79, further comprising a control display, wherein the detection of at least one control reagent by the at least one detector is visualized.
84. A system for stabilizing the diagnostic marker comprising:
a sample container having an opening and least one rigid chamber for containing a body fluid, the body fluid comprising at least one diagnostic marker;
a stabilizing agent contained within the sample container, the stabilizing agent capable of completely or substantially preventing the degradation or inactivation of the diagnostic marker; and
a securing device for closing the opening of the sample container;
wherein the stabilizing agent is coupled to a solid support.
85. The system of claim 84, wherein the stabilizing agent is adsorbed to the solid support.
PCT/US2011/031935 2010-04-09 2011-04-11 Devices, systems, and methods for biomarker stabilization WO2011127467A1 (en)

Applications Claiming Priority (98)

Application Number Priority Date Filing Date Title
US32276810P 2010-04-09 2010-04-09
US61/322,768 2010-04-09
US36497810P 2010-07-16 2010-07-16
US36496410P 2010-07-16 2010-07-16
US36496910P 2010-07-16 2010-07-16
US36498210P 2010-07-16 2010-07-16
US36517910P 2010-07-16 2010-07-16
US36497510P 2010-07-16 2010-07-16
US61/365,179 2010-07-16
US61/364,975 2010-07-16
US61/364,964 2010-07-16
US61/364,982 2010-07-16
US61/364,978 2010-07-16
US61/364,969 2010-07-16
US37361910P 2010-08-13 2010-08-13
US61/373,619 2010-08-13
US37896010P 2010-09-01 2010-09-01
US61/378,960 2010-09-01
US37959810P 2010-09-02 2010-09-02
US61/379,598 2010-09-02
US38340110P 2010-09-16 2010-09-16
US61/383,401 2010-09-16
US38534710P 2010-09-22 2010-09-22
US61/385,347 2010-09-22
US39000210P 2010-10-05 2010-10-05
US61/390,002 2010-10-05
US12/906,170 2010-10-18
US12/906,221 2010-10-18
US12/906,239 US20110195423A1 (en) 2010-02-10 2010-10-18 Systems and methods for diagnosing bacterial infections
US12/906,170 US20110195872A1 (en) 2010-02-10 2010-10-18 Devices and systems for biomarker detection
US12/906,236 US20110195440A1 (en) 2010-02-10 2010-10-18 Systems and methods for identifying allergies
US12/906,204 2010-10-18
US12/906,169 2010-10-18
US12/906,168 US20110195491A1 (en) 2010-02-10 2010-10-18 Devices and systems for biomarker detection
US12/906,167 US20110195487A1 (en) 2010-02-10 2010-10-18 Sample container for biomarker maintenance
US12/906,224 2010-10-18
US12/906,221 US20110195858A1 (en) 2010-02-10 2010-10-18 Systems and methods of analyzing biomarker levels
US12/906,225 2010-10-18
US12/906,236 2010-10-18
US12/906,245 US20110195394A1 (en) 2010-02-10 2010-10-18 Systems and methods for diagnosing viral infections
US12/906,162 2010-10-18
US12/906,162 US8574856B2 (en) 2010-02-10 2010-10-18 Methods for determining the efficiency of a therapeutic
US12/906,167 2010-10-18
US12/906,224 US20110195400A1 (en) 2010-02-10 2010-10-18 Systems and methods for diagnosing cancer
US12/906,273 2010-10-18
US12/906,171 US20110195488A1 (en) 2010-02-10 2010-10-18 Devices for collection and stabilization of biomarkers in liquid samples
US12/906,263 US20110195403A1 (en) 2010-02-10 2010-10-18 Systems and methods for fertility analysis
US12/906,211 US20110195857A1 (en) 2010-02-10 2010-10-18 Systems for analyzing stabilized biomarkers
US12/906,201 US20110195397A1 (en) 2010-02-10 2010-10-18 Drink mixture for the stabilization of diagnostic biomarkers
US12/906,225 US20110195401A1 (en) 2010-02-10 2010-10-18 Systems and methods for determining food allergies
US12/906,161 US20110195856A1 (en) 2010-02-10 2010-10-18 Methods of determining stabilization compounds for predictive biomarkers
US12/906,268 US20110195427A1 (en) 2010-02-10 2010-10-18 Systems and methods for detecting pregnancy
US12/906,164 US20110195851A1 (en) 2010-02-10 2010-10-18 Systems and methods for determining therapeutic potential
US12/906,255 2010-10-18
US12/906,202 US20110195398A1 (en) 2010-02-10 2010-10-18 Systems and methods for detecting insulin resistance biomarkers
US12/906,208 2010-10-18
US12/906,166 2010-10-18
US12/906,263 2010-10-18
US12/906,158 2010-10-18
US12/906,199 2010-10-18
US12/906,201 2010-10-18
US12/906,168 2010-10-18
US12/906,169 US20110194996A1 (en) 2010-02-10 2010-10-18 Transport container for stabilized liquid samples
US12/906,173 2010-10-18
US12/906,260 2010-10-18
US12/906,196 2010-10-18
US12/906,273 US20110195404A1 (en) 2010-02-10 2010-10-18 Systems and methods for determining calcium levels
US12/906,161 2010-10-18
US12/906,223 2010-10-18
US12/906,196 US20110195395A1 (en) 2010-02-10 2010-10-18 Rinse composition for the stabilization of diagnostic biomarkers
US12/906,166 US20110196085A1 (en) 2010-02-10 2010-10-18 Biomarker stabilizing agents
US12/906,239 2010-10-18
US12/906,211 2010-10-18
US12/906,223 US8293537B2 (en) 2010-02-10 2010-10-18 Systems and methods for hemoglobin analysis
US12/906,231 US20110195439A1 (en) 2010-02-10 2010-10-18 Systems and methods for determining cardiac conditions
US12/906,159 US8501669B2 (en) 2010-02-10 2010-10-18 Systems and methods of screening biomarkers in bodily fluids
US12/906,253 US20110195424A1 (en) 2010-02-10 2010-10-18 Systems and methods for diagnosing fungal infections
US12/906,260 US20110195425A1 (en) 2010-02-10 2010-10-18 Systems and methods for diagnosing parasite infections
US12/906,235 2010-10-18
US12/906,159 2010-10-18
US12/906,268 2010-10-18
US12/906,202 2010-10-18
US12/906,255 US20110195402A1 (en) 2010-02-10 2010-10-18 Systems and methods for detecting drug use
US12/906,204 US8318105B2 (en) 2010-02-10 2010-10-18 Systems and methods for biomarker analysis
US12/906,171 2010-10-18
US12/906,231 2010-10-18
US12/906,235 US8318641B2 (en) 2010-02-10 2010-10-18 Systems and methods for the detection of biomarkers
US12/906,227 2010-10-18
US12/906,253 2010-10-18
US12/906,173 US20110192239A1 (en) 2010-02-10 2010-10-18 Devices for collection and stabilization of biomarkers in liquid samples
US12/906,227 US8426151B2 (en) 2010-02-10 2010-10-18 Systems and methods for the detection of biomarkers
US12/906,245 2010-10-18
US12/906,158 US20110195421A1 (en) 2010-02-10 2010-10-18 Methods of identifying biomarkers for cancer
US12/906,208 US20110195399A1 (en) 2010-02-10 2010-10-18 Systems and methods for the inhibition of galactose oxidase
US12/906,199 US20110195396A1 (en) 2010-02-10 2010-10-18 Chewable compositions for the stabilization of diagnostic biomarkers
US12/906,157 US20110195855A1 (en) 2010-02-10 2010-10-18 Systems and methods for improving biomarker availability
US12/906,157 2010-10-18
US12/906,164 2010-10-18

Publications (1)

Publication Number Publication Date
WO2011127467A1 true WO2011127467A1 (en) 2011-10-13

Family

ID=44763311

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/031935 WO2011127467A1 (en) 2010-04-09 2011-04-11 Devices, systems, and methods for biomarker stabilization

Country Status (1)

Country Link
WO (1) WO2011127467A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9492444B2 (en) 2013-12-17 2016-11-15 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9707184B2 (en) 2014-07-17 2017-07-18 Pharmaceutical Manufacturing Research Services, Inc. Immediate release abuse deterrent liquid fill dosage form
US9918702B2 (en) 2014-08-12 2018-03-20 Nextgen Jane, Inc. System and method for monitoring health based on collected bodily fluid
US10172797B2 (en) 2013-12-17 2019-01-08 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US10195153B2 (en) 2013-08-12 2019-02-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
CN110376378A (en) * 2019-07-05 2019-10-25 中国医学科学院肿瘤医院 It can be used for the markers in detecting model of pulmonary cancer diagnosis
CN112394102A (en) * 2020-11-05 2021-02-23 上海交通大学医学院附属瑞金医院 Marker for detecting hypopituitarism and application thereof
US10959958B2 (en) 2014-10-20 2021-03-30 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form
CN113341009A (en) * 2021-05-25 2021-09-03 海南医学院 Urine biomarker for early diagnosis of angiostrongylus cantonensis disease, screening method and application
CN114487409A (en) * 2022-04-14 2022-05-13 启德医药科技(苏州)有限公司 Detection method and detection kit for activity of transpeptidase
US11446011B2 (en) 2016-04-13 2022-09-20 Nextgen Jane, Inc. Sample collection and preservation devices, systems and methods
CN115598344A (en) * 2022-11-09 2023-01-13 山东和佰生物科技有限公司(Cn) Detection kit for fecal pancreatic elastase 1 and preparation method thereof

Citations (9)

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

Patent Citations (9)

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

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10195153B2 (en) 2013-08-12 2019-02-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US10639281B2 (en) 2013-08-12 2020-05-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US10172797B2 (en) 2013-12-17 2019-01-08 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US10792254B2 (en) 2013-12-17 2020-10-06 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9492444B2 (en) 2013-12-17 2016-11-15 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9707184B2 (en) 2014-07-17 2017-07-18 Pharmaceutical Manufacturing Research Services, Inc. Immediate release abuse deterrent liquid fill dosage form
US9918702B2 (en) 2014-08-12 2018-03-20 Nextgen Jane, Inc. System and method for monitoring health based on collected bodily fluid
US10959958B2 (en) 2014-10-20 2021-03-30 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form
US11446011B2 (en) 2016-04-13 2022-09-20 Nextgen Jane, Inc. Sample collection and preservation devices, systems and methods
US11864740B2 (en) 2016-04-13 2024-01-09 Nextgen Jane, Inc. Sample collection and preservation devices, systems and methods
CN110376378A (en) * 2019-07-05 2019-10-25 中国医学科学院肿瘤医院 It can be used for the markers in detecting model of pulmonary cancer diagnosis
CN110376378B (en) * 2019-07-05 2022-07-26 中国医学科学院肿瘤医院 Marker combined detection model for lung cancer diagnosis
CN112394102A (en) * 2020-11-05 2021-02-23 上海交通大学医学院附属瑞金医院 Marker for detecting hypopituitarism and application thereof
CN112394102B (en) * 2020-11-05 2023-05-26 上海交通大学医学院附属瑞金医院 Marker for detecting hypopituitarism and application thereof
CN113341009A (en) * 2021-05-25 2021-09-03 海南医学院 Urine biomarker for early diagnosis of angiostrongylus cantonensis disease, screening method and application
CN113341009B (en) * 2021-05-25 2023-09-08 海南医学院 Early diagnosis urine biomarker for Guangzhou pipe-line nematode disease and application thereof
CN114487409B (en) * 2022-04-14 2022-07-19 启德医药科技(苏州)有限公司 Detection method and detection kit for activity of transpeptidase
CN114487409A (en) * 2022-04-14 2022-05-13 启德医药科技(苏州)有限公司 Detection method and detection kit for activity of transpeptidase
CN115598344A (en) * 2022-11-09 2023-01-13 山东和佰生物科技有限公司(Cn) Detection kit for fecal pancreatic elastase 1 and preparation method thereof
CN115598344B (en) * 2022-11-09 2023-04-18 山东和佰生物科技有限公司 Detection kit for fecal pancreatic elastase 1 and preparation method thereof

Similar Documents

Publication Publication Date Title
US8501669B2 (en) Systems and methods of screening biomarkers in bodily fluids
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: 11766859

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11766859

Country of ref document: EP

Kind code of ref document: A1