WO2021207647A1 - Small molecule drugs as targeted therapeutics - Google Patents

Small molecule drugs as targeted therapeutics Download PDF

Info

Publication number
WO2021207647A1
WO2021207647A1 PCT/US2021/026650 US2021026650W WO2021207647A1 WO 2021207647 A1 WO2021207647 A1 WO 2021207647A1 US 2021026650 W US2021026650 W US 2021026650W WO 2021207647 A1 WO2021207647 A1 WO 2021207647A1
Authority
WO
WIPO (PCT)
Prior art keywords
combination
disease
compound
salt
ester
Prior art date
Application number
PCT/US2021/026650
Other languages
French (fr)
Inventor
Ken Pingsheng HU
Original Assignee
Computational International LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Computational International LLC filed Critical Computational International LLC
Publication of WO2021207647A1 publication Critical patent/WO2021207647A1/en

Links

Classifications

    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H20/00ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance
    • G16H20/10ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance relating to drugs or medications, e.g. for ensuring correct administration to patients
    • 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
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H50/00ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics
    • G16H50/20ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics for computer-aided diagnosis, e.g. based on medical expert systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • A61K9/0078Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy for inhalation via a nebulizer such as a jet nebulizer, ultrasonic nebulizer, e.g. in the form of aqueous drug solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M11/00Sprayers or atomisers specially adapted for therapeutic purposes
    • A61M11/005Sprayers or atomisers specially adapted for therapeutic purposes using ultrasonics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M15/00Inhalators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M15/00Inhalators
    • A61M15/0085Inhalators using ultrasonics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M16/00Devices for influencing the respiratory system of patients by gas treatment, e.g. mouth-to-mouth respiration; Tracheal tubes
    • A61M16/06Respiratory or anaesthetic masks
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M16/00Devices for influencing the respiratory system of patients by gas treatment, e.g. mouth-to-mouth respiration; Tracheal tubes
    • A61M16/06Respiratory or anaesthetic masks
    • A61M16/0666Nasal cannulas or tubing
    • A61M16/0672Nasal cannula assemblies for oxygen therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M2205/00General characteristics of the apparatus
    • A61M2205/50General characteristics of the apparatus with microprocessors or computers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M2205/00General characteristics of the apparatus
    • A61M2205/50General characteristics of the apparatus with microprocessors or computers
    • A61M2205/52General characteristics of the apparatus with microprocessors or computers with memories providing a history of measured variating parameters of apparatus or patient
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M5/00Devices for bringing media into the body in a subcutaneous, intra-vascular or intramuscular way; Accessories therefor, e.g. filling or cleaning devices, arm-rests
    • A61M5/14Infusion devices, e.g. infusing by gravity; Blood infusion; Accessories therefor
    • A61M5/142Pressure infusion, e.g. using pumps
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A90/00Technologies having an indirect contribution to adaptation to climate change
    • Y02A90/10Information and communication technologies [ICT] supporting adaptation to climate change, e.g. for weather forecasting or climate simulation

Definitions

  • the method can comprise: a) administering to a subject a therapeutically effective amount of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof; b) directly or indirectly at least partially increasing in a subject a level, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof; c) directly or indirectly at least partially reducing in a subject a level, an activity, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, or any combination thereof;
  • a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be comprised in a pharmaceutical composition comprising an excipient, a diluent, or a carrier.
  • a pharmaceutical composition can comprise a first compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof; and a second compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • a pharmaceutical composition can be in unit dose form.
  • a pharmaceutical composition can comprise a molecule selected from a group consisting of: isoprene, decanal, 4- methylbenzaldehyde, 3,7-dimethyloctan-3-ol, l-methyl-4(l -methyl -2 -propenyl)-benzene, 1- ethenyl-3 -ethyl-benzene, l-ethenyl-4-ethyl-benzene, 1-phenylbut-l-ene, octan-2-one, heptan-
  • a pharmaceutical composition can further comprise albuterol or a salt thereof.
  • a pharmaceutical composition can be administered with an inhaler, a nebulizer, a vape device, a bronchodilator, a face mask, a nasal cannula, a diffuser, a tent, a container at least partially surrounding a subject’s head, or any combination thereof.
  • a method can further comprise administering a plant, a portion of a plant, an extract thereof, an herb, a portion of an herb, an extract thereof, or any combination thereof.
  • a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof can be comprised in an implant.
  • an implant at least partially can release a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, or a salt thereof, or any combination thereof over a time when at least partially implanted in a subject.
  • a starting material can be administered to a subject and a starting material can be transformed into a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or a combination thereof.
  • a compound of Table 1 a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or a combination thereof.
  • a compound of formula 1 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof ca ne covalently linked to an antibody or fragment thereof.
  • a compound of formula 1 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof ca ne covalently linked to an antibody or fragment thereof.
  • a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be covalently linked by a cleavable linker to an antibody or fragment thereof.
  • a cell endocytosis can be at least partially promoted in a subject.
  • a cell endocytosis can be of a polypeptide, a virus, a lipid, a polynucleotide, a molecule, or any combination thereof.
  • a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be administered and can at least partially cross a blood brain barrier in a subject.
  • an apoptosis or a cell death can be at least partially induced in a subject.
  • an induction can be at least in part a selective induction.
  • a cell growth, a cell communication, a cell metastasis, or any combination thereof can be at least partially inhibited in a subject.
  • an inhibition can be at least in part a selective inhibition.
  • a Reactive Oxygen Species can be at least partially induced within a subject.
  • a cell can be a cancer cell.
  • an immune cell can be at least partially deactivated in a subject.
  • a deactivation can be at least in part a selective deactivation.
  • an immune cell can be at least partially activated in a subject.
  • an activation can at least in part be a selective activation.
  • an immune cell can be at least partially induced into maturation, differentiation, or a combination thereof.
  • an immune cell can comprise a dendritic cell, a neutrophil, a granulocyte, a monocyte, a lymphocyte, or any combination thereof. In some embodiments, an immune cell can comprise a dendritic cell, a neutrophil, a granulocyte, a monocyte, a lymphocyte, or any combination thereof. In some embodiments, an immune cell can comprise a dendritic cell, a neutrophil, a granulocyte, a monocyte, a lymphocyte, or any combination thereof. In some embodiments, an immune cell can comprise a dendritic cell, a
  • a dendritic cell, a CD4 T-cell, a CD8 T-cell, or any combination thereof at least partially can directly or at least partially can indirectly suppress a tumor in a subject.
  • an immune cell can be a natural killer (NK) cell.
  • a NK cell at least partially can remove alpha-synuclein aggregates in a subject.
  • a treating or preventing disease can comprise administering a pharmaceutical composition comprising an excipient, a carrier, a diluent or any combination of these and a compound of
  • a treating or preventing can comprise administering a polynucleotide to a subject.
  • a polynucleotide can encode for a polypeptide or biologically active fragment thereof in a biochemical pathway for making a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • treating or preventing disease can comprise administering a genetically engineered virus comprising at least one polynucleotide coding for a polypeptide, or a biologically active fragment thereof, in a pathway to produce a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • treating or preventing disease can comprise administering a vector comprising at least one polynucleotide coding for a polypeptide, or a biologically active fragment thereof, in a pathway to produce a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • a vector can be a plasmid.
  • treating or preventing disease can comprise administering a genetically engineered cell to capture, to produce, or to chemically alter a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • a genetically engineered cell can be a eukaryotic cell.
  • a genetically engineered cell can be a prokaryotic cell.
  • a method can comprise altering of a composition of a food, a beverage, a nutrient, or any combination thereof consumed by a subject.
  • treating or preventing disease can comprise administering a compound to a subject.
  • a compound can be an at least partial inhibitor, a salt thereof, or a formulation containing at least one of these, of a polypeptide or biologically active fragment thereof involved in a competing biological pathway.
  • a production of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be altered.
  • treating or preventing disease can comprise administering an intermediate compound, a salt thereof, or a formulation thereof in a biological pathway.
  • a production of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be altered.
  • treating or preventing disease can comprise at least partially disrupting a communication between one or more cells in a subject.
  • a treating or preventing disease can comprise editing a polynucleotide.
  • a polynucleotide can encode for a polypeptide in a biochemical pathway for making a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, or a salt thereof.
  • a polynucleotide can comprise a ribose.
  • a polynucleotide can comprise a deoxyribose.
  • an edited polynucleotide can be comprised in an isolated cell and an isolated cell can be administered to a subject.
  • an isolated cell can be autologous.
  • an isolated cell can be allogenic.
  • a treating or preventing disease can comprise administering an at least partial inhibitor, a salt thereof, or a formulation containing at least one of these: of a polypeptide or biologically active fragment thereof, involved in a production of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • treating or preventing disease can comprise administering an at least partially interfering polynucleotide to bind to RNA or DNA to at least partially inhibit polypeptide production in a biochemical pathway for making a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof in a subject.
  • a polynucleotide can comprise a ribose.
  • a polynucleotide can comprise a deoxyribose.
  • a treating or preventing disease can comprise administering a CRISPR system to edit a polynucleotide to at least partially inhibit polypeptide production in a biochemical pathway for making a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof in a subject.
  • a polynucleotide can comprise a ribose.
  • a polynucleotide can comprise a deoxyribose.
  • an edited polynucleotide can comprise a nonsense mutation, a missense mutation, a deletion, or any combination thereof.
  • a treating or preventing disease can comprise administering an antibody, an aptamer, or a fragment thereof to at least partially bind to a compound of Table 1 a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof in a subject.
  • treating or preventing disease can comprise administering a molecule, a salt thereof, or a formulation containing at least one of these to capture a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof in a subject.
  • treating or preventing disease can comprise administering dialysis to a patient.
  • a dialysis can filter at least some of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof from a patient.
  • a disease can comprise a cancer, a tumor, an infectious disease, a skeletal disease, a urogenital disease, a digestive disease, a muscular disease, a neurological disease, an autoimmune disease, a respiratory disease, a cardiac disease, an endocrine disease, a liver disease, a hereditary disease, or any combination thereof.
  • a disease can comprise a cancer.
  • a cancer can comprise a sarcoma, a carcinoma, a melanoma, a lymphoma, leukemia, blastoma, germ cell tumor, myeloma, or any combination thereof.
  • a disease can comprise an infectious disease.
  • an infectious disease can comprise a viral infection.
  • a viral infection can comprise, influenza A, influenza B, SARS-CoV-2 (COVID-19), SARS-COV, MERS-CoV,
  • a disease can comprise an infectious disease.
  • an infectious disease can comprise a bacterial infection.
  • a bacterial infection can comprise Helicobacter pylori, Staphylococcus aureus,
  • Streptococcus pyogenes Neisseria gonorrhoeae, Treponema pallidum, Borrelia burgdorferi,
  • a disease can comprise an infectious disease.
  • an infectious disease can comprise a fungal, a parasitic infection, or any combination thereof.
  • an infection can be a drug resistant infection.
  • an infection can be a multi-drug resistant infection.
  • a disease can comprise a skeletal disease.
  • a skeletal disease can comprise osteoarthritis, osteoporosis, Paget disease of bone, or any combination thereof.
  • a disease can comprise an urogenital disease.
  • a urogenital disease can comprise benign prostate hyperplasia, urinary incontinence, kidney stones, kidney disease, erectile dysfunction, interstitial cystitis, prostatitis, overactive bladder, endometriosis, or any combination thereof.
  • a disease can comprise a digestive disease, wherein a digestive disease can comprise irritable bowel syndrome, celiac disease, gastroesophageal reflux disease, Chron’s disease, gastroparesis, or any combination thereof.
  • a disease can comprise a muscular disease.
  • a muscular disease can comprise muscular dystrophy, amyotrophic lateral sclerosis, Charcot-Marie-Tooth disease, myasthenia gravis, myopathy, myositis, peripheral neuropathy, or any combination thereof.
  • a disease can comprise a neurological disease.
  • a neurological disease can comprise a psychiatric disease, spinal cord injury, epilepsy, headache, migraine, dementia, vertigo, seizures, dizziness, sleep disorder, aneurysm, chronic fatigue syndrome, cerebral palsy, amyotrophic lateral sclerosis, neuralgia, neuropathy, or any combination thereof.
  • a disease can comprise an autoimmune disease.
  • an autoimmune disease can comprise type 1 diabetes, rheumatoid arthritis, psoriasis, multiple sclerosis, systemic lupus erythematosus, Sjogren’s syndrome, an allergy, or any combination thereof.
  • a disease can comprise a respiratory disease.
  • a respiratory disease can comprise asthma, chronic obstructive pulmonary disease, chronic bronchitis, emphysema, cystic fibrosis, plural effusion, or any combination thereof.
  • a disease can comprise a cardiac disease.
  • a cardiac disease can comprise a congenital heart disease, arrhythmia, coronary artery disease, dilated cardiomyopathy, myocardial infarction, congestive heart failure, high blood pressure, mitral regurgitation, pulmonary stenosis, or any combination thereof.
  • a disease can comprise an endocrine disease.
  • an endocrine disease can comprise type 2 diabetes, hypothyroidism, hyperthyroidism, low testosterone, obesity, Addison’s disease, Cushing’s syndrome, Graves’ disease, thyroiditis, prolactinoma, or any combination thereof.
  • a disease can comprise a liver disease.
  • a liver disease can be cirrhosis, fibrosis, hemochromatosis, Wilson’s disease, or any combination thereof.
  • a disease can comprise a hereditary disease.
  • a hereditary disease can comprise, an inherited mendelian disorder, a peroxisomal disorder, a mitochondrial disorder, cystic fibrosis, sickle-cell anemia, Marfan syndrome, Huntington’s disease, kidney disease, Tay-Sachs disease, phenylketonuria, hemophilia, or any combination thereof.
  • a treating or preventing a disease can comprise administering a second therapy.
  • a second therapy for cancer can comprise surgery, chemotherapy, radiation therapy, immunotherapy, hormone therapy, a checkpoint inhibitor, targeted drug therapy, CAR T-cell therapy, or a combination thereof.
  • administering of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof and a second administering can be concurrent.
  • administering of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof and a second administering can be sequential.
  • a method of treating or preventing disease can be administered by: injection, inhalation, catheterization, gastrostomy tube administration, intravenous administration, intraosseous administration, ocular administration, otic administration, topical administration, transdermal administration, oral administration, rectal administration, nasal administration, intravaginal administration, intracavernous administration, transurethral administration, buccal administration, sublingual administration, or a combination thereof.
  • an administration can comprise administering a nanoparticle, a viral vector, a viral-like particle, a liposome, an exosome, an extracellular vesicle, a microrobot, a microneedle, an implant, or a combination thereof.
  • an administration can comprise a topical administration, wherein a topical administration can comprise administering a patch, a lotion, a cream, a gel, a spray, an ointment, a liquid formulation, or a combination thereof.
  • a patch can comprise a microneedle patch, a single-layer drug in adhesive, a multi-layer drug in adhesive, a reservoir system, a matrix system, a vapour patch, or any combination thereof.
  • an administration can comprise an injection.
  • an injection can comprise administering an injector, intraarterial injection, intracerebroventricular injection, intraci sternal injection, intramuscular injection, intraorbital injection, intraparenchymal injection, intraperitoneal injection, intraspinal injection, intrathecal injection, intravenous injection, intraventricular injection, stereotactic injection, subcutaneous injection, or any combination thereof.
  • an injection can be administered directly to a site of a disease.
  • an administration can comprise an oral or rectal administration.
  • an oral or rectal administration can comprise administering a pill formulation, a liquid formulation, a powder formulation, or any combination thereof.
  • an administration can comprise an inhalation administration.
  • an inhalation administration can comprise administering an inhaler, a nebulizer, a vape device, a bronchodilator, a face mask, nasal cannula, a diffuser, a tent, a container at least partially surrounding a subject’s head, an aerosol formulation, a powder formulation, a liquid formulation, a gas formulation, or any combination thereof.
  • administering can be at least about once per day, twice per day, three times per day, or 4 times per day.
  • administering can be continuous.
  • administering can be by a pump or an implant.
  • a time of administration can comprise for at least about: 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months 11 months, 1 year, 2 years, 3 years, 4 years, 5 years, or for life.
  • a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can have an identical or a different dosage.
  • a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can have an identical or a different route of administration.
  • methods that can comprise; contacting an isolated cell with a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • a contact an isolated cell can undergo apoptosis or cell death.
  • a cell can be a cancer cell.
  • an isolated cell can produce ROS.
  • a method can be a method of screening.
  • a cell can be a cancer cell.
  • compositions that can comprise an excipient, a carrier, a diluent and a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • a pharmaceutical composition can be in unit dose form.
  • a pharmaceutical composition can comprise a molecule selected from a group consisting of: isoprene, decanal, 4- methylbenzaldehyde, 3,7-dimethyloctan-3-ol, l-methyl-4(l -methyl -2 -propenyl)-benzene, 1- ethenyl-3 -ethyl-benzene, l-ethenyl-4-ethyl-benzene, 1-phenylbut-l-ene, octan-2-one, heptan-
  • a pharmaceutical composition can further comprise albuterol a salt thereof, or any combination thereof.
  • a pharmaceutical composition can be administered with an inhaler, a nebulizer, a vape device, a bronchodilator, a face mask, nasal cannula, a diffuser, a tent, a container at least partially surrounding a subject’s head, or any combination thereof.
  • a method can be a method of diagnosing a disease or a condition and can further comprise at least one of a following: selecting a therapeutic regiment for treating a disease or a condition, selecting a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof to treat a disease or condition, treating a subject for a disease or condition, or any combination thereof.
  • a method can comprise detecting: a.) a presence and a level of a first compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof; b.) a presence and a level of a second compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof; c.) a presence and a level of a third compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof; and d.) comparing a first level to a first reference level, a second level to a second reference level, and a third level to
  • a method can be conducted at least in part on a diagnostic device, a screening device, or a diagnostic and a screening device comprising at least one of; a chromatography column, an eluant, a detector, and a mass spectrometer.
  • a chromatography column can comprise a gas chromatography column and an eluant can comprise a gas.
  • a diagnostic device, a screening device, or a diagnostic and a screening device can comprise a microelectromechanical system (MEMS).
  • MEMS microelectromechanical system
  • a diagnostic device, a screening device, or a diagnostic and a screening device can comprise a breath collection tube.
  • a device can comprise a self-contained power supply (e.g., a battery) and/or a wired power connection (e.g., a plug for electrical power).
  • a diagnostic device, a screening device, or a diagnostic and screening device can have: a) a positive volume of less than about 16 cubic feet, b) a method of transport, wherein a device can be moved by a robot, a drone, or a remote-control transport machine, c) a self- contained power supply, or d) or any combination of (a-c).
  • a robot, a drone, or a remote-control transport machine can be controlled autonomously.
  • a diagnostic device, a screening device, or a diagnostic a screening device can be linked wirelessly or directly to a processing station.
  • a diagnostic device, a screening device, or a diagnostic and screening device can be linked wirelessly by Wi-Fi, a broadband cellular network, or any combination thereof.
  • a processing station can analyze a data remotely.
  • a device can be about: 50 cm long, 30 cm wide, and 25 cm tall. In some embodiments, a device can be less than about: 12 cm long, 7 cm wide, and 3 cm tall. In some embodiments, an additional diagnostic test can be performed on a subject.
  • a sample can comprise a bodily fluid, a compound extruded by a body, a bodily part, a blood sample, a plasma sample, a urine sample, a sweat sample, a breath sample, a skin sample, a buccal sample, a hair sample, a fecal sample, a tear sample, a nasal sample, a nasal mucus sample, a biopsy sample, a saliva sample, a sputum sample or any combination thereof.
  • a compound extruded by a body can comprise an ester, a salt, or a combination thereof of a compound.
  • a compound extruded by a body can comprise a volatile compound.
  • a compound extruded by a body can comprise a metabolite.
  • a sample can further comprise an additive, a preservative, an anticoagulant, a membrane, an antibacterial compound, an antifungal compound, an antiviral, a chemical reagent, an antimicrobial compound, a diluent, or any combination thereof.
  • a sample can be treated with an electromagnetic wave, an ultrasound, a light wave, a radiation, a laser, adjusting a temperature of a sample, or any combination thereof.
  • a method can be used to identify a disease or condition, wherein a sample can be taken from a subject and applied to a device.
  • a computer can be used to at least partially compare a chromatogram in a subject having a disease to a chromatogram of a subject not having a disease, and determining independently a difference in a presence, an absence, a level, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • a chromatogram can comprise a signature of a disease or a condition.
  • a result generated by a method can be evaluated by an algorithm using a processor or microprocessor.
  • an algorithm can comprise a trained algorithm, a machine learning algorithm, or any combination thereof using a processor or microprocessor.
  • a computer can comprise a memory, one or more executable instructions, one or more processors, and a graphical user interface.
  • a result can be communicated via a communication medium.
  • a compound of Table 1 identified by a method, or a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be selected as a therapeutic.
  • a method can comprise a method of diagnosis, of prognosis, a theranostic method, a health level information method, or any combination thereof.
  • a subject can display an altered profile, a level of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be at least partially increased or at least partially decreased.
  • a measurement and/or an analysis e.g., comprising a subject’s metabolic profile
  • 2 hours from 2 hours to 6 hours, from 6 hours to 12 hours, from 12 hours to 24 hours, from 1 day to 2 days, from 2 days to 5 days, from 2 days to 1 week, from 1 week to 2 weeks, from 2 weeks to 1 month, from 1 month to 2 months, from 2 months to 6 months, from 6 months to 1 year, from 1 year to 2 years, from 2 years to 3 years, from 3 years to 5 years, from 5 years to 10 years, from 10 years to 20 years, at least 1 minute, at least 2 minutes, at least 3 minutes, at least 5 minutes, at least 10 minutes, at least 15 minutes, at least 20 minutes, at least 30 minutes, at least 1 hour, at least 2 hours, at least 6 hours, at least 12 hours, at least 24 hours, at least 2 days, at least
  • a healthy subject’s metabolic profile can be recorded for at least 2 weeks.
  • a healthy subject’s profile can be recorded a plurality of times over a period of at least two weeks.
  • a metabolic profile of a subject can be unknown, wherein a healthy metabolic profile can be determined from factors comprising a subject’s genetic profile, a diseased metabolic profile, a healthy population profile, or any combination thereof.
  • a method can be performing a diagnostic test and administering a therapeutic comprising a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • a diagnostic test can comprise a diagnostic test, a prognostic test or a theranostic test.
  • running a diagnostic test can comprise subjecting at least a portion of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof to a device comprising: a chromatography column, an eluant, a detector, and a mass spectrometer.
  • a device comprising: a chromatography column, an eluant, a detector, and a mass spectrometer.
  • a method can comprise: a.) administering to a subject a therapeutically effective amount of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof; b.) directly or indirectly at least partially increasing in a subject a level, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof; c.) directly or indirectly at least partially reducing in a subject a level, an activity, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof,
  • an administering can comprise administering an oral formulation.
  • an oral formulation ca be a dietary supplement.
  • a promoting or maintaining health can comprise promoting or maintaining: cardiovascular health, respiratory health, skeletal health, urogenital health, digestive health, muscular health, neural health, immune system health, liver health, endocrine health or any combination thereof.
  • compositions in unit dose form comprising, a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • a composition in unit dose form can be placed in a closed container and placed in a room at about 23 degrees Celsius with a relative atmospheric humidity of about 50% for at least about 1 month.
  • a time period at least about 80% of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof remains.
  • a time period can be about 1 month to about 2 years.
  • a method can comprise detecting a level of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof, from a sample of a subject having or suspected of having a disease or condition and comparing a level of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof, to a reference level and administering to a subject a therapeutically effective amount of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • a method can be conducted at least in part on a diagnostic device, a screening device, or a diagnostic and a screening device comprising at least one of; a chromatography column, an eluant, a detector, and a mass spectrometer.
  • a chromatography column can comprise a gas chromatography column and an eluant can comprise a gas.
  • a diagnostic device, a screening device, or a diagnostic and a screening device can comprise a microelectromechanical system (MEMS).
  • MEMS microelectromechanical system
  • a diagnostic device, a screening device, or a diagnostic and a screening device can comprise a breath collection tube.
  • an increase or decrease in concentration of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can indicate a treatment may be at least partially decreasing a presence of a disease, disease symptoms, or a combination thereof.
  • an increase or decrease in concentration of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can indicate a viral infection in a subject.
  • a diagnostic device can be operably linked to a database or may not linked to a database.
  • a database can comprise a profile pattern of different diseases.
  • a device compares a patient’s profile pattern with a database’s profile to identify a disease.
  • FIG. 1 shows a chromatogram of a patient with breast cancer, in accordance with embodiments.
  • FIG. 2 shows a chromatogram of a patient with kidney cancer, in accordance with embodiments.
  • FIG. 3A shows an overlay of chromatograms from gastric cancer patients’ blood, in accordance with embodiments.
  • FIG. 3B, FIG. 3C, and FIG. 3D show data from analysis of individual gastric cancer patients’ blood, in accordance with embodiments.
  • FIG. 4A shows an overlay of chromatograms from lung cancer patients’ blood, in accordance with embodiments.
  • FIG. 4B, FIG. 4C, and FIG. 4D show data from analysis of individual lung cancer patients’ blood, in accordance with embodiments.
  • FIG. 5A shows an overlay of chromatograms from gastric cancer patients’ urine, in accordance with embodiments.
  • FIG. 5B, FIG. 5C, and FIG. 5D show data from analysis of individual gastric cancer patients’ urine, in accordance with embodiments.
  • FIG. 6A shows an overlay of chromatograms from kidney cancer patients’ urine, in accordance with embodiments.
  • FIG. 6B, FIG. 6C, and FIG. 6D show data from analysis of individual kidney cancer patients’ urine, in accordance with embodiments.
  • FIG. 7A shows an overlay of chromatograms from lung cancer patients’ urine, in accordance with embodiments.
  • FIG. 7B, FIG. 7C, and FIG. 7D show data from analysis of individual lung cancer patients’ urine, in accordance with embodiments.
  • FIG. 8A shows an overlay of chromatograms from healthy control patients’ blood, in accordance with embodiments.
  • FIG. 8B and FIG. 8C show data from analysis of individual healthy control patients’ blood, in accordance with embodiments.
  • FIG. 9A shows an individual breast cancer patient’s metabolite volatile organic compounds (VOC) profile.
  • FIG. 9B shows an overlay of chromatograms of six breast cancer patients’ metabolite VOC profiles, in accordance with embodiments.
  • FIG. 10A shows an individual breast cancer patient’s metabolite volatile organic compounds (VOC) profile.
  • FIG. 10B shows an overlay of chromatograms of five kidney cancer patients’ metabolite VOC profiles, in accordance with embodiments, in accordance with embodiments.
  • FIG. 11A shows an individual breast cancer patient’s metabolite volatile organic compounds (VOC) profile.
  • FIG. 11B shows an overlay of chromatograms of five lymphoma patients’ metabolite VOC profiles, in accordance with embodiments, in accordance with embodiments, in accordance with embodiments.
  • FIG. 12A shows an overlay of chromatograms from breast and kidney cancer patients, in accordance with embodiments.
  • FIG. 12B, FIG. 12C, and FIG. 12D show data from analysis of individual renal cancer patients’ blood, in accordance with embodiments.
  • FIG. 12E, FIG. 12F, and FIG. 12G show data from analysis of individual breast cancer patients’ blood, in accordance with embodiments.
  • FIG. 13 shows an overlay of chromatograms from lung cancer and liver cancer patients, in accordance with embodiments.
  • FIG. 14 shows chromatograms from first thaw and second thaw analysis of kidney cancer patients’ urine, in accordance with embodiments.
  • FIG. 15 shows saliva chromatograms, in accordance with embodiments.
  • FIG. 16 shows chromatograms from four healthy volunteers’ breath (top row, second row) and a blank control (bottom left), in accordance with embodiments.
  • FIG. 17 shows a chromatogram from a cancer patient, in accordance with embodiments.
  • FIG. 18 shows a chromatogram from a cancer patient, in accordance with embodiments.
  • FIG. 19 shows a chromatogram from a lymphoma patient, in accordance with embodiments.
  • FIG. 20 shows a chromatogram from a lung cancer patient, in accordance with embodiments.
  • FIG. 21 shows a chromatogram from a prostate cancer patient, in accordance with embodiments.
  • FIG. 22 shows a chromatogram of a healthy control and cancer patients, in accordance with embodiments.
  • FIG. 23 shows a chromatogram of a healthy control and a lab standard, in accordance with embodiments.
  • FIG. 24 shows a chromatogram of blood and urine samples from kidney cancer patients, in accordance with embodiments.
  • FIG. 25 shows a computer control system that is programmed or otherwise configured to implement methods provided herein.
  • FIG. 26 is a diagram showing a method and system as disclosed herein.
  • FIG. 27A-B shows a gas chromatography (GC) device for testing a sample, in accordance with embodiments.
  • GC gas chromatography
  • FIG. 28A-B shows the GC device testing a sample on a subject, in accordance with embodiments.
  • a device as described herein can be portable, easy to operate, stand alone or in a cluster, low cost, fast, remote control capable, network connected or otherwise, gas chromatography device.
  • the device can use ambient air as well as other gases as a carrier gas to sample a compound, such as one or more Volatile Organic Compounds (VOCs) emission from any bodily fluid such as blood or urine, as well as any body parts such as skin, hair, nails, cells or whole tumor.
  • VOCs Volatile Organic Compounds
  • a device or method disclosed herein can comprise or comprise the use of a chemical that is not a VOC, e.g., for analysis of a sample from a subject and/or in the diagnosis of a condition (e.g., disease).
  • a device or method disclosed herein can comprise the use of a chemical that is not organic, e.g., for analysis of a sample from a subject and/or in the diagnosis of a condition (e.g., disease).
  • a device or method disclosed herein can comprise nitric oxide (NO) and/or nitrogen dioxide (N02) or the use thereof, e.g., for analysis of a sample from a subject and/or in the diagnosis of a condition (e.g., disease).
  • Data produced by the gas chromatography device can detect as well as separate individual VOCs as biomarkers, measure a presence or absence or concentration one or more compounds, such as VOCs. Data produced can be fed into a software comprising one or more algorithms capable of using these data to identify a presence or an absence of a disease such as Parkinson’s, infectious diseases, cancer or others.
  • the disease identification may be not limited to use a certain biomarker or biomarkers, but these data can also be used to construct patterns produced by the gas chromatography device.
  • these patterns can be used for disease screening, diagnostics, companion diagnostics, prognostics, theranostics, or any combination thereof.
  • a system can be for analyzing a gas mixture.
  • the system can comprise: a filter; a trap; a chromatographic column; a detector; and a pump.
  • the trap and the pump can be fluidly connected to form a first gas flow path.
  • the filter, the trap, the chromatographic column, the detector, and the pump can be fluidly connected to form a second gas flow path.
  • the detector and the pump can be fluidly connected to form a third gas flow path.
  • the chromatographic column can be a gas-solid adsorption chromatographic column.
  • the chromatographic column can be a gas-liquid gas chromatography column.
  • the trap further can comprise an adsorbent material.
  • the filter can be an activated charcoal filter.
  • the detector can be selected from the group consisting of a photo ionization detector, a mass spectrometer, a spectrophotometer, and a thermal conductivity detector.
  • the detector can be a photo ionization detector.
  • the pump can provide negative pressure.
  • the system can further comprise a housing. In some cases, the housing can be no larger than 216 cubic inches.
  • a method can comprise a method of analyzing a chemical compound in a gas mix.
  • the method can comprise directing flow of the gas mix through a trap to concentrate at least a quantity of the chemical compound; redirecting flow of the gas mix through a filter to provide a filtered gas flow to the trap; releasing at least a quantity of the concentrated chemical compound into the filtered gas flow; and analyzing at least a quantity of the released concentrated chemical compound.
  • the chemical compound can be an organic compound.
  • the organic compound can be a volatile organic compound.
  • analysis of at least a quantity of the released concentrated chemical compound can comprise running at least a quantity of the released concentrated chemical compound through a gas chromatography column.
  • the gas chromatography column can be a gas-solid adsorption chromatographic column.
  • the chromatographic column can be a gas-liquid gas chromatography column.
  • analysis of at least a quantity of the released concentrated chemical compound can comprise identifying the organic compound by a method selected from the group consisting of photo ionization, mass spectrometry, spectrophotometry, and thermal conductivity.
  • analysis of at least a quantity of the released concentrated chemical compound can further comprise quantifying the chemical compound.
  • the gas mix can be an environmental gas mix.
  • the gas mix can comprise gases exhaled or otherwise emitted by a living subject.
  • the gas mix can be air.
  • the term “about” or “approximately” can mean within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, e.g ., the limitations of the measurement system.
  • “about” can mean plus or minus 10%, per the practice in the art.
  • “about” can mean a range of plus or minus 20%, plus or minus 10%, plus or minus 5%, or plus or minus 1% of a given value.
  • the term can mean within an order of magnitude, within 5-fold, or within 2-fold, of a value.
  • the term “substantially” as used herein can refer to a value approaching 100% of a given value. In some cases, the term can refer to an amount that can be at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9%, or 99.99% of a total amount. In some cases, the term can refer to an amount that can be about 100% of a total amount.
  • the term “homology” can refer to a % identity of a sequence to a reference sequence. As a practical matter, whether any particular sequence can be at least 50%, 60%, 70%, 80%, 85%, 90%, 92%, 95%, 96%, 97%, 98% or 99% identical to any sequence described herein (which can correspond with a particular nucleic acid sequence described herein), such particular polypeptide sequence can be determined conventionally using known computer programs such the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, Wis. 53711).
  • the parameters can be set such that the percentage of identity is calculated over the full length of the reference sequence and that gaps in homology of up to 5% of the total reference sequence are allowed.
  • administer can refer to methods that can be used to enable delivery of compounds or compositions described herein, to the desired site of biological action.
  • delivery can include injection, inhalation, catheterization, gastrostomy tube administration, intravenous administration, intraosseous administration, ocular administration, otic administration, topical administration, transdermal administration, oral administration, rectal administration, nasal administration, intravaginal administration, intracavemous administration, transurethral administration, buccal administration, sublingual administration, or a combination thereof.
  • Delivery can include direct application to the affect tissue or region of the body.
  • Delivery can include a parenchymal injection, an intra-thecal injection, an intra ventricular injection, or an intra-ci sternal injection.
  • a composition provided herein can be administered by any method.
  • a method of administration can be by inhalation, intraarterial injection, intracerebroventricular injection, intraci sternal injection, intramuscular injection, intraorbital injection, intraparenchymal injection, intraperitoneal injection, intraspinal injection, intrathecal injection, intravenous injection, intraventricular injection, stereotactic injection, subcutaneous injection, epidural, or any combination thereof.
  • Delivery can include parenteral administration (including intravenous, subcutaneous, intrathecal, intraperitoneal, intramuscular, intravascular or infusion), oral administration, inhalation administration, intraduodenal administration, rectal administration.
  • delivery can comprise a nanoparticle, a viral vector, a viral-like particle, a liposome, an exosome, an extracellular vesicle, a microrobot, a microneedle, an implant, or a combination thereof.
  • the implant can partially or fully release a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, or a salt thereof.
  • delivery can be from a device.
  • delivery can be administered by a pump, an infusion pump or a combination thereof.
  • delivery can be by a contact lens, an eye drop, a nasal spray, an ear drop, or any combination thereof.
  • Delivery can include, a pill, an enema, an eye drop, an ear drop, a gas, a mouthwash, a toothpaste, a film, a tea, an elixir, a osmotic delivery system, a dietary supplement, a food, a beverage, a chewing gum, an oil, a lozenge, a mouth spray, a lip balm, a shampoo, a dental implant, a dental ware (e.g.
  • delivery can comprise an inhaler, a humidifier, a diffuser, a vape device, a bronchodilator, a face mask, a nasal cannula, a tent, a container at least partially surrounding the subjects head, a nebulizer, a jet nebulizer, an ultrasonic nebulizer, or a combination thereof.
  • an inhaler can comprise a metered-dose inhaler, a dry powder inhaler or any combination thereof.
  • Delivery can include topical administration (such as a lotion, a cream, a patch, a gel, a spray, a drip, a liquid formulation, an ointment) to an external surface of a surface, such as a skin.
  • a patch can comprise a membrane, a microneedle patch, a single-layer drug in adhesive, a multi-layer drug in adhesive, a reservoir system, a matrix system, a vapour patch or any combination thereof.
  • a subject can administer the composition in the absence of supervision.
  • a subject can administer the composition under the supervision of a medical professional (e.g., a physician, nurse, physician’s assistant, orderly, hospice worker, etc.).
  • a medical professional can administer the composition.
  • a cosmetic professional can administer the composition.
  • subject refers to animals, typically mammalian animals. Any suitable mammal can be administered a composition as described herein (such as an engineered oligonucleotide) or treated by a method as described herein.
  • Non-limiting examples of mammals include humans, non-human primates (e.g., apes, gibbons, chimpanzees, orangutans, mon-keys, macaques, and the like), domestic animals (e.g., dogs and cats), farm animals (e.g., horses, cows, goats, sheep, pigs) and experimental animals (e.g., mouse, rat, rabbit, guinea pig).
  • Mammals can be any age or at any stage of development, for example a mammal can be neonatal, infant, adolescent, adult or in utero. In some embodiments a mammal can be a human.
  • Humans can be more than about: 1, 2, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 years of age. Humans can be less than about: 1, 2, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 years of age.
  • a mammal can be male or female.
  • a subject can be a human.
  • a subject may be suspected of having a disease or condition.
  • the subject can be a patient, such as a patient being treated for a condition or a disease, such as a cancer, an infectious disease, or a neurological condition.
  • the subject may be predisposed to a risk of developing a condition or a disease such as cancer.
  • the subject may be in remission from a condition or a disease, such as a cancer patient.
  • the subject may be healthy.
  • a cell can refer to any mammalian cell, typically a human cell.
  • the human cell can be a skin cell, a cervical cell, a prostate cell, a stem cell, a bone cell, a blood cell, a muscle cell, a fat cell, a nerve cell, an endothelial cell, sperm cell, egg cell, cancer cell, barrier cell, hormone-secreting cell, exocrine-secretory cell, epithelial cell, oral cell, sensory transducer cell, autonomic neuron cell, peripheral neuron cell, central nervous neuron cell, secretory cell, barrier cell, muscle cell, cardiac muscle cell, white blood cell, germ cell, nurse cell, kidney cell, or any combination thereof.
  • the cell will be a cancer cell.
  • treating a disease or condition can include one or more of: reducing the frequency, the severity, or any combination thereof, of a symptom.
  • treating a disease can comprise elimination of a symptom, elimination of an underlying cause, and improvement or remediation of damage.
  • treatment of cancer can include, for example, relieving the pain experienced by a subject suffering from cancer, or causing the regression or disappearance of cancer. Treating may also include: reduced malaise, cessation of cancer side effects, abatement of tumors, or any combination thereof.
  • Treating can include administering a compound of Table 1
  • “treating” of a urogenital disease can include one or more of: reducing the frequency, the severity of symptoms, the elimination of symptoms, the elimination of a symptoms underlying cause, and improvement or remediation of damage.
  • treatment of urogenital disease can include, for example, relieving pain experienced by a subject suffering from a kidney stone, or causing the regression of kidney stones. Treating may also include: reduction of malaise, cessation of urinary side effects, abatement of kidney damage, or any combination thereof.
  • treating of an infectious disease can include one or more of: reducing the frequency or severity of a symptom, elimination of a symptom, elimination of a symptom’s underlying cause, elimination of the infectious disease, and improvement or remediation of damage.
  • treatment of a viral infection can include, for example, relieving the cough experienced by a subject suffering from a viral infection or causing the regression, the disappearance, or any combination thereof of a viral infection. Treating may also include: reduction of malaise, cessation of viral infection side effects, abatement of a viral infection, or any combination thereof.
  • Treatment can include administration to the subject in need thereof the compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or a salt thereof, or a pharmaceutical composition as described herein.
  • Treatment can include one or more than one of the compounds of Table 1, the homologs thereof, the derivatives thereof, the esters thereof, the enantiomers thereof, the diastereomers thereof, the racemates thereof, or the salts thereof or any combination thereof delivered in a single dose or in multiple doses.
  • preventing a disease or a condition can include inhibiting a disease from occurring in a subject.
  • a diagnostic test can comprise an imaging procedure, a blood count analysis, a tissue pathology analysis, a biomarker analysis, a biopsy, a magnetic resonance image procedure, a physical examination, a urine test, an ultrasonography procedure, a genetic test, a liver function test, a positron emission tomography procedure, a X-ray, serology, an angiography procedure, an electrocardiography procedure, an endoscopy, a diagnostic polymerase chain reaction test (PCR), a pap smear, a hematocrit test, a skin allergy test, a urine test, a colonoscopy, an enzyme-linked immunosorbent assay (ELISA), microscopy analysis, bone marrow examination, rapid diagnostic test, pregnancy test, organ function test, toxicology test, infectious disease test, bodily fluids test, or any combination thereof.
  • PCR diagnostic polymerase chain reaction test
  • a pap smear a hematocrit test
  • a skin allergy test a urine test
  • a diagnostic test can be comprised of the unit described herein.
  • the unit described herein diagnostic test can comprise a chromatography column, an eluant, a detector, a mass spectrometer, a microelectromechanical system (MEMS), a sample collection system, a breath collection tube or any combination thereof.
  • MEMS microelectromechanical system
  • a biomolecule disclosed herein can be a polypeptide, a protein, a lipid, a nucleic acid, a carbohydrate, a saccharide, a polysaccharide, a modified protein, a glycosylated protein, a modified lipid, a lipoprotein or any combination thereof.
  • a compound disclosed herein can be a small molecule.
  • a small molecule can be a low molecular weight organic compound. In some embodiments the small molecule can be less than about: 10 Daltons, 50 Daltons, 100 Daltons,
  • the small molecule can be more than about: 10 Daltons,
  • a small molecule can comprise, a carbon, a hydrogen, a nitrogen, an oxygen, a phosphorus, a sulfur, an organic atom, an inorganic atom, a plurality of molecular atoms, or any combination thereof.
  • a small molecule can bind to a biological molecule such as a protein, a nucleic acid, a polysaccharide, a lipid, or any combination thereof.
  • a small molecule can be an enzyme inhibitor, an enzyme activator, a cell signaling molecule, metabolite, a hormone, a drug, a pesticide, or any combination thereof. In some cases, a small molecule can bind to a biological channel, receptor, or any combination thereof.
  • a small molecule can be a natural product (i.e. a sugar, a lipid, an amino acid, a fatty acid, a phenolic compound, an alkaloid or any combination thereof). In some cases, a small molecule can be a synthetic product. In some embodiments, a small molecule can be chemically stable, thermally stable, biologically stable or any combination thereof. In some embodiments a small molecule can bind to a single molecule.
  • a small molecule may bind to multiple molecules.
  • a small molecule can have a half-life from about: 5 seconds to 10 seconds, 10 seconds to 30 seconds, 30 seconds to 60 seconds, 1 minute to 5 minutes, 5 minutes to 10 minutes, 10 minutes to 30 minutes, 30 minutes to 60 minutes, 1 hour to 2 hours, 2 hours to 4 hours, 4 hours to 8 hours, 8 hours to 12 hours, 12 hours to 16 hours, 16 hours to 24 hours, 1 day to 2 days, 2 days to 4 days, 4 days to 7 days, 1 week to 2 weeks, 2 weeks to 3 weeks, 3 weeks to 4 weeks, 1 month to 2 months, 2 months to 4 months, 4 months to 6 months, 6 months to 12 months or 1 year to about 2 years.
  • the compounds disclosed in here can be an isomer.
  • an isomer can be a compound having the same molecular formula but a different structure. Isomers which may differ in configuration, conformation, or a combination thereof can be referred to as stereoisomers.
  • An isomer can also be used to refer to an enantiomer.
  • An enantiomer can be used to describe one of a pair of molecular isomers which can be a mirror image of each other and non-superimposable.
  • a compound disclosed herein can have an asymmetric center and can exist in different enantiomeric and diastereomeric forms.
  • the compounds disclosed herein can relate to the use of all optical isomers and stereoisomers of a compound disclosed herein, a mixture thereof, a pharmaceutical composition, and to a method of treatment that can contain them.
  • a compound disclosed herein can exist as a racemate.
  • a racemate can be a racemic mixture that can contain both enantiomers of a compound in equal amounts.
  • a compound disclosed herein can exist as a tautomer.
  • a compound disclosed herein can relate to the use of a tautomer, a mixture thereof, a pharmaceutical composition, and to a method of treatment that can contain a tautomer.
  • a homolog of a compound disclosed herein can refer to a structure that can be similar to the parent structure.
  • a homolog can refer to similarity with respect to a structure, a function, a chemical composition or any combination thereof.
  • the homolog may not be readily derived synthetically from the parent structure.
  • the homolog can be derived synthetically from the parent structure.
  • a derivative of a compound disclosed herein can refer to a chemical substance related structurally a compound disclosed herein.
  • a derivative can be made from the structurally-related parent compound in one or more steps. The general physical and chemical properties of a derivative can be similar to a parent compound.
  • bioisosteric groups can have similar spatial or electronic requirements to the parent group but can exhibit differing or improved physicochemical or other properties. For example, some moieties are described in Patini et al., Chem. Rev, 1996, 96, 3147-3176.
  • the compounds disclosed herein can be an ester.
  • an ester can be derived from a carboxylic acid.
  • a carboxylic acid can contain the — COOH group, and in an ester the hydrogen in this group can be replaced by a hydrocarbon group.
  • the hydrocarbon group can be an alkyl group (e.g. methyl or ethyl, or one comprising a benzene ring.)
  • an ester can refer to a chemical moiety with formula — COOR, where R can represent an alkyl, a cycloalkyl, an aryl, a heteroaryl, or a heteroalicyclic.
  • R can represent an alkyl, a cycloalkyl, an aryl, a heteroaryl, or a heteroalicyclic.
  • R can represent an alkyl, a cycloalkyl, an aryl, a heteroaryl, or a heteroalicyclic.
  • an ester can be an alkoxycarbonyl group appended to the parent molecule on an available carbon atom.
  • an ester can be an alkoxycarbonyl group appended to the parent molecule on one or more available aryl groups, cycloalkyl groups, heterocycle groups, or any combination thereof.
  • a compound containing an ester can be a compound considered to be prodrug.
  • an ester group can comprise, pivaloyloxymethyl, acetoxymethyl, phthalidyl, indanyl and methoxymethyl, or any combination thereof.
  • a “salt” can include a pharmaceutically acceptable salt.
  • pharmaceutically acceptable salts can include those salts prepared by reaction of a compound disclosed herein with a mineral, organic acid or inorganic base, such salts including, acetate, acrylate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, bisulfite, bitartrate, bromide, butyrate, butyn-l,4-dioate, camphorate, camphorsulfonate, caproate, caprylate, chlorobenzoate, chloride, citrate, cyclopentanepropionate, decanoate, digluconate, dihydrogenphosphate, dinitrobenzoate, dodecyl sulfate, ethanesulfonate, formate, fumarate, glucoheptanoate, glycerophosphate, glycolate, hemisulfate, heptanoate,
  • metaphosphate methanesulfonate, methoxybenzoate, methylbenzoate, monohydrogenphosphate, 1-napthalenesulfonate, 2-napthalenesulfonate, nicotinate, nitrate, palmoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, pyrosulfate, pyrophosphate, propiolate, phthalate, phenyl acetate, phenylbutyrate, propanesulfonate, salicylate, succinate, sulfate, sulfite, succinate, suberate, sebacate, sulfonate, tartrate, thiocyanate, tosylate, undeconate, and xylenesulfonate.
  • a compound disclosed herein can be prepared as pharmaceutically acceptable salts formed by reacting the free base form of the compound with a pharmaceutically acceptable inorganic or organic acid, including, but not limited to, inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid metaphosphoric acid, and the like; and organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, Q-toluenesulfonic acid, tartaric acid, trifluoroacetic acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, arylsulfonic acid, methanesulfonic acid, ethanesulfonic acid, 1,2- ethane
  • a compound disclosed herein which can comprise a free acid group reacts with a suitable base, such as the hydroxide, carbonate, bicarbonate, sulfate, of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary or tertiary amine.
  • a suitable base such as the hydroxide, carbonate, bicarbonate, sulfate, of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary or tertiary amine.
  • Representative alkali or alkaline earth salts can include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts and the like.
  • bases can include sodium hydroxide, potassium hydroxide, choline hydroxide, sodium carbonate, N+(Cl-4 alkyl)4, and the like.
  • Representative organic amines useful for the formation of base addition salts can include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like. It may be understood that a compound disclosed herein can also include the quaternization of any basic nitrogen-containing groups they contain. In some embodiments, water or oil-soluble or dispersible products can be obtained by such quatemization.
  • a compound disclosed herein can be prepared as pharmaceutically acceptable salts formed when an acidic proton present in the parent compound either can be replaced by a metal ion, for example an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base.
  • base addition salts can be also prepared by reacting the free acid form of a compound disclosed herein with a pharmaceutically acceptable inorganic or organic base, including, but not limited to organic bases such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like and inorganic bases such as aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like.
  • the salt forms of the disclosed compounds can be prepared using salts of the starting materials or intermediates.
  • a sample can be a bodily fluid, a body odor, a compound extruded from the body, a bodily part, a blood sample, a plasma sample, a urine sample, a sweat sample, a breath sample, a skin sample, a buccal sample, a hair sample, a fecal sample, a tear sample, a nasal sample, a nasal mucus sample, a biopsy sample, a saliva sample, a sputum sample, or any combination thereof.
  • the compound extruded from the body can be a metabolite.
  • the compound extruded from the body can be an ester, a salt, or a combination thereof of the compound.
  • a sample can come from a body fluid.
  • a body fluid can be, amniotic fluid, aqueous humour, vitreous humour, bile, whole blood, blood derivative, breast milk, cerebrospinal fluid, cerumen (earwax), chyle, chyme, endolymph and perilymph, exudates, nipple discharge.
  • a sample can be from a body odor.
  • a chemical extruded from the body can be a breath, a flatus, a burp, an odor of the body, recovered from an item that contacted a body, or any combination thereof.
  • a sample can be from a body part, an organ, a cell or any combination thereof.
  • a sample can be a live cell, or dead cell, a hair, a nail, or any combination thereof.
  • a sample can comprise, an additive, a preservative, an anticoagulant, a membrane, an antibacterial compound, an antifungal compound, an antiviral, a chemical reagent, an antimicrobial compound, a diluent, or any combination thereof.
  • a sample can be treated with an electromagnetic wave, an ultrasound, a light wave, a radiation, a laser, adjusting a temperature of the sample, or any combination thereof.
  • the sample can be heated.
  • the sample can be cooled (e.g. placed in a refrigerator, a freezer, or a combination thereof for a period of time.)
  • a pharmaceutical composition can comprise a small molecule. In some embodiments, a pharmaceutical composition may not comprise a small molecule. In some cases, a pharmaceutical composition can comprise a biomolecule. In some cases, a pharmaceutical composition can comprise both a small molecule and a biomolecule. In some embodiments a small molecule can be a compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof. In some cases, a pharmaceutical composition can comprise a molecule from Table 2.
  • a pharmaceutical composition can comprise a molecule from Table 2 and a compound of Table 1 a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof as described herein.
  • a pharmaceutical composition can comprise a first active ingredient.
  • the first active ingredient can comprise a compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof as described herein.
  • a pharmaceutical composition can comprise more than about: 1 active ingredient, 2 active ingredients, 3 active ingredients, 4 active ingredients, 5 active ingredients, 6 active ingredients, 7 active ingredients, 8 active ingredients, 9 active ingredients, 10 active ingredients, 11 active ingredients ,12 active ingredients, 13 active ingredients, 14 active ingredients, 15 active ingredients, 20 active ingredients, 25 active ingredients, 30 active ingredients, 35 active ingredients, 40 active ingredients, 45 active ingredients, 50 active ingredients, 60 active ingredients, 70 active ingredients, 80 active ingredients ,90 active ingredients, 100 active ingredients, 125 active ingredients, 150 active ingredients, 175 active ingredients, 200 active ingredients, 250 active ingredients, 300 active ingredients, 400 active ingredients, 500 active ingredients, 1000 active ingredients, 5000 active ingredients, or 10,000 active ingredients.
  • the pharmaceutical composition can be formulated in unit dose form.
  • a pharmaceutical composition can comprise 1 to 734 compounds of the compounds of Table 1, the homologs thereof, the derivatives thereof, the esters thereof, the enantiomers thereof, the diastereomers thereof, the racemates thereof, the salts thereof or any combination thereof.
  • the pharmaceutical composition can comprise at least about 1, 2, 3, 4, 5, 6, 7, 8, 9,
  • the pharmaceutical composition can comprise at least 1 to about 734 molecules of Table 1, 50 to about 600 molecules of Table 1, 100 to about 400 molecules of Table 1, 10 to about 100 molecules of Table 1, 20 to about 60 molecules of Table 1, 15 to about 45 molecules of Table 1 or any combination thereof.
  • the pharmaceutical composition can comprise a pharmaceutically acceptable excipient, diluent, or carrier.
  • a pharmaceutical composition can comprise albuterol, beclomethasone, budesonide, formoterol, ciclesonide, fluticasone, fluticasone propionate, fluticasone furoate, fluticasone propionate, salmeterol, fluticasone furonate, umeclidinium, vilanterol, mometasone furoate, memetasone, a steroid, a bronchodilator.
  • a pharmaceutical composition can comprise a solid, a liquid, an aerosol, a gas, or a combination thereof.
  • a composition described herein can compromise an excipient.
  • An excipient can comprise a pH agent (to minimize oxidation or degradation of a component of the composition), a stabilizing agent (to prevent modification or degradation of a component of the composition), a buffering agent (to enhance temperature stability), a solubilizing agent (to increase protein solubility), or any combination thereof.
  • An excipient can comprise a surfactant, a sugar, an amino acid, an antioxidant, a salt, a non-ionic surfactant, a solubilizer, a triglyceride, an alcohol, or any combination thereof.
  • An excipient can comprise sodium carbonate, acetate, citrate, phosphate, poly-ethylene glycol (PEG), human serum albumin
  • HSA sorbitol
  • sucrose trehalose
  • polysorbate 80 sodium phosphate, sucrose, disodium phosphate, mannitol, polysorbate 20, histidine, citrate, albumin, sodium hydroxide, glycine, sodium citrate, trehalose, arginine, sodium acetate, acetate, HC1, disodium edetate, lecithin, glycerin, xanthan rubber, soy isoflavones, polysorbate 80, ethyl alcohol, water, teprenone, or any combination thereof.
  • An excipient can be an excipient described in the Handbook of
  • a carrier can refer to reagents, cells, compounds, materials, compositions, dosage forms, or any combination thereof that can be compatible with a small molecule or other agents that can be administered therapeutically.
  • a carrier can be suitable for use in contact with a tissue of a subject.
  • a carrier may not have a toxicity, an irritation, an allergic response, or any combination thereof.
  • a carrier that may be suitable for use can include a liquid, an aerosol, a semi-solid (e.g., a gel), and a solid material (e.g., a pill, a patch, or an implant).
  • a carrier can be designed to resist degradation within the body (non-biodegradable) or they may be designed to degrade within the body
  • a biodegradable material can further be bioresorbable or bioabsorbable. In some cases, a biodegradable material can be degraded and eliminated from the body by conversion into other materials or breakdown and elimination through natural pathways.
  • a pharmaceutical composition can comprise isoprene, decanal, 4- methylbenzaldehyde, 3,7-dimethyloctan-3-ol, l-methyl-4(l -methyl -2 -propenyl)-benzene, 1- ethenyl-3 -ethyl-benzene, l-ethenyl-4-ethyl-benzene, 1-phenylbut-l-ene, octan-2-one, heptan-
  • a pharmaceutical composition can comprise a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or a salt thereof.
  • the pharmaceutical composition can be inhaled.
  • the pharmaceutical composition can be encapsulated.
  • the pharmaceutical composition can be formulated as a liquid, an aerosol or as a semi-solid, such as a gel, or a solid.
  • the pharmaceutical composition can be formulated at a solution.
  • the pharmaceutical composition can be formulated as an injectable.
  • the pharmaceutical composition can be formulated as a subdermal implant.
  • the pharmaceutical composition can be implantable.
  • the pharmaceutical composition can be formulated for oral delivery.
  • the pharmaceutical composition can be delivered by any method.
  • Administration or application of a composition disclosed herein can be performed for a treatment duration of at least about at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14,
  • a treatment duration can be from about 1 to about 30 days, from about 2 to about 30 days, from about 3 to about 30 days, from about 4 to about 30 days, from about 5 to about 30 days, from about 6 to about 30 days, from about 7 to about 30 days, from about 8 to about 30 days, from about 9 to about 30 days, from about 10 to about 30 days, from about 11 to about 30 days, from about 12 to about 30 days, from about 13 to about 30 days, from about 14 to about 30 days, from about 15 to about 30 days, from about 16 to about 30 days, from about 17 to about 30 days, from about 18 to about 30 days, from about 19 to about 30 days, from about 20 to about 30 days, from about 21 to about 30 days, from about 22 to about 30 days, from about 23 to about 30 days, from about 24 to about 30 days, from about 25 to about
  • 30 days from about 26 to about 30 days, from about 27 to about 30 days, from about 28 to about 30 days, or from about 29 to about 30 days.
  • Administration or application of a composition disclosed herein can be performed for a treatment duration of at least about 1 week, at least about 1 month, at least about 1 year, at least about 2 years, at least about 3 years, at least about 4 years, at least about 5 years, at least about 6 years, at least about 7 years, at least about 8 years, at least about 9 years, at least about 10 years, at least about 15 years, at least about 20 years, or for life.
  • Administration can be performed repeatedly over a lifetime of a subject, such as once a month or once a year for the lifetime of a subject.
  • Administration can be performed repeatedly over a substantial portion of a subject’s life, such as once a month or once a year for at least about 1 year, 5 years, 10 years, 15 years, 20 years, 25 years, 30 years, or more.
  • composition disclosed herein can be performed at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 times a day.
  • administration or application of a composition disclosed herein can be performed continuously throughout an entire day, for example, when an implant can be used for administration.
  • an implant can release a composition continuously for about: 5 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 8 hours, 12 hours, 16 hours, 20 hours, 1 day, 2 days, 4 days, 7 days, 2 weeks, 4 weeks, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year or more.
  • an implant can have noncontinuous administration.
  • administration or application of composition disclosed herein can be performed at least 1, 2,
  • composition disclosed herein can be performed at least 1, 2,
  • a composition can be administered/applied as a single dose or as divided doses.
  • the compositions described herein can be administered at a first time point and a second time point.
  • a composition can be administered such that a first administration can be administered before the other with a difference in administration time of 1 hour, 2 hours, 4 hours, 8 hours, 12 hours, 16 hours, 20 hours, 1 day,
  • a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or a salt thereof when stored in a closed container placed in a room for a time period will remain at least about 80% of an initial amount of the compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or a salt thereof.
  • the compound of Table 1 will remain at least about 70% the initial amount.
  • the compound of Table 1 will remain at least about 90% the initial amount.
  • the compound of Table 1 will remain at least about: 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%.
  • the compound of Table 1 can be at least about 60% to about at least 80%.
  • the compound of Table 1 can be at least about 80% to at least about 99%.
  • the time period of storage can be at least 1 month. In some cases, the time period of storage can be at least about 3 months. In some cases, the time period of storage can be at least about 1 year. In some cases, the time period of storage can be at least about 1, 2, 4, 6, 8, 12, 18, 24, 36, 48 or 60 months. In some cases, the time period of storage can be at least about 1 month to about at least 1 year. In some cases, the time period of storage can be at least about 6 months to at least about 2 years. In some cases, the time period of storage can be at least about 1 month to at least about 5 years. [0075]
  • tissue as used herein, can be any tissue sample.
  • a tissue can be a tissue suspected or confirmed of having a disease or condition.
  • a tissue can be a sample that may be substantially healthy, substantially benign, or otherwise substantially free of a disease or a condition.
  • a tissue can be a tissue removed from a subject, such as a tissue biopsy, a tissue resection, an aspirate (such as a fine needle aspirate), a tissue washing, a cytology specimen, a bodily fluid, or any combination thereof.
  • a tissue can comprise cancerous cells, tumor cells, non-cancerous cells, or a combination thereof.
  • a tissue can comprise a blood sample (such as a cell-free DNA sample).
  • a tissue can be a sample that may be genetically modified.
  • Methods as described herein can be utilized for preventing, diagnosing and treating one or more diseases described herein. Methods can be utilized for preventing one or more diseases. Methods can be utilized for diagnosing one or more diseases. Methods can be utilized for treating, such as at least partially treating or substantially reducing one or more symptoms of one or more diseases.
  • a disease can comprise a cancer, a tumor, an infectious disease, a skeletal disease, a urogenital disease, a digestive disease, a muscular disease, a neurological disease, an autoimmune disease, a respiratory disease, a cardiac disease, an endocrine disease, a liver disease, a hereditary disease, or any combination thereof.
  • a disease or condition can comprise a cancer.
  • a cancer may comprise, a sarcoma, a carcinoma, a melanoma, a lymphoma, leukemia, blastoma, germ cell tumor, myeloma, or any combination thereof.
  • cancer may comprise a thyroid cancer, adrenal cortical cancer, anal cancer, aplastic anemia, bile duct cancer, bladder cancer, bone cancer, bone metastasis, central nervous system (CNS) cancers, peripheral nervous system (PNS) cancers, breast cancer, Castleman's disease, cervical cancer, childhood Non-Hodgkin's lymphoma, lymphoma, colon and rectum cancer, endometrial cancer, esophagus cancer, Ewing's family of tumors (e.g.
  • Ewing's sarcoma eye cancer, gallbladder cancer, gastrointestinal carcinoid tumors, gastrointestinal stromal tumors, gestational trophoblastic disease, hairy cell leukemia, Hodgkin's disease, Kaposi's sarcoma, kidney cancer, laryngeal and hypopharyngeal cancer, acute lymphocytic leukemia, acute myeloid leukemia, children's leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, liver cancer, lung cancer, lung carcinoid tumors, Non-Hodgkin's lymphoma, male breast cancer, malignant mesothelioma, multiple myeloma, myelodysplastic syndrome, myeloproliferative disorders, nasal cavity and paranasal cancer, nasopharyngeal cancer, neuroblastoma, oral cavity and oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, penile cancer
  • a condition or a disease can include hyperproliferative disorders.
  • Malignant hyperproliferative disorders can be stratified into risk groups, such as a low risk group and a medium-to-high risk group.
  • Hyperproliferative disorders can include but may not be limited to cancers, hyperplasia, or neoplasia.
  • the hyperproliferative cancer can be breast cancer such as a ductal carcinoma in duct tissue of a mammary gland, medullary carcinomas, colloid carcinomas, tubular carcinomas, and inflammatory breast cancer; ovarian cancer, including epithelial ovarian tumors such as adenocarcinoma in the ovary and an adenocarcinoma that has migrated from the ovary into the abdominal cavity; uterine cancer; cervical cancer such as adenocarcinoma in the cervix epithelial including squamous cell carcinoma and adenocarcinomas; prostate cancer, such as a prostate cancer selected from the following: an adenocarcinoma or an adenocarcinoma that has mi-grated to the bone; pancreatic cancer such as epithelioid carcinoma in the pancreatic duct tissue and an adenocarcinoma in a pancreatic duct; bladder cancer such as a transitional cell carcinoma in urinary bladder,
  • AIDS-related lympho-ma such as diffuse large B-cell lymphoma, B-cell immunoblastic lymphoma and small non-cleaved cell lymphoma; Kaposi's Sarcoma; viral-induced cancers including hepatitis B virus (HBV), hepatitis C virus (HCV),and hepatocellular carcinoma; human lymphotropic virus-type 1 (HTLV-1) and adult T-cell leukemia/lymphoma; and human papilloma virus (HPV) and cervical cancer; central nervous system (CNS) cancers such as primary brain tumor, which includes gliomas (astrocytoma, anaplastic astrocytoma, or glioblastoma multiforme), oligodendrogliomas, ependymomas, meningiomas, lymphomas, schwannomas, and medulloblastomas; peripheral nervous system (PNS) cancers such as acoustic neuromas and malignant peripheral nerve sheath tumors (
  • Miillerian tumors oral cavity and oropharyngeal cancer such as hypopharyngeal cancer, laryngeal cancer, nasopharyngeal cancer, and oropharyngeal cancer; stomach cancer such as lymphomas, gastric stromal tumors, and carcinoid tumors; testicular cancer such as germ cell tumors (GCTs), which include seminomas and nonseminomas, and gonadal stromal tumors, which include Leydig cell tumors and Sertoli cell tumors; thymus cancer such as to thymomas, thymic carcinomas, Hodgkin disease, non-Hodgkin lymphomas carcinoids or carcinoid tumors; rectal cancer; and colon cancer.
  • GCTs germ cell tumors
  • thymus cancer such as to thymomas, thymic carcinomas, Hodgkin disease, non-Hodgkin lymphomas carcinoids or carcinoid tumors
  • rectal cancer and
  • the diseases stratified, classified, characterized, or diagnosed by the methods of the present disclosure include but may not be limited to thyroid disorders such as for example benign thyroid disorders including but not limited to follicular adenomas, Hurthle cell adenomas, lymphocytic thyroiditis, and thyroid hyperplasia.
  • the diseases stratified, classified, characterized, or diagnosed by the methods of the present disclosure include but may not be limited to malignant thyroid disorders such as for example follicular carcinomas, follicular variant of papillary thyroid carcinomas, medullary carcinomas, and papillary carcinomas.
  • urogenital disease can comprise benign prostate hyperplasia, urinary incontinence, kidney stones, kidney disease, erectile dysfunction, interstitial cystitis, prostatitis, overactive bladder, endometriosis, hypospadias, interstitial cystitis, posterior urethral valve, undescended testes, ureteropelvic junction obstruction, vesicoureteral reflux, or any combination thereof.
  • a digestive disease can comprise irritable bowel syndrome, celiac disease, gastroesophageal reflux disease, Crohn’s disease, gastroparesis, gas, lactose intolerance, diarrhea, acid reflux, constipation, Hirschsprung disease, celiac disease, gallstones, abdominal adhesions, Barrett’s esophagus, bowl control problems, colon polyps, diverticulosis, food poisoning, gastritis, gastroparesis, GI bleeding, hemorrhoids, indigestion, hernia, microscopic colitis, pancreatitis, peptic ulcers, proctitis, short bowel syndrome, ulcerative colitis, Whipple disease, Zollinger-Ellison syndrome, or any combination thereof.
  • a respiratory disease can comprise asthma, chronic obstructive pulmonary disease, chronic bronchitis, emphysema, cystic fibrosis, plural effusion, pneumonia, obstructive condition, alpha- 1 antitrypsin deficiency, bronchiectasis, pulmonary fibrosis, sarcoidosis, or any combination thereof.
  • a cardiac disease can comprise congenital heart disease, arrhythmia, coronary artery disease, cardiac arrest, peripheral artery disease, dilated cardiomyopathy, myocardial infarction, stroke, structural heart disease, congestive heart failure, high blood pressure, mitral regurgitation, pulmonary stenosis, or any combination thereof.
  • an endocrine disease can comprise type 2 diabetes, hypothyroidism, hyperthyroidism, low testosterone, obesity, Addison’s disease, Cushing’s syndrome, Graves’ disease, thyroiditis, prolactinoma, type 1 diabetes, type 2 diabetes, osteoporosis, thyroid cancer, gestational diabetes, growth hormone deficiency, Hashimoto’s thyroiditis, hyperglycemia, hyperparathyroidism, hypoglycemia, hypoparathyroidism, hypothyroidism, menopause, obesity, pre-diabetes, thyroid nodules, or any combination thereof.
  • a liver disease can comprise cirrhosis, fibrosis, hemochromatosis, Wilson’s disease, Alagille syndrome, alcohol -related liver disease, alpha-1 antitrypsin deficiency, autoimmune hepatitis, benign liver tumor, biliary atresia, Crigler- Najjar Syndrome, Galactosemia, Gilbert Syndrome, Hemochromatosis, Hepatic Encephalopathy, Hepatitis A, Hepatitis B, Hepatitis C, hepatorenal syndrome, intrahepatic cholestasis of pregnancy, lysosomal acid lipase deficiency, liver cysts, newborn jaundice, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, primary biliary cholangitis, primary sclerosing cholangitis, progressive familial intrahepatic cholestasis, Reye syndrome, type 1 glycogen storage disease or any combination thereof.
  • a disease or condition can comprise a skeletal disease can comprise osteoarthritis, osteoporosis, Pagets disease of bone, back pain, bursitis, fibromyalgia, fibrous dysplasia, growth plate injuries, heritable disorders of connective tissue, osteonecrosis, scoliosis, shoulder problems, hip problems, knee problems, joint problems, spinal stenosis, tendinitis, bone breaks, bone spurs, osteopetrosis, or any combination thereof.
  • a disease or condition can comprise a neurological disorder.
  • a neurological disorder may comprise Acquired Epileptiform Aphasia, Acute
  • Alzheimer's disease Amyotrophic lateral sclerosis (see Motor Neuron Disease)
  • Blepharo-spasm Bloch-Sulzberger syndrome, Brachial plexus injury, Brain abscess, Brain damage, Brain in-jury, Brain tumor, Brown-Sequard syndrome, Canavan disease, Carpal tunnel syndrome (CTS), Causalgia, Central pain syndrome, Central pontine myelinolysis,
  • Centronuclear myopathy Cephalic disorder, Cerebral aneurysm, Cerebral arteriosclerosis,
  • Cerebral atrophy Cerebral gigantism
  • Cerebral palsy Charcot-Marie-Tooth disease
  • Chiari malformation Cerebral malformation
  • Chorea Chronic inflammatory de-myelinating polyneuropathy (CIDP)
  • Chronic pain Chronic regional pain syndrome, Coffin Lowry syndrome, Coma, including
  • Dermatomyositis Neurological Dyspraxia, Diabetic neuropathy, Diffuse sclerosis, Dysautonomia, Dyscalculia, Dysgraphia, Dyslexia, Dystonia, , Early infantile epileptic encephalopathy, Empty sella syndrome, Encephalitis, Encephalocele, Encephalotrigeminal angiomatosis, Encopresis, Epilepsy, Erb's palsy, Erythromelalgia, Essential tremor, Fabry's disease, Fahr's syndrome, Fainting, Familial spastic paralysis, Febrile seizures, Fisher syndrome, Friedreich's ataxia, FART Syndrome, Gaucher's disease, Gerstmann's syndrome,
  • Giant cell arteritis Giant cell inclusion disease, Globoid cell Leukodystrophy, Gray matter heterotopia, Guillain-Barre syndrome, HTLV-1 associated myelopathy, Hallervorden-Spatz disease, Head injury, Headache, Hemifacial Spasm, Hereditary Spastic Paraplegia,
  • Neuron Disease Motor skills disorder, Moyamoya disease, Mucopolysaccharidoses, Multi-
  • Muscular dystrophy Myalgic encephalomyelitis, Myasthenia gravis, Myelinoclastic diffuse sclerosis, Myoclonic Encephalopathy of infants, Myoclonus, Myopathy, Myotubular myopathy, Myotonia congenita, Narcolepsy, Neurofibromatosis, Neuroleptic malignant syndrome, Neurological manifestations of AIDS, Neurological sequelae of lupus,
  • Neuromyotonia Neuronal ceroid lipofuscinosis, Neuronal migration disorders, Niemann-Pick disease, Non 24-hour sleep-wake syndrome, Nonverbal learning disorder, O'Sullivan
  • McLeod syndrome Occipital Neuralgia, Occult Spinal Dysraphism Sequence, Ohtahara syndrome, Olivopontocerebellar atrophy, Opsoclonus myoclonus syndrome, Optic neuritis,
  • Paramyotonia Con-genita Paraneoplastic diseases, Paroxysmal attacks, Parry-Romberg syndrome, Rombergs Syndrome, Pelizaeus-Merzbacher disease, Periodic Paralyses,
  • Peripheral neuropathy Persistent Vegetative State, Pervasive neurological disorders, Photic sneeze reflex, Phytanic Acid Storage disease, Pick's disease, Pinched Nerve, Pituitary
  • Prader-Willi syndrome Primary Lateral Sclerosis, Prion diseases, Progressive Hemifacial
  • Atrophy also known as Rombergs Syndrome, Progressive multifocal leukoencephalopathy,
  • Septooptic dysplasia Shaken baby syndrome, Shingles, Shy-Drager syndrome, Sjogren's syndrome, Sleep apnea, Sleeping sickness, Snatiation, Sotos syndrome, Spasticity, Spina bifida, Spinal cord injury, Spinal cord tumors, Spinal muscular atrophy, Spinal stenosis,
  • a disease or condition may comprise an autoimmune disease.
  • an autoimmune disease may comprise acute disseminated encephalomyelitis (ADEM), acute necrotizing hemorrhagic leukoencephalitis, Addison's disease, agammaglobulinemia, allergic asthma, allergic rhinitis, alopecia areata, amyloidosis, ankylosing spondylitis, anti- GBM/anti-TBM nephritis, antiphospholipid syndrome (APS), autoimmune aplastic anemia, autoimmune dysautonomia, autoimmune hepatitis, autoimmune hyperlipidemia, autoimmune immunodeficiency, autoimmune inner ear disease (AIED), autoimmune myocarditis, autoimmune pancreatitis, autoimmune retinopathy, autoimmune thrombocytopenic purpura (ATP), autoimmune thyroid disease, axonal & neuronal neuropathies, Balo disease, Bechet’s disease, bullous pepomyelitis, agamm
  • Devic's disease (neuromyelitis optica), discoid lupus, Dressler's syndrome, endometriosis, eosinophillic fasciitis, erythema nodosum, experimental allergic encephalomyelitis, Evan's syndrome, fibromyalgia, fibrosing alveolitis, giant cell arteritis (temporal arteritis), glomerulonephritis, Goodpasture's syn-drome, Grave's disease, Guillain-Barre syndrome,
  • Hashimoto's encephalitis Hashimoto's thyroiditis, hemolytic anemia, Henock-Schoniein purpura, herpes gestationis, hypogammaglobulinemia, idiopathic thrombocytopenic purpura
  • IGP insulin-dependent diabetes
  • type 1 insulin- dependent diabetes
  • interstitial cystitis juvenile arthritis
  • juvenile diabetes Kawasaki syndrome
  • Lambert-Eaton syndrome leukocytoclastic vasculitis
  • lichen planus lichen sclerosis
  • CLI linear IgA disease
  • SLE Lupus
  • MCTD mixed connective tissue disease
  • autoimmune polyglandular syndromes polymyalgia rheumatic, polymyositis, postmyocardial infarction syndrome, postpericardiotomy syndrome, progesterone dermatitis, primary biliary cirrhosis, primary sclerosing cholangitis, psoriasis, psoriatic arthritis, idiopathic pulmonary fibrosis, pyoderma gangrenosum, pure red cell aplasis, Raynaud's phenomena, reflex sympathetic dystrophy, Reiter's syndrome, relapsing polychondritis, restless legs syndrome, retroperitoneal fibrosis, rheumatic fever, rheumatoid arthritis, sarcoidosis, Schmidt syn-drome, scleritis, scleroderma, Slogren's syndrome, sperm and testicular autoimmunity, stiff person syndrome, subacute bacterial endocarditis (SBE), sympathetic o
  • cystic fibrosis vasculitides acquired hypoparathyroidism, coronary artery disease, pemphigus foliaceus, pemphigus vulgaris, Rasmussen encephalitis, autoimmune gastritis, insulin hypoglycemic syndrome (Hirata disease), Type B insulin resistance, acanthosis, systemic lupus erythematosus (SLE), pernicious anemia, treatment-resistant Lyme arthritis, polyneuropathy, demyelinating diseases, atopic dermatitis, autoimmune hypothyroidism, vitiligo, thyroid associated ophthalmopathy, autoimmune coeliac disease, ACTH deficiency, dermatomyositis, Sjogren syndrome, systemic sclerosis, progressive systemic sclerosis, morphea, primary antiphospholipid syndrome, chronic idiopathic urticaria, connective tissue syndromes, necrotizing and crescentic glomerulonephritis (NCGN), systemic vascu
  • Raynaud syndrome chronic liver disease, visceral leishmaniasis, autoimmune Cl deficiency, membrane proliferative glomerulonephritis (MPGN), prolonged coagulation time, immunodeficiency, atherosclerosis, neuronopathy, paraneoplastic pemphigus, paraneoplastic stiff man syndrome, paraneoplastic encephalomyelitis, subacute autonomic neuropathy, cancer-associated retinopathy, paraneoplastic opsoclonus myoclonus ataxia, lower motor neuron syndrome and Lambert-Eaton myasthenic syndrome.
  • MPGN membrane proliferative glomerulonephritis
  • a disease or a condition may comprise an infectious disease.
  • an infectious disease can comprise a viral infection, a bacterial infection, a parasitic infection, a fungal infection or any combination thereof.
  • an infectious disease can comprise, AIDS, anthrax, botulism, brucellosis, chancroid, chlamydial infection, cholera, coccidioidomycosis, cryptosporidiosis, cyclosporiasis, dipheheria, ehrlichiosis, arboviral encephalitis, enterohemorrhagic Helicobacter pylori, Staphylococcus aureus, Mycobacterium tuberculosis, Escherichia coli, Campylobacter jejuni, Salmonella, Klebsiella pneumonia,
  • Streptococcus pyogenes Neisseria gonorrhoeae, Treponema pallidum, Borrelia burgdorferi,
  • Neisseria meningitidis Chlamydia trachomatis, Enterobacteriaceae, Vibrio sp., giardiasis,
  • Enterobius vermicularis Ancylostoma duodenale, Necator americanus, Entamoeba histolytica, Trypanosoma cruzi, Cyclospora cayetanenensis, Haemophilus influenza, Hansen's disease (Leprosy), trichomoniasis, ascariasis, taenia infection, lymphatic filariasis, African sleeping sickness, onchocerciasis, leishmaniasis, fungal nail infections, vaginal candidiasis, ringworm, Candida infections, blastomycosis, coccidioidomycosis, Cryptococcus gattii,
  • Cryptococcus neoformans pneumoncystis pneumona, talaromycosis, mucormycosis, fungal eye infection, mycetoma, sporotrichosis, Tinea versicolor, ringworm, hantavirus pulmonary syndrome, hemolytic uremic syndrome, hepatitis A, hepatitis B, hepatitis C, human immunodeficiency virus (HIV), legionellosis, listeriosis, Lyme disease, malaria, measles.
  • HIV human immunodeficiency virus
  • Meningococcal disease Meningococcal disease, mumps, pertussis (whooping cough), plague, paralytic poliomyelitis, psittacosis, Q fever, rabies, rocky mountain spotted fever, rubella, congenital rubella syndrome , shigellosis, smallpox, rotavirus, norovirus, streptococcal disease (invasive group
  • streptococcal toxic shock syndrome streptococcus pneumonia, syphilis, tetanus, toxic shock syndrome, trichinosis, tuberculosis, tularemia, typhoid fever, Chikungunya virus, varicella, yellow fever, variant Creutzfeldt-Jakob disease (vCJD), Ebola hemorrhagic fever,
  • Echinococcosis enterovirus, herpes simplex 1, herpes simplex 2, varicella zoster virus, dengue virus, Hendra virus, Nipah virus, human monkey-pox, influenza, influenza A, influenza B, H5N1, lassa fever, Margurg hemorrhagic fever, Nipah virus, O'nyong fever, Rift valley fever, Herpes, SARS-CoV-2 (COVID-19), SARS-CoV (SARS), MERS-CoV (MERS),
  • an infectious disease can be a drug resistant infection. In some cases, an infectious disease can be a multi-drug resistant infection.
  • Conditions or diseases of the present disclosure can include a genetic disorder.
  • a genetic disorder can be a hereditary disease.
  • a heredity disease can be inherited mendelian disorders, peroxisomal disorders, mitochondrial disorders, cystic fibrosis, sickle-cell anemia, Marfan syndrome, Huntington’s disease, kidney disease, Tay-Sachs disease, phenylketonuria, hemophilia, or any combination thereof.
  • a genetic disorder may be an illness caused by abnormalities in genes or chromosomes. Genetic disorders can be grouped into two categories: single gene disorders and multifactorial and polygenic (complex) disorders. A single gene disorder can be the result of a single mutated gene.
  • Inheriting a single gene disorder can include but not be limited to autosomal dominant, autosomal recessive, X-linked dominant, X-linked recessive, Y-linked and mitochondrial inheritance.
  • one mutated copy of the gene can be necessary for a person to be affected by an autosomal dominant disorder.
  • autosomal dominant type of disorder can include but are not be limited to Huntington's disease, Neurofibromatosis 1, Marfan Syndrome, Hereditary nonpolyposis colorectal cancer, or Hereditary multiple exostoses.
  • two copies of the gene can be mutated for a subject to be affected by an autosomal recessive disorder.
  • Examples of this type of disorder can include but may not be limited to cystic fibrosis, sickle-cell disease (also partial sickle cell disease), Tay-Sachs disease, Niemann-Pick disease, or spinal muscular atrophy.
  • X-linked dominant disorders are caused by mutations in genes on the X chromosome such as X-linked hypophosphatemic rickets. Some X-linked dominant conditions such as Rett syndrome, Incontinentia Pigmenti type 2 and Aicardi Syndrome can be fatal.
  • X-linked recessive disorders are also caused by mutations in genes on the X chromosome.
  • Examples of this type of disorder can include but are not limited to Hemophilia A, Duchenne muscular dystrophy, red-green colorblindness, muscular dystrophy and Androgenetic alopecia.
  • Y-linked disorders are caused by mutations on the Y chromosome. Examples can include but are not limited to
  • the genetic disorder of mitochondrial inheritance also known as maternal inheritance, can apply to genes in mitochondrial DNA such as in
  • Genetic disorders may also be complex, multifactorial or polygenic.
  • Polygenic genetic disorders can be associated with the effects of multiple genes in combination with lifestyle and environmental factors. Although complex genetic disorders can cluster in families, they do not have a clear-cut pattern of inheritance.
  • Multifactorial or polygenic disorders can include heart disease, diabetes, asthma, autism, autoimmune diseases such as multiple sclerosis, cancers, ciliopathies, cleft palate, hypertension, inflammatory bowel disease, mental retardation or obesity.
  • Other genetic disorders can include but may not be limited to lp36 deletion syndrome, 21 -hydroxylase deficiency, 22qll.2 deletion syndrome, aceruloplasminemia, achondrogenesis, type II, achondroplasia, acute intermittent porphyria, adenylosuccinate lyase deficiency, Adrenoleu-kodystrophy, Alexander disease, alkaptonuria, alpha- 1 antitrypsin deficiency, Alstrom syndrome, Alzheimer's disease (type 1, 2, 3, and 4), Amelogenesis Imperfecta, amyotrophic lateral sclerosis, Amyotrophic lateral sclerosis type 2, Amyotrophic lateral sclerosis type 4, amyotrophic lateral sclerosis type 4, androgen insensitivity syndrome, Anemia, Angelman syndrome, Apert syndrome, ataxia-telangiectasia, Beare-Stevenson cutis gyrata syndrome, Benjamin syndrome, beta thalassemia, biotimidase defic
  • Congenital heart disease congenital hypothyroidism
  • Connective tissue disease Cowden syndrome
  • Cri du chat syndrome Crohn's disease
  • fibrostenosing Crouzon syndrome
  • Danlos syndrome arthrochalasia type Ehlers-Danlos syndrome classical type, Ehlers-Danlos syndrome dermatosparaxis type, Ehlers-Danlos syndrome kyphoscoliosis type, vascular type, erythropoietic protoporphyria, Fabry's disease, Facial injuries and disorders, factor V Leiden thrombophilia, familial adenomatous polyposis, familial dysautonomia, fanconi anemia, FG syndrome, fragile X syndrome, Friedreich ataxia, Friedreich's ataxia, G6PD deficiency, galactosemia, Gaucher's disease (type 1, 2, and 3), Genetic brain disorders, Glycine encephalopathy, Haemochromatosis type 2, Haemochromatosis type 4, Harlequin Ichthyosis,
  • Kennedy disease Klinefelter syndrome, Kniest dysplasia, Krabbe disease, Learning disability, Lesch-Nyhan syndrome, Leukodystrophies, Li-Fraumeni syndrome, lipoprotein lipase deficiency, familial, Male genital disorders, Marfan syndrome, McCune- Albright syndrome, McLeod syndrome, Mediterranean fever, familial, Menkes disease, Menkes syndrome, Metabolic disorders, methemoglobinemia beta-globin type, Methemoglobinemia congenital methaemoglobinaemia, methylmalonic acidemia, Micro syndrome, Microcephaly,
  • Movement disorders Mowat-Wilson syndrome, Mucopolysaccharidosis (MPS I), Muenke syndrome, Muscular dystrophy, Muscular dystrophy, Duchenne and Becker type, muscular dystrophy, Duchenne and Becker types, myotonic dystrophy, Myotonic dystrophy type 1 and type 2, Neonatal hemochromatosis, neurofibromatosis, neurofibromatosis 1, neurofibromatosis 2, Neurofibromatosis type I, neurofibromatosis type II, Neurologic diseases, Neuromuscular disorders, Niemann-Pick disease, Nonketotic hyperglycinemia, nonsyndromic deafness, Nonsyndromic deafness autosomal recessive, Noonan syn-drome, osteogenesis imperfecta (type I and type III), otospondylomegaepiphyseal dysplasia, pantothenate kinase-associated neurodegeneration, Patau Syndrome (Trisomy 13), Pendred syndrome,
  • Gaucher disease pseudoxanthoma elasticum, Retinal disorders, retinoblastoma, retinoblastoma FA — Friedreich ataxia, Rett syndrome, Rubinstein-Taybi syndrome, Sandhoff disease, sensory and autonomic neuropathy type III, sickle cell anemia, skeletal muscle regeneration, Skin pigmentation disorders, Smith Lemli Opitz Syn-drome, Speech and communication disorders, spinal muscular atrophy, spinal-bulbar muscular atrophy, spinocerebellar ataxia, spondyloepimetaphyseal dysplasia, Strudwick type, spondyloepiphyseal dysplasia congenita, Stickler syndrome, Stickler syndrome COL2A1,
  • Tay-Sachs disease tetrahydrobiopterin deficiency, thanatophoric dysplasia, thiamine- responsive megaloblastic anemia with diabetes mellitus and sensorineural deafness, Thyroid disease, Tourette's Syndrome, Treacher Collins syndrome, triple X syndrome, tuberous sclerosis, Turner syndrome, Usher syndrome, variegate porphyria, von Hippel-Lindau disease, Waardenburg syndrome, Weissenbacher-Zweymiiller syndrome, Wilson disease, Wolf-Hirschhom syndrome, Xeroderma Pigmentosum, X-linked severe combined immunodeficiency, X-linked sideroblastic anemia, or X-linked spinal-bulbar muscle atrophy.
  • compositions and methods disclosed herein for treating or preventing a disease or a condition can comprise administering to a subject a therapeutically effective amount of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • a portion of a plant, an extract thereof, an herb, a portion of an herb, an extract thereof, or any combination thereof can be administered with a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • a plant or an herb can comprise ginseng, ginkgo, mushrooms, wolfberry, dang gui, astragalus, atractylodes, bupleurum, cinnamon, coptis chinensis, ginger, licorice, ephedra, peon, rehmania, rubarb, salvia, cannabis, mint, velvet leaf, Chinese goldthread, poppy, prgining croton, lilac daphne, devil’s trumpet, jimson weed, noble dendrobium, blue evergreen, kudzu, snakeroot, foxglove, stephania root, skullcap, pagoda tree, Indian stringbush or any combination thereof.
  • a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be increased or decreased in a subject by altering a composition of a food, a beverage, a nutrient, or any combination thereof. In some cases, the increase or decrease can achieved by altering the diet of a subject, administering a supplement, or by adding exercise routine.
  • a method can comprise detecting a presence and a level of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof, from a sample of a subject may have or may be suspected of having a disease or condition and comparing the level of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof, to a reference level; or detecting the presence, the levels, the elution times, of a plurality of compounds of
  • the presence and the level can be a concentration.
  • the elution time can be in seconds, in minutes, in hours or a combination thereof.
  • the reference pattern can be compiled from a single patient result or a plurality of patient results.
  • the reference pattern can be obtained from a lab sample, a clinical sample, or a reference sample.
  • the reference pattern can be one compound or a plurality of compounds.
  • a level can indicate a viral infection, a bacterial infection, a fungal infection, a parasitic infection, a cancer, a tumor, an infectious disease, a skeletal disease, a urogenital disease, a digestive disease, a muscular disease, a neurological disease, an autoimmune disease, a respiratory disease, a cardiac disease, an endocrine disease, a liver disease, a hereditary disease or any combination thereof.
  • the method can be a method of diagnosing a disease or a condition and further can comprise selecting a therapeutic regiment for treating the disease or the condition.
  • the therapeutic regiment can comprise selecting a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof to treat the disease or condition in a subject.
  • the method can comprise detecting a presence and a level of a first compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, a biomolecule of Table 2 or any combination thereof; a presence and a level of a second compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, a biomolecule of Table 2 or any combination thereof; a presence and a level of a third compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, a biomolecule of Table 2 or any combination thereof; and comparing the first level to a first reference level, the second level to a second reference level, and the third level to
  • the method can comprise detecting the presence and level of less than about: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17,
  • a method can be conducted at least in part on a diagnostic device, a screening device, or a diagnostic and a screening device comprising at least one of; a chromatography column, an eluant, a detector, and a mass spectrometer.
  • a chromatography column can comprise a gas chromatography column, a liquid chromatography column or any combination thereof and the eluant can comprise, a gas, a liquid or any combination thereof.
  • a gas can be helium, hydrogen, or nitrogen.
  • the chromatograph column can comprise a paper, a silica gel, a cellulose, an aluminum oxide, or any combination thereof.
  • a diagnostic device, a screening device, or a diagnostic and a screening device can comprise a microelectromechanical system (MEMS), a nanoelectromechanical systems (NEMS), or any combination thereof.
  • MEMS and NEMS device can comprise a central unit to process data, a microsensor, and an energy source.
  • a diagnostic device, a screening device, or a diagnostic and a screening device can comprise a breath collection tube, a sample collection port, or any combination thereof.
  • the breath collection tube can be more than about: 0.1 meters, 0.2 meters, 0.3 meters, 0.4 meters, 0.5 meters, 0.6 meters, 0.7 meters, 0.8 meters, 0.9 meters, 1.0 meters, 1.5 meters, 2 meters. 2.5 meters. 3 meters. 3.5 meters or 4 meters. In some cases, the breath collection tube can be less than about: 0.1 meters, 0.2 meters, 0.3 meters, 0.4 meters, 0.5 meters, 0.6 meters, 0.7 meters,
  • a diagnostic device, a screening device, or a diagnostic and a screening device may have: a positive volume of less than about 16 cubic feet, a method of transport, wherein the device can be moved by a robot, a drone, or a remote-control transport machine; a self-contained power supply, or any combination thereof.
  • a diagnostic device, a screening device, or a diagnostic and a screening device may have a volume of less than about: 25 cubic feet, 24 cubic feet, 23 cubic feet, 22 cubic feet, 21 cubic feet, 20 cubic feet, 19 cubic feet, 18 cubic feet, 17 cubic feet, 16 cubic feet, 15 cubic feet, 14 cubic feet, 13 cubic feet, 12 cubic feet, 11 cubic feet, 10 cubic feet, 9 cubic feet, 8 cubic feet, 7 cubic feet, 6 cubic feet, 5 cubic feet, 4 cubic feet, 3 cubic feet, 2 cubic feet, 1 cubic foot, or 0.5 cubic foot.
  • a robot, a drone, or a remote-control transport machine can be controlled autonomously.
  • a diagnostic device, a screening device, or a diagnostic and screening device can link wirelessly or directly to a processing station.
  • a wireless connection can be a Wi-Fi connection, a broadband cellular connection, a Bluetooth® connection, or any combination thereof.
  • a processing station can analyze the data remotely.
  • the device can have a length of about: 65 cm, 64 cm, 63 cm, 62 cm, 61 cm, 60 cm, 59 cm, 58 cm, 57 cm, 56 cm, 55 cm, 54 cm, 53 cm, 52 cm,
  • the device can have a width of about: 45 cm, 44 cm, 43 cm, 42 cm, 41 cm, 40 cm, 39 cm, 38 cm, 37 cm, 36 cm, 35 cm, 34 cm, 33 cm, 32 cm, 31 cm, 30 cm, 29 cm, 28 cm, 27 cm, 26 cm, 25 cm, 24 cm, 23 cm, 22 cm, 21 cm, or 20 cm.
  • the device can have a height of about: 40 cm, 39 cm, 38 cm, 37 cm, 36 cm, 35 cm, 34 cm, 33 cm, 32 cm, 31 cm, 30 cm, 29 cm, 28 cm, 27 cm, 26 cm, 25 cm, 24 cm, 23 cm, 22 cm, 21 cm, or 20 cm, 19 cm, 18 cm, 17 cm, 16 cm, or 15 cm.
  • a device can have a length of less than about: 16 cm, 15, cm, 14 cm, 13 cm, 12 cm, 11 cm, 10 cm, 9 cm, 8 cm, 7 cm, or 6 cm.
  • a device can have a width of less than about: 11 cm, 10 cm, 9 cm, 8 cm, 7 cm, 6 cm, 5 cm, or 4 cm.
  • a device can have a height of less than about: 6 cm, 5 cm, 4 cm, 3 cm, 2 cm, 1 cm or 0.5 cm.
  • the method can be used to identify a disease or condition, wherein the sample can be taken from a subject and applied to a device.
  • a computer can be used to at least partially compare a chromatogram in a subject having a disease to a chromatogram of a subject not having the disease, and determining independently the difference in the presence, the absence, the level, or any combination thereof of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, a biomolecule from Table 2 or any combination thereof.
  • the method can be used to identify a disease or condition, wherein the sample can be taken from a subject and applied to a device.
  • a computer can be used to at least partially compare data (e.g., measured or recorded data that may or may not be normalized to a control, e.g., in the form of a chromatogram, a heat map, or a scalogram) in a subject having a disease to data (e.g., measured or recorded data that may or may not be normalized to a control, e.g., in the form of a chromatogram, a heat map, or scalogram) from a subject not having the disease, and determining independently the difference in the presence, the absence, the level, or any combination thereof of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, a biomolecule from Table 2 or any combination thereof.
  • a heat map may be generated during analysis of data produced from embodiments described herein, e.g., which can comprise the use of MEMS.
  • a heat map can be used for analysis of data measured or recorded from embodiments described herein, e.g., which can comprise the use of MEMS, for example, to determine the presence of a condition such as a cancer in a subject, the risk of a condition such as cancer in a subject, or a prognosis of a subject..
  • a chromatogram or scalogram can be generated during analysis of data produced from embodiments described herein, e.g., which can comprise the use of GCMS or MEMS.
  • a chromatogram or scalogram can be used for analysis of data measured or recorded from embodiments described herein, e.g., which can comprise the use of GCMS or
  • the chromatogram can comprise a signature of a disease or a condition. In some cases, the chromatogram can comprise a signature of more than one disease or condition. In some cases, the signature can be compiled from a single patient result or a plurality of patient results. In some cases, the signature can be obtained from a lab sample, a clinical sample, or a reference sample. In some cases, the signature can be a pattern. In some cases, the pattern can be one compound or a plurality of compounds. In some cases, the pattern can be one biomolecule or a plurality of biomolecules. In some cases, the pattern can comprise both biomolecules and compounds.
  • the method may include using an algorithm.
  • the algorithm may be employed to perform one or more comparisons of the method, such as comparing one or more chromatograms.
  • the algorithm may be a supervised learning algorithm, a trained algorithm, a machine learning algorithm or other.
  • the algorithm may comprise unsupervised learning algorithm.
  • the algorithm may comprise a hierarchical clustering. Examples of algorithms may include support vector machine (SVM) algorithm, a random forest algorithm, a logistical regression algorithm, a neural network, a decision tree algorithm, a naive bayes algorithm, nearest neighbor algorithm, or any combination thereof.
  • SVM support vector machine
  • the method may not comprise use of an algorithm.
  • a portion of the method may employ use of an algorithm.
  • an algorithm can use a processor or a microprocessor.
  • a computer can comprise a memory, one or more executable instructions, one or more processors and a graphical user interface.
  • a result can be communicated via a communication medium.
  • a communication medium can be a screen, an audio description, a physical readout, or any combination thereof.
  • a communication medium can be the internet, a private server, a cell phone, a computer, a television, a hologram, a smart device, a speaker, a printer, or any combination thereof.
  • the compound of Table 1 identified by the method, or the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof can be selected as a therapeutic
  • a biomolecule of Table 2 can be identified by the method and can be selected as a therapeutic.
  • a compound from Table 1 and a biomolecule from Table 2 can be identified by the method.
  • the method can include diagnosing a presence or an absence of a disease or condition in a subject.
  • the method can include a prognosis of a disease or condition in a subject, such as a prognosis prior to, during, or following administration of one or more pharmaceutical compositions.
  • the method can include a theranostic method, such as administration of a first component to a subject to diagnose a disease or condition and administration of a second component to the subject to at least partially treat or reduce one or more symptom of the disease or condition.
  • the method can include measuring a health level information, such as a heart rate, a blood pressure, a body temperature, an oxygen saturation, a protein, a chemokine or a hormone level, a medical history, or any combination thereof.
  • a health level information such as a heart rate, a blood pressure, a body temperature, an oxygen saturation, a protein, a chemokine or a hormone level, a medical history, or any combination thereof.
  • the subject may be symptomatic for the disease or condition.
  • the subject may be asymptomatic for the disease or condition.
  • the subject may have received a previous diagnosis for the disease or condition.
  • the subject may be suspected of having the disease or condition.
  • the subject can display an altered profile, a level of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, the biomolecule of Table 2 or any combination thereof may be at least partially increased or at least partially decreased.
  • the altered profile can be visually represented as chromatogram.
  • a measurement and/or an analysis (e.g., comprising a measurement and/or analysis of a healthy subject’s metabolic profile or a metabolic profile of a subject having a condition such as cancer, wherein either profile can include VOC) can be made one or more times over a period of 1 minute, 2 minutes, 3 minutes, 5 minutes, 10 minutes, 15 minutes, 20 minutes, 30 minutes, 1 hour, 2 hours, 6 hours, 12 hours, 24 hours, 2 days, 5 days, 1 week, 2 weeks, 1 month, 2 months, 6 months, 1 year, 2 years, 3 years, 5 years, 10 years, 20 years, less than 1 minute, from 1 minute to 2 minutes, from 2 minutes to 3 minutes, from 3 minutes to 5 minutes, from 5 minutes to 10 minutes, from 10 minutes to 15 minutes, from 15 minutes to
  • 10 years from 10 years to 20 years, at least 1 minute, at least 2 minutes, at least 3 minutes, at least 5 minutes, at least 10 minutes, at least 15 minutes, at least 20 minutes, at least 30 minutes, at least 1 hour, at least 2 hours, at least 6 hours, at least 12 hours, at least 24 hours, at least 2 days, at least 5 days, at least 1 week, at least 2 weeks, at least 1 month, at least 2 months, at least 6 months, at least 1 year, at least 2 years, at least 3 years, at least 5 years, at least 10 years, at least 20 years, at most 1 minute, at most 2 minutes, at most 3 minutes, at most 5 minutes, at most 10 minutes, at most 15 minutes, at most 20 minutes, at most 30 minutes, at most 1 hour, at most 2 hours, at most 6 hours, at most 12 hours, at most 24 hours, at most 2 days, at most 5 days, at most 1 week, at most 2 weeks, at most 1 month, at most 2 months, at most 6 months, at most 1 year, at most 2 years, at most 3 years,
  • a healthy subject’s profile can be recorded for at least about: 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months 9 months, 10 months, 11 months, 1 year, 2 years, 3 years, 4 years, 5 years, 6 years, 7 years, 8 ears, 9 years, 10 years, 11 years, 12 years, 13 years, 14 years, 15 years, 20 years, 25 years, 30 years, 35 years, 40 years, 50 years, 60 years, 70 years, 80 years, 90 years or 100 years.
  • a healthy subject’s metabolic profile can be recorded from about: 1 day to about 100 days, 1 day to about 50 days, 1 day to about 25 days, 1 day to about 15 days, 1 day to about 13 days, 1 day to about 12 days, 1 day to about 11 days, 1 day to about 10 days,
  • a subject’s profile can be recorded once over a time period. In some cases, a subjects profile can be recorded at least about: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15,
  • a metabolic profile of the subject can be unknown.
  • the healthy metabolic profile can be determined from factors comprising a subject’s genetic profile, a diseased metabolic profile, a healthy population profile, a diseased population profile, a reference profile, or any combination thereof.
  • a method can comprise performing a diagnostic test and administering a therapeutic comprising the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof.
  • the diagnostic test can comprise subjecting a sample or at least a portion of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof to a device comprising: a chromatography column, an eluant, a detector, and a mass spectrometer.
  • a method can be promoting or maintain health in a subject. Promoting or maintaining health in a subject can comprise promoting or maintaining: ear, nose and throat health, eye health, cardiovascular health, respiratory health, skeletal health, skin health, urogenital health, digestive health, muscular health, neural health, immune system health, liver health, mental health, sexual health, circulatory system health, joint health, dermatological health, lung health, dental health, bone health, central nervous system health, peripheral nervous system health, spine health, kidney health, heart health, intestine health, podiatry health, endocrine health or any combination thereof.
  • health can include pediatric health, infant health, adult health or any combination thereof.
  • a starting material can be administered to a subject and the starting material can be transformed into the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof.
  • the starting material can be a compound of a biological pathway, an intermediate in a biological pathway, a prodrug, or any combination thereof.
  • the transformation can be a phase 1 reaction, a phase 2 reaction, a microbial biotransformation, a digestive biproduct, a metabolic biproduct, a degradation biproduct or any combination thereof.
  • a compound can be administered to a subject to alter the production of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, a enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • the production of a biomolecule in Table 2 can be altered.
  • the compound can be an inhibitor, an activator, a metabolite, a toxin, an intermediate compound of a biological pathway, or any combination thereof.
  • the compound can be an at least partial inhibitor or a complete inhibitor to a polypeptide or a biologically active fragment thereof.
  • the compound can have a direct or an indirect effect on a biological pathway.
  • the biological pathway can be a competing biological pathway.
  • the production of the compound from Table 1, a homolog thereof, a derivative thereof, an ester thereof, a enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can increase or decrease.
  • the compound from Table 1 can be structurally modified.
  • the compound of Table 1, homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof can be covalently linked to an antibody or fragment thereof.
  • the linkage can be a cleavable linkage.
  • a non-cleavable linkage can comprise a thioether linker, a maleimide alkane linker, a maleimide cyclohexane linker, or any combination thereof.
  • the cleavable linkage can comprise an acid-labile linker, a reducible linker, a disulfide-linker, a hydrazone linker, a peptide linker, or any combination thereof.
  • the method of treating or preventing a disease can include at least partially inducing, promoting, stimulating, or any combination thereof endocytosis from a cell in the subject.
  • the endocytosis can be of a polypeptide, a virus, a lipid, a polynucleotide, a molecule, a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or the salt thereof.
  • the endocytosis can be measured by intravital microscopy, fluorescence microscopy, and labeling endocytosed substrates. In some cases, the endocytosis can be measured by comparing endocytosis in untreated cells with cells that have been treated.
  • the cell can be an isolated cell from the subject. In some cases, the isolated cell can be manipulated in vitro. In some cases, the method can include at least partially inhibiting a cell growth, a cell communication, a cell metastasis or any combination thereof in the subject. In some cases, the cell growth, the cell communication, or the cell metastasis can be of a cancer cell, a tumor cell, a mutated cell, or any combination thereof.
  • the cell growth, the cell communication, or the cell metastasis can be inhibited by a direct or indirect contact with the pharmaceutical composition.
  • the cell growth, the cell communication, or the cell metastasis can be measured by a biochemical assay, intravital microscopy, fluorescence microscopy, tumor shrinkage, cancer cell depletion, cancer remission, cell death, or any combination thereof.
  • the cell growth, the cell communication or the cell metastasis can be measured by comparing cell growth, the cell communication or the cell metastasis in an untreated cell with the cell growth, the cell communication or the cell metastasis in a treated cell.
  • the cell can be an isolated cell from the subject.
  • the isolated cell can be manipulated in vitro and shown to be have inhibited cell growth, cellular communication, or cellular metastasis.
  • the method can include at least partially inducing, promoting, stimulating, or any combination thereof apoptosis or cell death in the subject.
  • the apoptosis or cell death can be of a cancer cell, a tumor cell, a mutated cell, or any combination thereof.
  • apoptosis or cell death can be stimulated by a direct or indirect contact with the pharmaceutical composition.
  • the cell death or apoptosis can be measured by a biochemical assay, intravital microscopy, fluorescence microscopy, tumor shrinkage, cancer cell depletion, cancer remission or any combination thereof.
  • the cell can be an isolated cell from the subject.
  • the isolated cell can be manipulated in vitro and shown to be have increased cell death or apoptosis.
  • the method can include at least partially inducing, promoting, stimulating or any combination thereof Reactive Oxygen Species (ROS) within the subject.
  • ROS Reactive Oxygen Species
  • the ROS can be produced by a direct or indirect contact with the pharmaceutical composition.
  • the ROS production can occur in a cancer cell, a tumor cell, a mutated cell, or any combination thereof.
  • the ROS production can be measured by an electron paramagnetic resonance assay, a cytochrome c reduction assay, nitroblue tetrazolium assay, fluorescence analysis of dihydroethidium or hydroethidium, fluorescence analysis of hydrocyans, chemiluminescence assays, electrochemical detection, a probe, in vivo redox imaging using photon-electron double-resonance imagine, fluorescence microscopy, or any combination thereof.
  • the cell can be an isolated cell from the subject.
  • the ROS can be measured by comparing ROS production in an untreated cell with ROS production in a treated cell
  • the isolated cell can be manipulated in vitro and shown to have increased ROS production.
  • the method can comprise at least partially activating or deactivating an immune cell.
  • the immune cell can be activated or deactivated by a direct or indirect contact with a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or the salt thereof.
  • the immune cell can be a dendritic cell, a neutrophil, a granulocyte, a monocyte, a lymphocyte, a B-cell, a T-cell, a natural killer (NIC) cell, a neutrophil, a mast cell, a basophil, an eosinophil, a memory B-cell, a plasma cell, a memory T-cell, a CD4 T-cell, a CD8 T-cell, a Thl T-cell, a Thl7 T-cell, a Treg T-cell, a Th2 T-cell, a precursor cell type to any of these or any combination thereof.
  • an immune cell can be induced into maturation, senescence, differentiation or any combination thereof.
  • a myeloblast can be induced into a granulocyte or a CD4 T-cell can be induced into a TH17 T-cell.
  • a hyperactive immune cell can be induced into senescence.
  • an activated immune cell can indirectly suppress a tumor or a cancer in a subject (e.g. a CD4 T-Cell a CD8 T-cell, an NK cell or a combination thereof).
  • an immune cell can be deactivated and suppress a tumor, for example, in a lymphoma or a leukemia.
  • an immune cell can be activated and repress an infection, for example, activation of a CD8 T-Cell to suppress a viral infection or activation of a neutrophil to suppress a bacterial or fungal infection.
  • the immune cell can be activated to remove excess protein or other toxic molecules, for example, a NK cell can be activated to remove alpha-synuclein aggregates in a subject.
  • the method can comprise at least partially disrupting a communication between one or more cells in the subject.
  • cell communication can be inhibited, enhanced, altered, or any combination thereof.
  • cell communication can be altered in one cell type, or multiple cell types.
  • disrupting cell communication can lead to cell death or apoptosis.
  • disrupting cell communication can increase or decrease an immune response.
  • disrupting cell communication can increase or decrease neural cell communication.
  • the method of treating or preventing a disease can comprise directly or indirectly at least partially increasing in the subject a level, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
  • the treating or preventing disease can comprise administering a genetically engineered cell to capture, to produce, or to chemically alter the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof.
  • the genetically engineered cell can be a prokaryote.
  • the prokaryote can be a bacterium (e.g. E. coli or a Lactobacillus species) or an Archaea.
  • the genetically engineered cell can be a eukaryotic cell.
  • the genetically engineered cell can be autologous or allogenic.
  • the genetically engineered cell can comprise a polynucleotide.
  • the polynucleotide encodes for a polypeptide or a biologically active fragment that can produce, alter, capture, or degrade the compound of
  • the method of treating or preventing a disease can comprise directly or indirectly at least partially reducing in the subject a level, an activity, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, a biomolecule of Table 2 or any combination thereof.
  • the treating or preventing disease can comprise editing a polynucleotide.
  • editing can be performed by a CRISPR Cas system or a similar nuclease.
  • the polynucleotide can encode for a polypeptide in a biochemical pathway for making the compound, or for degrading the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof.
  • the polynucleotide can comprise a ribose or a deoxyribose.
  • the edited polynucleotide can be comprised in an isolated cell and the isolated cell can be administered to the subject. In some cases, the isolated cell can be autologous or allogenic.
  • the treating or preventing disease can comprise administering an at least partially interfering polynucleotide to bind to RNA or DNA to at least partially inhibit polypeptide production in a biochemical pathway for making the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the salt thereof, a biomolecule of Table 2 or any combination thereof.
  • the interfering polynucleotide can be a miRNA molecule or a siRNA molecule.
  • the treating or preventing disease can comprise administering a CRISPR system to edit a polynucleotide to at least partially inhibit polypeptide production in a biochemical pathway for making the compound.
  • the CRISPR system can be a class 1 system, a class 2 system.
  • the CRISPR system can comprise, a Cas3, a Cas8a, a Cas5, a
  • an edited polynucleotide can have a nonsense mutation, a missense mutation, a deletion or any combination thereof.
  • a polynucleotide can be engineered.
  • an engineered polynucleotide can comprise a plurality of nucleotides.
  • an engineered oligonucleotide can comprise an artificial nucleic acid analogue.
  • an engineered oligonucleotide can comprise DNA, can comprise cell-free DNA, cDNA, fetal
  • an oligonucleotide can comprise miRNA, shRNA, or siRNA.
  • the treating or preventing disease can comprise administering an antibody, an aptamer, or a fragment thereof to at least partially bind to the compound of Table 1 the homolog thereof, the derivative thereof, the ester thereof, or the salt thereof.
  • the antibody can be monoclonal or polyclonal.
  • the antibody or fragment thereof can be from an IgG, IgA, IgM.
  • the binding can decrease bioavailability, decrease half-life, inhibit the action of the compound, or any combination thereof.
  • the treating or preventing disease can comprise administering a molecule, a salt thereof, or a formulation containing at least one of these to capture the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, or the salt thereof.
  • the molecule can contain functional groups that bind to the compound.
  • the binding can be a covalent bond an ionic bond or a combination thereof.
  • the treating or preventing disease can comprise administering dialysis to a patient, wherein the dialysis filters at least some of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, or the salt thereof.
  • the dialysis can comprise hemodialysis, peritoneal dialysis, or any combination thereof.
  • the dialysis can be hemodialysis, peritoneal dialysis, or any combination thereof.
  • the compound can be filtered from the blood of a subject.
  • a second therapy can include chemotherapy, radiation, bone marrow transplantation, a checkpoint inhibitor, CAR T-cell therapy, immunotherapy, hormone therapy, cryotherapy, surgical procedure (such as tumor resection) or any combination thereof.
  • a second therapy can include administration of a pharmaceutical composition, such as a small molecule.
  • a second therapy can include administration of a pharmaceutical composition, such as one or more antiviral drugs, for example, interferon, oseltamivir, ribavirin, daclatasvir, sofosbuvir, velpatasvir, voxilapresvir, remdesivir, indomethacin, or any combination thereof.
  • a second therapy can include administration of a pharmaceutical composition, such as one or more antibiotics.
  • a second therapy can comprise administration of a muscle relaxant, an anti-depressant, a steroid, an opioid, a cannabis-based therapeutic, acetaminophen, a non-steroidal anti-inflammatory, a neuropathic agent, a cannabis, a progestin, a progesterone, or any combination thereof.
  • a neuropathic agent may comprise gabapentin.
  • a non-steroidal anti-inflammatory can comprise naproxen, ibuprofen, a COX-2 inhibitor, or any combination thereof.
  • a second therapy can comprise administration of a heart medication such as a high blood pressure medication, a beta blocker, an anticoagulant, an antiplatelet agent, an ACE inhibitor, an angiotensin II receptor blocker, an angiotensin receptor-neprilysin inhibitor, a calcium channel blocker, a cholesterol-lowering medication, a diuretic, a vasodilator or any combination thereof.
  • a heart medication such as a high blood pressure medication, a beta blocker, an anticoagulant, an antiplatelet agent, an ACE inhibitor, an angiotensin II receptor blocker, an angiotensin receptor-neprilysin inhibitor, a calcium channel blocker, a cholesterol-lowering medication, a diuretic, a vasodilator or any combination thereof.
  • a second therapy can comprise administration of a biologic agent, cellular therapy, regenerative medicine therapy, a tissue engineering approach, a stem cell transplantation or any combination thereof.
  • a second therapy can comprise a medical procedure.
  • a medical procedure can comprise an epidural injection (such as a steroid injection), acupuncture, exercise, physical therapy, an ultrasound, a surgical therapy, a chiropractic manipulation, an osteopathic manipulation, a chemonucleolysis, or any combination thereof.
  • a second therapy can comprise use of a breathing assist device or a ventilator.
  • a second therapy can comprise administration of a regenerative therapy or an immunotherapy such as a protein, a stem cell, a cord blood cell, an umbilical cord tissue, a tissue, or any combination thereof.
  • a second therapy can comprise a biosimilar.
  • a second therapy can alter the levels of compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof in a subject.
  • a second therapy may not alter the levels of compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof in a subject.
  • the administering of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or a salt thereof and a second therapy can be concurrent.
  • the administering of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or a salt thereof and a second therapy can be sequential.
  • the treating or preventing can comprise administering a polynucleotide to a subject, a genetically engineered virus, a vector, or any combination thereof to a subject to directly or indirectly produce the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, or the salt thereof or any combination thereof in a subject.
  • the genetically engineered virus, the vector or any combination thereof can comprise at least one polynucleotide.
  • a polynucleotide can encode for a polypeptide or biologically active fragment thereof in a biochemical pathway for making the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, or the salt thereof.
  • the polynucleotide can encode for a gene, a biologically active RNA, such as siRNA or miRNA, or any combination thereof.
  • the vector can be a plasmid.
  • a vector can comprise RNA, DNA or any combination thereof.
  • a vector can be used for drug delivery, in an in vitro setting, in vivo setting, or any combination thereof.
  • the vector can comprise a recombinant vector.
  • the vector can be a vector that can be modified from a naturally occurring vector.
  • the vector can comprise at least a portion of a non-naturally occurring vector. Any vector can be utilized.
  • a vector can deliver the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof.
  • the vector can be targeted to but may not be limited to a subject, or a specific organ, or a specific cell, or any combination thereof.
  • the vector can comprise any composition described herein.
  • the vector can be comprised of a liposome, a nanoparticle or any combination thereof.
  • the liposome can include but may not be limited to unilamellar liposome, multilamellar liposome, archaeosome, noisome, novasome, cryptosome, emulsome, vesosome, or a derivative of any of these, or any combination thereof.
  • the nanoparticle can include but may not be limited to biopolymeric nanoparticle, alginate nanoparticle, xanthan gum nanoparticle, cellulose nanoparticle, dendrimer, polymeric micelle, polyplexed, inorganic nanoparticle, nanocrystal, metallic nanoparticle, quantum dot, protein nanoparticle, polysaccharide nanoparticle, or a derivative of any of these, or any combination thereof.
  • the vector can be an RNA viral vector which can include but may not be limited to a retrovirus, lentivirus, coronavirus, alphavirus, flavivirus, rhabdovirus, morbillivirus, picornavirus, coxsackievirus, or picornavirus or portions of any of these, or fragments of any of these, or any combination thereof.
  • the vector can be a DNA viral vector which can include but may not be limited to an adeno-associated viral (AAV) vector, adenovirus, hybrid adenoviral system, hepadnavirus, parvovirus, papillomavirus, polyomavirus, herpesvirus, poxvirus, a portion of any of these, or a fragment of any of these, or any combination thereof.
  • AAV adeno-associated viral
  • a subject may not have been previously diagnosed with a disease or condition. In some cases, a subject may have been diagnosed with a disease or condition. In some cases, a subject may not have received a definitive diagnosis of a disease or condition.
  • a subject may have previously had a disease or condition.
  • a subject may be in remission.
  • a subject may be at risk of developing a disease or condition (such as based at least in part on a previous condition, a lifestyle factor, a genetic variant, or any combination thereof).
  • a subject may have received a diagnostic test.
  • a subject may receive a second diagnostic test in addition to the diagnostic device, the screening device, or the diagnostic and screening device described herein.
  • a diagnostic test can include an imaging procedure, a blood count analysis, a tissue pathology analysis, a biomarker analysis, a rapid diagnostic test, a polymerase chain reaction diagnostic test, an enzyme-linked immunosorbent assay (ELISA), a biopsy, a colonoscopy, an electrocardiogram, an electroencephalogram, a gastroscopy, an eye test, an magnetic resonance imaging (MRI scan), a positron emission tomography (PET) scan, an ultra sound, an X-ray, a computed tomography (CT) scan, a physical examination, an organ function test, a pregnancy test, a toxicology test, a uranalysis, a thyroid function test, a pap smear, a gastric fluid analysis, a fecal sample analysis, a histology test, a genetic test, a mental function test, or any combination thereof.
  • MRI scan magnetic resonance imaging
  • PET positron emission tomography
  • CT computed tomography
  • the disease or condition may comprise a cancer, a tumor, an infectious disease, a skeletal disease, a urogenital disease, a digestive disease, a muscular disease, a neurological disease, an autoimmune disease, a respiratory disease, a cardiac disease, an endocrine disease, a liver disease, a hereditary disease, or any combination thereof.
  • the disease or condition may be a viral infection, such as a SARS-CoV infection, a SARS-CoV-2 infection, a MERS-CoV infection, a HIV infection, or any combination thereof.
  • the disease or condition may be cancer, such as a lymphoma, a carcinoma, a sarcoma, a skin cancer, a cervical cancer, a prostate cancer, or any combination thereof.
  • Methods can include in vivo or in vitro delivery methods. Methods can include contacting a cell, such as a cell in vivo with the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, or the salt thereof. Methods can include contacting a cell, such as an isolated and purified cell (such as a cell in vitro) with the compound of Table
  • Methods can include contacting a tissue, such as an in vivo tissue or an isolated in vitro tissue, with a compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the salt thereof, the nucleic acid construct, or the pharmaceutical composition as described herein.
  • FIG. 1 shows a gas chromatography mass spectrometry
  • GCMS chromatogram of a patient with breast cancer, in accordance with embodiments.
  • FIG. 2 shows a GCMS chromatogram of a patient with kidney cancer, in accordance with embodiments.
  • FIG. 3A-3D show GCMS chromatograms from gastric patients’ blood, in accordance with embodiments.
  • FIG. 4A-4D show this figure shows GCMS chromatograms from lung cancer patients’ blood.
  • FIG. 5A-5D show this figure shows GCMS chromatograms from gastric cancer patients’ urine, in accordance with embodiments.
  • FIG. 6A-6D these figures show this figure shows GCMS chromatograms from kidney cancer patients’ urine, in accordance with embodiments.
  • FIG. 7A-7D these figures show this figure shows GCMS chromatograms from lung cancer patients’ urine, in accordance with embodiments.
  • FIG. 8A-8C show this figure shows GCMS chromatograms from health control’s blood, in accordance with embodiments.
  • FIG. 9A-B shows chromatograms of breast cancer patients, in accordance with embodiments.
  • FIG. 9A shows one breast cancer patient’s metabolite volatile organic compound (VOC) profile.
  • FIG. 9B shows an overlay of six breast cancer patients’ metabolite VOC profile.
  • VOC volatile organic compound
  • FIG. 10A-B shows chromatograms of kidney cancer patients.
  • FIG. 10A shows one kidney cancer patients metabolite VOC profile.
  • FIG.10B shows an overlay of six kidney cancer patients’ metabolite VOC profiles.
  • FIG. 11A-B shows chromatograms of lymphoma patients, in accordance with embodiments.
  • FIG. 11A shows one lymphoma VOC profile.
  • FIG. 11B shows an overlay of six lymphoma patients’ metabolite VOC profiles.
  • FIG. 12A-12G show this figure shows chromatograms of breast and kidney cancer patients’ blood, in accordance with embodiments.
  • FIG. 13 shows chromatograms from lung cancer and liver cancer patients’ blood, in accordance with embodiments.
  • FIG. 14 this figure shows chromatograms from kidney cancer patients’ urine, in accordance with embodiments.
  • FIG. 15 this figure shows saliva chromatograms, in accordance with embodiments.
  • FIG. 16 this figure shows chromatograms from volunteers’ breath, in accordance with embodiments.
  • Portable gas chromatography (GC) chromatograms in the top and middle rows are from volunteers.
  • the chromatogram at the lower left is a blank control chromatogram.
  • FIG. 17 this figure shows a chromatogram from a cancer patient, in accordance with embodiments.
  • the portable GC chromatogram is from the blood of a male lung cancer patient.
  • FIG. 18 shows a chromatogram from a cancer patient, in accordance with embodiments.
  • the portable GC chromatogram is from the blood of a female endometrial cancer patient.
  • FIG. 19 shows a chromatogram from a lymphoma patient, in accordance with embodiments.
  • the portable GC chromatogram is from the blood of a female lymphoma cancer patient.
  • FIG. 20 shows a chromatogram from a lung cancer patient, in accordance with embodiments.
  • the portable GC chromatogram is from the saliva of a female lung cancer patient.
  • FIG. 21 shows a chromatogram from a prostate cancer patient, in accordance with embodiments.
  • the portable GC chromatogram is from the blood of a male with prostate cancer.
  • FIG. 22 shows a chromatogram of a healthy control and cancer patients, in accordance with embodiments.
  • the portable GC chromatogram is an overlay of a healthy control patient, a breast cancer patient, a lung cancer patient, a kidney cancer patient.
  • FIG. 23 shows a chromatogram of a healthy control and a lab standard, in accordance with embodiments.
  • the portable GC chromatogram of a healthy controls urine and a lab made synthetic standard urine sample to identify VOCs.
  • FIG. 24 this figure shows a gas chromatography (GC) chromatogram analysis of heated blood and urine samples from kidney cancer patients.
  • GC gas chromatography
  • FIG. 27A-B this figure shows a GC device for testing a sample.
  • FIG.27A shows the connected to a computer that displays the chromatogram.
  • FIG. 27B shows the device connected to a breath sampling mask.
  • FIG. 28A-B shows a GC device testing a sample from a subject.
  • FIG.28A shows the device connected to a PC which can transmit or present the data.
  • a robotic drone is pictured moving the device.
  • FIG. 28B shows a device that can transmit the data wirelessly via cellular connections or Wi-Fi. The device is autonomously moved by a robot.
  • a sample 202 containing a blood sample can be obtained from a subject 201, such as a human subject.
  • a sample 202 can be subjected to one or more methods as described herein, such as adding the sample to a chromatography column, an eluant, a detector, a mass spectrometer, or any combination thereof.
  • an assay can comprise obtaining a chromatograph.
  • One or more results from a method can be input into a processor 204.
  • One or more input parameters such as a sample identification, subject identification, sample type, a reference, or other information can be input into a processor 204.
  • One or more metrics from an assay can be input into a processor 204 such that the processor can produce a result, such a diagnosis, a prognosis, a theranostic, of a viral infective disease or a recommendation for a treatment.
  • a processor can send a result, an input parameter, a metric, a reference, or any combination thereof to a display 205, such as a visual display or graphical user interface.
  • a processor 204 can (i) send a, an input parameter, a metric, or any combination thereof result wirelessly or directly to a server 207, (ii) receive a result, an input parameter, a metric, or any combination thereof from a server 207, (iii) or a combination thereof.
  • the output can be a chromatograph.
  • FIG. 25 shows a computer system 101 that is programmed or otherwise configured to perform gas chromatography on a blood sample.
  • the computer system 101 can regulate various aspects of the present disclosure, such as, diagnosis, prognosis, a theranostic method, and a health level information method
  • the computer system 101 can be an electronic device of a user or a computer system, a handheld system, a cell phone or any combination thereof that is remotely located with respect to the electronic device.
  • the computer system 101 includes a central processing unit (CPU, also “processor” and “computer processor” herein) 105, which can be a single core or multi core processor, or a plurality of processors for parallel processing.
  • the computer system 101 also includes memory or memory location 110 (e.g., random-access memory, read-only memory, flash memory), electronic storage unit 115 (e.g., hard disk), communication interface 120 (e.g., network adapter) for communicating with one or more other systems, and peripheral devices 125, such as cache, other memory, data storage and/or electronic display adapters.
  • the memory 110, storage unit 115, interface 120 and peripheral devices 125 are in communication with the CPU 105 through a communication bus (solid lines), such as a motherboard.
  • the storage unit 115 can be a data storage unit (or data repository) for storing data.
  • the computer system 101 can be operatively coupled to a computer network (“network”) 130 with the aid of the communication interface 120.
  • the network 130 can be the Internet, an internet and/or extranet, or an intranet and/or extranet that is in communication with the Internet.
  • the network 130 in some cases is a telecommunication and/or data network.
  • the network 130 can include one or more computer servers, which can enable distributed computing, such as cloud computing.
  • the network 130 in some cases with the aid of the computer system 101, can implement a peer-to-peer network, which may enable devices coupled to the computer system 101 to behave as a client or a server.
  • the CPU 1105 can execute a sequence of machine-readable instructions, which can be embodied in a program or software.
  • the instructions may be stored in a memory location, such as the memory 110.
  • the instructions can be directed to the CPU 105, which can subsequently program or otherwise configure the CPU 105 to implement methods of the present disclosure. Examples of operations performed by the CPU 105 can include fetch, decode, execute, and writeback.
  • the CPU 105 can be part of a circuit, such as an integrated circuit.
  • a circuit such as an integrated circuit.
  • One or more other components of the system 101 can be included in the circuit.
  • the circuit is an application specific integrated circuit (ASIC).
  • ASIC application specific integrated circuit
  • the storage unit 115 can store files, such as drivers, libraries and saved programs.
  • the storage unit 115 can store user data, e.g., user preferences and user programs.
  • the computer system 101 in some cases can include one or more additional data storage units that are external to the computer system 101, such as located on a remote server that is in communication with the computer system 101 through an intranet or the Internet.
  • the computer system 101 can communicate with one or more remote computer systems through the network 130. For instance, the computer system 101 can communicate with a remote computer system of a user (e.g., a physician).
  • remote computer systems examples include personal computers (e.g., portable PC), slate or tablet PC’s (e.g., Apple® iPad, Samsung® Galaxy Tab), telephones, Smart phones (e.g., Apple® iPhone, Android- enabled device, Blackberry®), or personal digital assistants.
  • the user can access the computer system 101 via the network 130.
  • Methods as described herein can be implemented by way of machine (e.g., computer processor) executable code stored on an electronic storage location of the computer system 101, such as, for example, on the memory 110 or electronic storage unit 115.
  • the machine executable or machine readable code can be provided in the form of software.
  • the code can be executed by the processor 105.
  • the code can be retrieved from the storage unit 1115 and stored on the memory 110 for ready access by the processor 105.
  • the electronic storage unit 115 can be precluded, and machine- executable instructions are stored on memory 110.
  • the code can be pre-compiled and configured for use with a machine having a processer adapted to execute the code, or can be compiled during runtime.
  • the code can be supplied in a programming language that can be selected to enable the code to execute in a pre-compiled or as-compiled fashion.
  • aspects of the systems and methods provided herein can be embodied in programming.
  • Various aspects of the technology may be thought of as “products” or “articles of manufacture” typically in the form of machine (or processor) executable code and/or associated data that is carried on or embodied in a type of machine readable medium.
  • Machine-executable code can be stored on an electronic storage unit, such as memory (e.g., read-only memory, random-access memory, flash memory) or a hard disk.
  • “Storage” type media can include any or all of the tangible memory of the computers, processors or the like, or associated modules thereof, such as various semiconductor memories, tape drives, disk drives and the like, which may provide non-transitory storage at any time for the software programming. All or portions of the software may at times be communicated through the Internet or various other telecommunication networks. Such communications, for example, may enable loading of the software from one computer or processor into another, for example, from a management server or host computer into the computer platform of an application server.
  • another type of media that may bear the software elements includes optical, electrical and electromagnetic waves, such as used across physical interfaces between local devices, through wired and optical landline networks and over various air-links.
  • a machine readable medium such as computer-executable code
  • a tangible storage medium such as computer-executable code
  • Non-volatile storage media include, for example, optical or magnetic disks, such as any of the storage devices in any computer(s) or the like, such as may be used to implement the databases, etc. shown in the drawings.
  • Volatile storage media include dynamic memory, such as main memory of such a computer platform.
  • Tangible transmission media include coaxial cables; copper wire and fiber optics, including the wires that comprise a bus within a computer system.
  • Carrier-wave transmission media may take the form of electric or electromagnetic signals, or acoustic or light waves such as those generated during radio frequency (RF) and infrared (IR) data communications.
  • RF radio frequency
  • IR infrared
  • Common forms of computer-readable media therefore include for example: a floppy disk, a flexible disk, hard disk, magnetic tape, any other magnetic medium, a CD-ROM, DVD or DVD- ROM, any other optical medium, punch cards paper tape, any other physical storage medium with patterns of holes, a RAM, a ROM, a PROM and EPROM, a FLASH-EPROM, any other memory chip or cartridge, a carrier wave transporting data or instructions, cables or links transporting such a carrier wave, or any other medium from which a computer may read programming code and/or data.
  • Many of these forms of computer readable media may be involved in carrying one or more sequences of one or more instructions to a processor for execution.
  • the computer system 101 can include or be in communication with an electronic display 135 that comprises a user interface (UI) 140 for providing, for example, a chromatogram of the subject.
  • UI user interface
  • Examples of UFs include, without limitation, a graphical user interface (GUI) and web-based user interface.
  • GUI graphical user interface
  • Methods and systems of the present disclosure can be implemented by way of one or more algorithms.
  • An algorithm can be implemented by way of software upon execution by the central processing unit 105. The algorithm can, for example, determine if a subject’s chromatogram matches that of a healthy patient’s profile or that it matches that of a patient with a known disease.
  • a patient is diagnosed with pancreatic cancer and the patient’s breath is tested with the gas chromatography device described herein.
  • the patient’s chromatogram displays reduced levels of two volatile organic compounds from Table 1 as compared to a subject without the disease.
  • the two compounds are administered to the patient who has pancreatic cancer by a nebulizer twice a day for six weeks and during this time the patient’s chromatogram is monitored for changes in the presence and concentration of volatile organic compounds. After one week, the levels of the two chemicals begin to rise in the patient.
  • An imaging diagnostic test is administered, and cancer is shown to be decreasing in size as compared to before the treatment with the two compounds.
  • a biopsy is taken of the pancreatic cancer. A portion of the cancer cells are tested in vitro, and administration of the compound to the cancer cells demonstrates that they are killed on contact by the two compounds.
  • a patient is diagnosed with lymphatic cancer and the patient’s serum is tested with the gas chromatography device described herein.
  • the patient’s chromatogram displays reduced levels of a volatile organic compound from Table 1 as compared to a subject without the disease.
  • the compound is administered to the patient who has lymphatic cancer by intravenous administration each day for eight weeks.
  • the patient’s volatile organic compound chromatogram is monitored every three days by the device for changes in the presence and concentration of volatile organic compounds. After two weeks, the levels of the compound begin to rise in the patient.
  • a biopsy sample is administered at the end of treatment, and the lymphatic cancer cells are shown to be reduced in number as compared to before the treatment with the compound. A portion of the cancer cells are tested in vitro , and administration of the compound for two hours to the cancer cells kills the cancer cells.
  • a patient is diagnosed with endometrial cancer and the patient’s serum is tested with the gas chromatography device described herein.
  • the patient’s chromatogram displays increased levels of three volatile organic compound from Table 1 as compared to a subject without the disease.
  • the compounds are removed by dialysis every four days for ten weeks.
  • the patient’s volatile organic compound chromatogram is monitored every day by the device for changes in the presence and concentration of volatile organic compounds. After three weeks, the levels of the three chemicals begin to rise in the patient.
  • An imaging diagnostic test and a blood diagnostic is administered at the end of treatment, and the cancer is shown to be in remission as compared to before the dialysis treatment that removed the three compounds.
  • a patient arrives to a health care provider and is having trouble breathing.
  • a patient’s breath and saliva are tested with the gas chromatography device described herein.
  • the patient’s chromatogram displays reduced levels of two volatile organic compound from Table 1 as compared to a subject without the disease. This chromatogram pattern is associated with lung cancer.
  • An imaging diagnostic test is performed and confirms the patient has lung cancer.
  • the compounds are administered to the patient who has lung cancer by an inhaler each day for twenty weeks.
  • the patient’s volatile organic compound chromatogram is monitored every two days by the device for changes in the presence and concentration of volatile organic compounds. After six weeks, the levels of the compounds begin to rise in the patient.
  • An imaging diagnostic test and a biopsy sample is administered at the end of treatment. The diagnostic image displays a reduction in the size of the cancer.
  • a portion of the cancer cells from the biopsy are tested in vitro , and administration of the two compounds inhibit cell growth of the cancer cells.
  • a patient is diagnosed with a melanoma and the patient’s skin is tested with the gas chromatography device described herein.
  • the patient’s chromatogram displays reduced levels of four volatile organic compounds from Table 1 as compared to a subject without melanoma.
  • the four compounds are administered to the patient who has melanoma by a applying a patch that contains the four compounds directly to the site of the melanoma, each day for nine weeks.
  • the patients skin sample is tested throughout the treatment for changes in the presence and concentration of the volatile organic compounds. After four days, the levels of the four chemicals begin to rise in the patient.
  • An imaging diagnostic test is administered, and cancer is shown to be decreasing in size as compared to before the treatment with the four compounds.
  • a biopsy is taken of the melanoma and the cancer cells are reduced.
  • a portion of the patient’s cancer cells and the patient’s natural killer (NK) cells are cocultured in vitro, and administration of the four compounds to the coculture demonstrates the NK cells begin to kill the cancer cells after administration of the four compounds.
  • a patient arrives to a health care provider and is having trouble breathing.
  • a patient’s breath is tested with the gas chromatography device described herein.
  • the patient’s chromatogram displays increased levels of two volatile organic compound from Table 1 as compared to a subject without the disease.
  • This chromatogram pattern is associated with SARS-CoV-2.
  • a treatment is administered for the SARS-CoV-2 infection and the patient’s volatile organic compound chromatogram is monitored every day by the device for changes in the presence and concentration of volatile organic compounds. After three days, the levels of the two compounds begin to rise in the patient.
  • a polymerase chain reaction (PCR) diagnostic test is administered at the end of treatment and the patient displays a normal level of the two compounds. The diagnostic test shows the patient is no longer positive for the virus.
  • PCR polymerase chain reaction
  • a patient’s blood is tested with the gas chromatography device described herein.
  • the patient’s chromatogram displays decreased levels of two volatile organic compound from Table 1 as compared to a subject without the disease. This chromatogram pattern is associated with dementia.
  • the compounds are administered to the patient by a pill once a day for ten years.
  • the patient’s volatile organic compound chromatogram is monitored every week by the device for changes in the presence and concentration of volatile organic compounds. After five weeks, the levels of the two compounds begin to rise in the patient.
  • a physical diagnostic test is administered after four months of treatment. The test shows the dementia symptoms have not progressed. After 8 months of treatment the patient is displaying normal levels of the two compounds and the symptoms of dementia have diminished.
  • a basal cell carcinoma cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
  • a squamous cell carcinoma cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
  • An adenocarcinoma cancer cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
  • An osteosarcoma cancer cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
  • a soft tissue sarcoma cancer cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
  • a rhabdomyosarcoma cancer cell is treated with a volatile organic compound from Table 1
  • a B-cell acute lymphoblastic leukemia cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
  • a S. aureus cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
  • An E. coli cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
  • AM tuberculosis cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
  • AN gonorrhoeae cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
  • a C. trachomatic cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
  • Plasmodium cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
  • the device is used to diagnose an infectious disease the SARS-CoV-2 virus. A new outbreak occurs and there is no current diagnostic available.
  • the device disclosed herein is immediately used to diagnose patients with a different pattern in their chromatogram. The chromatography profile is improved when more people are tested by algorithms.
  • the device is linked to a database, so information is updated immediately from other testing sites.
  • the metabolite profile pattern recognition algorithm is be updated over the air remotely and in real time. The disease can be diagnosed before antibodies appear.
  • While exemplary embodiments have been shown and described herein, such embodiments are by way of example only. Numerous variations, changes, and substitutions can be performed on the exemplary embodiments. It should be understood that various alternatives to the embodiments described herein may be employed.

Abstract

Disclosed herein are methods for treating or preventing a disease or condition. Also disclosed herein are methods of detecting a presence or an absence of a compound from a subject suspected of having a disease or condition. Also disclosed herein, are compounds and pharmaceutical compositions for treating or preventing a disease or condition.

Description

SMALL MOLECULE DRUGS AS TARGETED THERAPEUTICS
CROSS REFERENCE
[0001] This application claims priority to United States Provisional Patent Application No. 63/007,889 filed April 9, 2020, which is incorporated herein by reference in its entirety.
SUMMARY
[0002] Disclosed herein are methods of treating or preventing diseases or conditions. In some embodiments, the method can comprise: a) administering to a subject a therapeutically effective amount of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof; b) directly or indirectly at least partially increasing in a subject a level, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof; c) directly or indirectly at least partially reducing in a subject a level, an activity, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof, or any combination of (a-c). In some embodiments, a method can comprise administering to a subject a therapeutically effective amount of a compound of
Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some embodiments, a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be comprised in a pharmaceutical composition comprising an excipient, a diluent, or a carrier. In some embodiments, a pharmaceutical composition can comprise a first compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof; and a second compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some embodiments, a pharmaceutical composition can be in unit dose form. In some embodiments, a pharmaceutical composition can comprise a molecule selected from a group consisting of: isoprene, decanal, 4- methylbenzaldehyde, 3,7-dimethyloctan-3-ol, l-methyl-4(l -methyl -2 -propenyl)-benzene, 1- ethenyl-3 -ethyl-benzene, l-ethenyl-4-ethyl-benzene, 1-phenylbut-l-ene, octan-2-one, heptan-
3-one, hexan-3-one 5-methyl, butanoic acid 2-methyl-ethyl ester, propionic acid, 2- m ethoxy ethanol, acetaldehyde, propanal, n-propyl acetate, methyl methacrylate, styrene, 1,1- dipropoxypropane, a homolog of any of these, a derivative of any of these, an ester of any of these, a enantiomer of any of these, a diastereomer of any of these, a racemate of any of these, a salt of any of these, and any combination of any of these. In some embodiments, a pharmaceutical composition can further comprise albuterol or a salt thereof. In some embodiments, a pharmaceutical composition can be administered with an inhaler, a nebulizer, a vape device, a bronchodilator, a face mask, a nasal cannula, a diffuser, a tent, a container at least partially surrounding a subject’s head, or any combination thereof. In some embodiments, a method can further comprise administering a plant, a portion of a plant, an extract thereof, an herb, a portion of an herb, an extract thereof, or any combination thereof.
In some embodiments, a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof can be comprised in an implant. In some embodiments, an implant at least partially can release a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, or a salt thereof, or any combination thereof over a time when at least partially implanted in a subject. In some embodiments, a starting material can be administered to a subject and a starting material can be transformed into a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or a combination thereof. In some embodiments, a compound of Table
1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof ca ne covalently linked to an antibody or fragment thereof. In some embodiments, a compound of
Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be covalently linked by a cleavable linker to an antibody or fragment thereof. In some embodiments, a cell endocytosis can be at least partially promoted in a subject. In some embodiments, a cell endocytosis can be of a polypeptide, a virus, a lipid, a polynucleotide, a molecule, or any combination thereof. In some embodiments, a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be administered and can at least partially cross a blood brain barrier in a subject. In some embodiments, an apoptosis or a cell death can be at least partially induced in a subject. In some embodiments, an induction can be at least in part a selective induction. In some embodiments, a cell growth, a cell communication, a cell metastasis, or any combination thereof can be at least partially inhibited in a subject. In some embodiments, an inhibition can be at least in part a selective inhibition. In some embodiments, a Reactive Oxygen Species (ROS) can be at least partially induced within a subject. In some embodiments, a cell can be a cancer cell. In some embodiments, an immune cell can be at least partially deactivated in a subject. In some embodiments, a deactivation can be at least in part a selective deactivation. In some embodiments, an immune cell can be at least partially activated in a subject. In some embodiments, an activation can at least in part be a selective activation. In some embodiments, an immune cell can be at least partially induced into maturation, differentiation, or a combination thereof. In some embodiments, an immune cell can comprise a dendritic cell, a neutrophil, a granulocyte, a monocyte, a lymphocyte, or any combination thereof. In some embodiments, an immune cell can comprise a dendritic cell, a
CD4 T-cell, a CD8 T-cell, or any combination thereof. In some embodiments, a dendritic cell, a CD4 T-cell, a CD8 T-cell, or any combination thereof at least partially can directly or at least partially can indirectly suppress a tumor in a subject. In some embodiments, an immune cell can be a natural killer (NK) cell. In some embodiments, a NK cell at least partially can remove alpha-synuclein aggregates in a subject. In some embodiments, a treating or preventing disease can comprise administering a pharmaceutical composition comprising an excipient, a carrier, a diluent or any combination of these and a compound of
Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some embodiments, a treating or preventing can comprise administering a polynucleotide to a subject. In some embodiments, a polynucleotide can encode for a polypeptide or biologically active fragment thereof in a biochemical pathway for making a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some embodiments, treating or preventing disease can comprise administering a genetically engineered virus comprising at least one polynucleotide coding for a polypeptide, or a biologically active fragment thereof, in a pathway to produce a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some embodiments, treating or preventing disease can comprise administering a vector comprising at least one polynucleotide coding for a polypeptide, or a biologically active fragment thereof, in a pathway to produce a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some embodiments, a vector can be a plasmid. In some embodiments, treating or preventing disease can comprise administering a genetically engineered cell to capture, to produce, or to chemically alter a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some embodiments, a genetically engineered cell can be a eukaryotic cell. In some embodiments, a genetically engineered cell can be a prokaryotic cell. In some embodiments, a method can comprise altering of a composition of a food, a beverage, a nutrient, or any combination thereof consumed by a subject. In some embodiments, treating or preventing disease can comprise administering a compound to a subject. In some embodiments, a compound can be an at least partial inhibitor, a salt thereof, or a formulation containing at least one of these, of a polypeptide or biologically active fragment thereof involved in a competing biological pathway. In some embodiments, a production of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be altered. In some embodiments, treating or preventing disease can comprise administering an intermediate compound, a salt thereof, or a formulation thereof in a biological pathway. In some embodiments, a production of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be altered. In some embodiments, treating or preventing disease can comprise at least partially disrupting a communication between one or more cells in a subject. In some embodiments, a treating or preventing disease can comprise editing a polynucleotide. In some embodiments, a polynucleotide can encode for a polypeptide in a biochemical pathway for making a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, or a salt thereof. In some embodiments, a polynucleotide can comprise a ribose. In some embodiments, a polynucleotide can comprise a deoxyribose. In some embodiments, an edited polynucleotide can be comprised in an isolated cell and an isolated cell can be administered to a subject. In some embodiments, an isolated cell can be autologous. In some embodiments, an isolated cell can be allogenic. In some embodiments, a treating or preventing disease can comprise administering an at least partial inhibitor, a salt thereof, or a formulation containing at least one of these: of a polypeptide or biologically active fragment thereof, involved in a production of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some embodiments, treating or preventing disease can comprise administering an at least partially interfering polynucleotide to bind to RNA or DNA to at least partially inhibit polypeptide production in a biochemical pathway for making a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof in a subject. In some embodiments, a polynucleotide can comprise a ribose. In some embodiments, a polynucleotide can comprise a deoxyribose.
In some embodiments, a treating or preventing disease can comprise administering a CRISPR system to edit a polynucleotide to at least partially inhibit polypeptide production in a biochemical pathway for making a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof in a subject. In some embodiments, a polynucleotide can comprise a ribose. In some embodiments, a polynucleotide can comprise a deoxyribose.
In some embodiments, an edited polynucleotide can comprise a nonsense mutation, a missense mutation, a deletion, or any combination thereof. In some embodiments, a treating or preventing disease can comprise administering an antibody, an aptamer, or a fragment thereof to at least partially bind to a compound of Table 1 a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof in a subject. In some embodiments, treating or preventing disease can comprise administering a molecule, a salt thereof, or a formulation containing at least one of these to capture a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof in a subject. In some embodiments, treating or preventing disease can comprise administering dialysis to a patient. In some embodiments, a dialysis can filter at least some of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof from a patient. In some embodiments, a disease can comprise a cancer, a tumor, an infectious disease, a skeletal disease, a urogenital disease, a digestive disease, a muscular disease, a neurological disease, an autoimmune disease, a respiratory disease, a cardiac disease, an endocrine disease, a liver disease, a hereditary disease, or any combination thereof. In some embodiments, a disease can comprise a cancer.
In some embodiments, a cancer can comprise a sarcoma, a carcinoma, a melanoma, a lymphoma, leukemia, blastoma, germ cell tumor, myeloma, or any combination thereof. In some embodiments, a disease can comprise an infectious disease. In some embodiments, an infectious disease can comprise a viral infection. In some embodiments, a viral infection can comprise, influenza A, influenza B, SARS-CoV-2 (COVID-19), SARS-COV, MERS-CoV,
HKU1 coronavirus, OC43 coronavirus, NL63 coronavirus, 229E coronavirus, hepatitis A, hepatitis B, hepatitis C, norovirus, rotavirus, measles, rubella, HIV, enterovirus, herpes simplex 1, herpes simplex 2, varicella zoster virus, ebola virus, dengue virus or any combination thereof. In some embodiments, a disease can comprise an infectious disease. In some embodiments, an infectious disease can comprise a bacterial infection. In some embodiments, a bacterial infection can comprise Helicobacter pylori, Staphylococcus aureus,
Mycobacterium tuberculosis, Escherichia coli, Campylobacter jejuni, Klebsiella pneumonia,
Streptococcus pyogenes, Neisseria gonorrhoeae, Treponema pallidum, Borrelia burgdorferi,
Neisseria meningitidis, Chlamydia trachomatis, Salmonella, Vibrio, Enterobacteriaceae, or any combination thereof. In some embodiments, a disease can comprise an infectious disease.
In some embodiments, an infectious disease can comprise a fungal, a parasitic infection, or any combination thereof. In some embodiments, an infection can be a drug resistant infection.
In some embodiments, an infection can be a multi-drug resistant infection. In some embodiments, a disease can comprise a skeletal disease. In some embodiments, a skeletal disease can comprise osteoarthritis, osteoporosis, Paget disease of bone, or any combination thereof. In some embodiments, a disease can comprise an urogenital disease. In some embodiments, a urogenital disease can comprise benign prostate hyperplasia, urinary incontinence, kidney stones, kidney disease, erectile dysfunction, interstitial cystitis, prostatitis, overactive bladder, endometriosis, or any combination thereof. In some embodiments, a disease can comprise a digestive disease, wherein a digestive disease can comprise irritable bowel syndrome, celiac disease, gastroesophageal reflux disease, Chron’s disease, gastroparesis, or any combination thereof. In some embodiments, a disease can comprise a muscular disease. In some embodiments, a muscular disease can comprise muscular dystrophy, amyotrophic lateral sclerosis, Charcot-Marie-Tooth disease, myasthenia gravis, myopathy, myositis, peripheral neuropathy, or any combination thereof. In some embodiments, a disease can comprise a neurological disease. In some embodiments, a neurological disease can comprise a psychiatric disease, spinal cord injury, epilepsy, headache, migraine, dementia, vertigo, seizures, dizziness, sleep disorder, aneurysm, chronic fatigue syndrome, cerebral palsy, amyotrophic lateral sclerosis, neuralgia, neuropathy, or any combination thereof. In some embodiments, a disease can comprise an autoimmune disease.
In some embodiments, an autoimmune disease can comprise type 1 diabetes, rheumatoid arthritis, psoriasis, multiple sclerosis, systemic lupus erythematosus, Sjogren’s syndrome, an allergy, or any combination thereof. In some embodiments, a disease can comprise a respiratory disease. In some embodiments, a respiratory disease can comprise asthma, chronic obstructive pulmonary disease, chronic bronchitis, emphysema, cystic fibrosis, plural effusion, or any combination thereof. In some embodiments, a disease can comprise a cardiac disease. In some embodiments, a cardiac disease can comprise a congenital heart disease, arrhythmia, coronary artery disease, dilated cardiomyopathy, myocardial infarction, congestive heart failure, high blood pressure, mitral regurgitation, pulmonary stenosis, or any combination thereof. In some embodiments, a disease can comprise an endocrine disease. In some embodiments, an endocrine disease can comprise type 2 diabetes, hypothyroidism, hyperthyroidism, low testosterone, obesity, Addison’s disease, Cushing’s syndrome, Graves’ disease, thyroiditis, prolactinoma, or any combination thereof. In some embodiments, a disease can comprise a liver disease. In some embodiments, a liver disease can be cirrhosis, fibrosis, hemochromatosis, Wilson’s disease, or any combination thereof. In some embodiments, a disease can comprise a hereditary disease. In some embodiments, a hereditary disease can comprise, an inherited mendelian disorder, a peroxisomal disorder, a mitochondrial disorder, cystic fibrosis, sickle-cell anemia, Marfan syndrome, Huntington’s disease, kidney disease, Tay-Sachs disease, phenylketonuria, hemophilia, or any combination thereof. In some embodiments, a treating or preventing a disease, can comprise administering a second therapy. In some embodiments, a second therapy for cancer can comprise surgery, chemotherapy, radiation therapy, immunotherapy, hormone therapy, a checkpoint inhibitor, targeted drug therapy, CAR T-cell therapy, or a combination thereof. In some embodiments, administering of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof and a second administering can be concurrent. In some embodiments, administering of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof and a second administering can be sequential. In some embodiments, a method of treating or preventing disease can be administered by: injection, inhalation, catheterization, gastrostomy tube administration, intravenous administration, intraosseous administration, ocular administration, otic administration, topical administration, transdermal administration, oral administration, rectal administration, nasal administration, intravaginal administration, intracavernous administration, transurethral administration, buccal administration, sublingual administration, or a combination thereof. In some embodiments, an administration can comprise administering a nanoparticle, a viral vector, a viral-like particle, a liposome, an exosome, an extracellular vesicle, a microrobot, a microneedle, an implant, or a combination thereof. In some embodiments, an administration can comprise a topical administration, wherein a topical administration can comprise administering a patch, a lotion, a cream, a gel, a spray, an ointment, a liquid formulation, or a combination thereof. In some embodiments, a patch can comprise a microneedle patch, a single-layer drug in adhesive, a multi-layer drug in adhesive, a reservoir system, a matrix system, a vapour patch, or any combination thereof. In some embodiments, an administration can comprise an injection. In some embodiments, an injection can comprise administering an injector, intraarterial injection, intracerebroventricular injection, intraci sternal injection, intramuscular injection, intraorbital injection, intraparenchymal injection, intraperitoneal injection, intraspinal injection, intrathecal injection, intravenous injection, intraventricular injection, stereotactic injection, subcutaneous injection, or any combination thereof. In some embodiments, an injection can be administered directly to a site of a disease. In some embodiments, an administration can comprise an oral or rectal administration. In some embodiments, an oral or rectal administration can comprise administering a pill formulation, a liquid formulation, a powder formulation, or any combination thereof. In some embodiments, an administration can comprise an inhalation administration. In some embodiments, an inhalation administration can comprise administering an inhaler, a nebulizer, a vape device, a bronchodilator, a face mask, nasal cannula, a diffuser, a tent, a container at least partially surrounding a subject’s head, an aerosol formulation, a powder formulation, a liquid formulation, a gas formulation, or any combination thereof. In some embodiments, administering can be at least about once per day, twice per day, three times per day, or 4 times per day. In some embodiments, administering can be continuous. In some embodiments, administering can be by a pump or an implant. In some embodiments, a time of administration can comprise for at least about: 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months 11 months, 1 year, 2 years, 3 years, 4 years, 5 years, or for life. In some embodiments, a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can have an identical or a different dosage. In some embodiments, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can have an identical or a different route of administration.
[0003] Also disclosed herein, are methods that can comprise; contacting an isolated cell with a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some embodiments, after a contact an isolated cell can undergo apoptosis or cell death. In some embodiments, a cell can be a cancer cell.
[0004] Also disclosed herein are methods that can comprise; contacting an isolated cell with a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some embodiments, an isolated cell can produce ROS. In some embodiments, a method can be a method of screening. In some embodiments, a cell can be a cancer cell.
[0005] Also disclosed herein are pharmaceutical compositions that can comprise an excipient, a carrier, a diluent and a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some embodiments, a pharmaceutical composition can be in unit dose form. In some embodiments, a pharmaceutical composition can comprise a molecule selected from a group consisting of: isoprene, decanal, 4- methylbenzaldehyde, 3,7-dimethyloctan-3-ol, l-methyl-4(l -methyl -2 -propenyl)-benzene, 1- ethenyl-3 -ethyl-benzene, l-ethenyl-4-ethyl-benzene, 1-phenylbut-l-ene, octan-2-one, heptan-
3-one, hexan-3-one 5-methyl, butanoic acid 2-methyl-ethyl ester, propionic acid, 2- m ethoxy ethanol, acetaldehyde, propanal, n-propyl acetate, methyl methacrylate, styrene, 1,1- dipropoxypropane, a homolog of any of these, a derivative of any of these, an ester of any of these, a salt of any of these, and any combination of any of these. In some embodiments, a pharmaceutical composition can further comprise albuterol a salt thereof, or any combination thereof. In some embodiments, a pharmaceutical composition can be administered with an inhaler, a nebulizer, a vape device, a bronchodilator, a face mask, nasal cannula, a diffuser, a tent, a container at least partially surrounding a subject’s head, or any combination thereof.
[0006] Also disclosed herein are methods that can comprise detecting a presence and a level of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof, from a sample of a subject that can have or may be suspected of having a disease or condition and comparing a level of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof, to a reference level; or detecting a presence, a level, an elution time, of a plurality of compounds of Table 1, homologs thereof, derivatives thereof, esters thereof, enantiomers thereof, diastereomers thereof, racemates thereof, salts thereof, or any combination thereof and comparing a pattern thereof to a reference pattern. In some embodiments, a method can be a method of diagnosing a disease or a condition and can further comprise at least one of a following: selecting a therapeutic regiment for treating a disease or a condition, selecting a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof to treat a disease or condition, treating a subject for a disease or condition, or any combination thereof.
[0007] In some embodiments, a method can comprise detecting: a.) a presence and a level of a first compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof; b.) a presence and a level of a second compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof; c.) a presence and a level of a third compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof; and d.) comparing a first level to a first reference level, a second level to a second reference level, and a third level to a third reference level.
[0008] In some embodiments, a method can be conducted at least in part on a diagnostic device, a screening device, or a diagnostic and a screening device comprising at least one of; a chromatography column, an eluant, a detector, and a mass spectrometer. In some embodiments, a chromatography column can comprise a gas chromatography column and an eluant can comprise a gas. In some embodiments, a diagnostic device, a screening device, or a diagnostic and a screening device can comprise a microelectromechanical system (MEMS). In some embodiments, a diagnostic device, a screening device, or a diagnostic and a screening device can comprise a breath collection tube. In some cases, a device can comprise a self-contained power supply (e.g., a battery) and/or a wired power connection (e.g., a plug for electrical power). In some embodiments, a diagnostic device, a screening device, or a diagnostic and screening device can have: a) a positive volume of less than about 16 cubic feet, b) a method of transport, wherein a device can be moved by a robot, a drone, or a remote-control transport machine, c) a self- contained power supply, or d) or any combination of (a-c). In some embodiments, a robot, a drone, or a remote-control transport machine can be controlled autonomously. In some embodiments, a diagnostic device, a screening device, or a diagnostic a screening device can be linked wirelessly or directly to a processing station. In some embodiments, a diagnostic device, a screening device, or a diagnostic and screening device can be linked wirelessly by Wi-Fi, a broadband cellular network, or any combination thereof. In some embodiments, a processing station can analyze a data remotely. In some embodiments, a device can be about: 50 cm long, 30 cm wide, and 25 cm tall. In some embodiments, a device can be less than about: 12 cm long, 7 cm wide, and 3 cm tall. In some embodiments, an additional diagnostic test can be performed on a subject. In some embodiments, a sample can comprise a bodily fluid, a compound extruded by a body, a bodily part, a blood sample, a plasma sample, a urine sample, a sweat sample, a breath sample, a skin sample, a buccal sample, a hair sample, a fecal sample, a tear sample, a nasal sample, a nasal mucus sample, a biopsy sample, a saliva sample, a sputum sample or any combination thereof. In some embodiments, a compound extruded by a body can comprise an ester, a salt, or a combination thereof of a compound. In some embodiments, a compound extruded by a body can comprise a volatile compound. In some embodiments, a compound extruded by a body can comprise a metabolite. In some embodiments, a sample can further comprise an additive, a preservative, an anticoagulant, a membrane, an antibacterial compound, an antifungal compound, an antiviral, a chemical reagent, an antimicrobial compound, a diluent, or any combination thereof. In some embodiments, a sample can be treated with an electromagnetic wave, an ultrasound, a light wave, a radiation, a laser, adjusting a temperature of a sample, or any combination thereof. In some embodiments, a method can be used to identify a disease or condition, wherein a sample can be taken from a subject and applied to a device. In some embodiments, a computer can be used to at least partially compare a chromatogram in a subject having a disease to a chromatogram of a subject not having a disease, and determining independently a difference in a presence, an absence, a level, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some embodiments, a chromatogram can comprise a signature of a disease or a condition. In some embodiments, a result generated by a method can be evaluated by an algorithm using a processor or microprocessor. In some embodiments, an algorithm can comprise a trained algorithm, a machine learning algorithm, or any combination thereof using a processor or microprocessor. In some embodiments, a computer can comprise a memory, one or more executable instructions, one or more processors, and a graphical user interface. In some embodiments, a result can be communicated via a communication medium. In some embodiments, a compound of Table 1 identified by a method, or a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be selected as a therapeutic. In some embodiments, a method can comprise a method of diagnosis, of prognosis, a theranostic method, a health level information method, or any combination thereof. In some embodiments, a subject can display an altered profile, a level of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be at least partially increased or at least partially decreased. In some cases, a measurement and/or an analysis (e.g., comprising a subject’s metabolic profile) can be made one or more times over a period of 1 minute, 2 minutes, 3 minutes, 5 minutes, 10 minutes,
15 minutes, 20 minutes, 30 minutes, 1 hour, 2 hours, 6 hours, 12 hours, 24 hours, 2 days, 5 days,
1 week, 2 weeks, 1 month, 2 months, 6 months, 1 year, 2 years, 3 years, 5 years, 10 years, 20 years, less than 1 minute, from 1 minute to 2 minutes, from 2 minutes to 3 minutes, from 3 minutes to 5 minutes, from 5 minutes to 10 minutes, from 10 minutes to 15 minutes, from 15 minutes to 20 minutes, from 20 minutes to 30 minutes, from 30 minutes to 1 hour, from 1 hour to
2 hours, from 2 hours to 6 hours, from 6 hours to 12 hours, from 12 hours to 24 hours, from 1 day to 2 days, from 2 days to 5 days, from 2 days to 1 week, from 1 week to 2 weeks, from 2 weeks to 1 month, from 1 month to 2 months, from 2 months to 6 months, from 6 months to 1 year, from 1 year to 2 years, from 2 years to 3 years, from 3 years to 5 years, from 5 years to 10 years, from 10 years to 20 years, at least 1 minute, at least 2 minutes, at least 3 minutes, at least 5 minutes, at least 10 minutes, at least 15 minutes, at least 20 minutes, at least 30 minutes, at least 1 hour, at least 2 hours, at least 6 hours, at least 12 hours, at least 24 hours, at least 2 days, at least
5 days, at least 1 week, at least 2 weeks, at least 1 month, at least 2 months, at least 6 months, at least 1 year, at least 2 years, at least 3 years, at least 5 years, at least 10 years, at least 20 years, at most 1 minute, at most 2 minutes, at most 3 minutes, at most 5 minutes, at most 10 minutes, at most 15 minutes, at most 20 minutes, at most 30 minutes, at most 1 hour, at most 2 hours, at most
6 hours, at most 12 hours, at most 24 hours, at most 2 days, at most 5 days, at most 1 week, at most 2 weeks, at most 1 month, at most 2 months, at most 6 months, at most 1 year, at most 2 years, at most 3 years, at most 5 years, at most 10 years, at most 20 years. In some embodiments, a healthy subject’s metabolic profile can be recorded for at least 2 weeks. In some embodiments, a healthy subject’s profile can be recorded a plurality of times over a period of at least two weeks. In some embodiments, a metabolic profile of a subject can be unknown, wherein a healthy metabolic profile can be determined from factors comprising a subject’s genetic profile, a diseased metabolic profile, a healthy population profile, or any combination thereof. In some embodiments, a method can be performing a diagnostic test and administering a therapeutic comprising a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some embodiments, a diagnostic test can comprise a diagnostic test, a prognostic test or a theranostic test. In some embodiments, running a diagnostic test can comprise subjecting at least a portion of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof to a device comprising: a chromatography column, an eluant, a detector, and a mass spectrometer. [0009] Also disclosed herein, are methods of promoting or maintaining health in a subject. In some embodiments, a method can comprise: a.) administering to a subject a therapeutically effective amount of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof; b.) directly or indirectly at least partially increasing in a subject a level, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof; c.) directly or indirectly at least partially reducing in a subject a level, an activity, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof, or any combination of (a-c). In some embodiments, an administering can comprise administering an oral formulation. In some embodiments, an oral formulation ca be a dietary supplement. In some embodiments, a promoting or maintaining health can comprise promoting or maintaining: cardiovascular health, respiratory health, skeletal health, urogenital health, digestive health, muscular health, neural health, immune system health, liver health, endocrine health or any combination thereof.
[0010] Also disclosed herein are pharmaceutical compositions in unit dose form comprising, a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some embodiments, a composition in unit dose form can be placed in a closed container and placed in a room at about 23 degrees Celsius with a relative atmospheric humidity of about 50% for at least about 1 month. In some embodiments, at a end of a time period at least about 80% of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof remains. In some embodiments, a time period can be about 1 month to about 2 years.
[0011] Also disclosed herein, are methods of treating or preventing a disease or condition. In some embodiments, a method can comprise detecting a level of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof, from a sample of a subject having or suspected of having a disease or condition and comparing a level of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof, to a reference level and administering to a subject a therapeutically effective amount of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some embodiments, a method can be conducted at least in part on a diagnostic device, a screening device, or a diagnostic and a screening device comprising at least one of; a chromatography column, an eluant, a detector, and a mass spectrometer. In some embodiments, a chromatography column can comprise a gas chromatography column and an eluant can comprise a gas. In some embodiments, a diagnostic device, a screening device, or a diagnostic and a screening device can comprise a microelectromechanical system (MEMS). In some embodiments, a diagnostic device, a screening device, or a diagnostic and a screening device can comprise a breath collection tube. In some embodiments, an increase or decrease in concentration of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof, can indicate a treatment may be at least partially decreasing a presence of a disease, disease symptoms, or a combination thereof. In some embodiments, an increase or decrease in concentration of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can indicate a viral infection in a subject. In some embodiments, a diagnostic device can be operably linked to a database or may not linked to a database. In some embodiments, a database can comprise a profile pattern of different diseases. In some embodiments, a device compares a patient’s profile pattern with a database’s profile to identify a disease.
INCORPORATION BY REFERENCE
[0012] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. To the extent publications and patents or patent applications incorporated by reference contradict the disclosure contained in the specification, the specification is intended to supersede and/or take precedence over any such contradictory material.
DESCRIPTION OF THE DRAWINGS
[0013] The novel features of exemplary embodiments are set forth with particularity in the appended claims. A better understanding of the features and advantages will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of exemplary embodiments are utilized, and the accompanying drawings of which:
[0014] FIG. 1 shows a chromatogram of a patient with breast cancer, in accordance with embodiments.
[0015] FIG. 2 shows a chromatogram of a patient with kidney cancer, in accordance with embodiments.
[0016] FIG. 3A shows an overlay of chromatograms from gastric cancer patients’ blood, in accordance with embodiments. FIG. 3B, FIG. 3C, and FIG. 3D show data from analysis of individual gastric cancer patients’ blood, in accordance with embodiments. [0017] FIG. 4A shows an overlay of chromatograms from lung cancer patients’ blood, in accordance with embodiments. FIG. 4B, FIG. 4C, and FIG. 4D show data from analysis of individual lung cancer patients’ blood, in accordance with embodiments.
[0018] FIG. 5A shows an overlay of chromatograms from gastric cancer patients’ urine, in accordance with embodiments. FIG. 5B, FIG. 5C, and FIG. 5D show data from analysis of individual gastric cancer patients’ urine, in accordance with embodiments.
[0019] FIG. 6A shows an overlay of chromatograms from kidney cancer patients’ urine, in accordance with embodiments. FIG. 6B, FIG. 6C, and FIG. 6D show data from analysis of individual kidney cancer patients’ urine, in accordance with embodiments.
[0020] FIG. 7A shows an overlay of chromatograms from lung cancer patients’ urine, in accordance with embodiments. FIG. 7B, FIG. 7C, and FIG. 7D show data from analysis of individual lung cancer patients’ urine, in accordance with embodiments.
[0021] FIG. 8A shows an overlay of chromatograms from healthy control patients’ blood, in accordance with embodiments. FIG. 8B and FIG. 8C show data from analysis of individual healthy control patients’ blood, in accordance with embodiments.
[0022] FIG. 9A shows an individual breast cancer patient’s metabolite volatile organic compounds (VOC) profile. FIG. 9B shows an overlay of chromatograms of six breast cancer patients’ metabolite VOC profiles, in accordance with embodiments.
[0023] FIG. 10A shows an individual breast cancer patient’s metabolite volatile organic compounds (VOC) profile. FIG. 10B shows an overlay of chromatograms of five kidney cancer patients’ metabolite VOC profiles, in accordance with embodiments, in accordance with embodiments.
[0024] FIG. 11A shows an individual breast cancer patient’s metabolite volatile organic compounds (VOC) profile. FIG. 11B shows an overlay of chromatograms of five lymphoma patients’ metabolite VOC profiles, in accordance with embodiments, in accordance with embodiments, in accordance with embodiments. [0025] FIG. 12A shows an overlay of chromatograms from breast and kidney cancer patients, in accordance with embodiments. FIG. 12B, FIG. 12C, and FIG. 12D show data from analysis of individual renal cancer patients’ blood, in accordance with embodiments.
FIG. 12E, FIG. 12F, and FIG. 12G show data from analysis of individual breast cancer patients’ blood, in accordance with embodiments.
[0026] FIG. 13 shows an overlay of chromatograms from lung cancer and liver cancer patients, in accordance with embodiments.
[0027] FIG. 14 shows chromatograms from first thaw and second thaw analysis of kidney cancer patients’ urine, in accordance with embodiments.
[0028] FIG. 15 shows saliva chromatograms, in accordance with embodiments.
[0029] FIG. 16 shows chromatograms from four healthy volunteers’ breath (top row, second row) and a blank control (bottom left), in accordance with embodiments.
[0030] FIG. 17 shows a chromatogram from a cancer patient, in accordance with embodiments.
[0031] FIG. 18 shows a chromatogram from a cancer patient, in accordance with embodiments.
[0032] FIG. 19 shows a chromatogram from a lymphoma patient, in accordance with embodiments.
[0033] FIG. 20 shows a chromatogram from a lung cancer patient, in accordance with embodiments.
[0034] FIG. 21 shows a chromatogram from a prostate cancer patient, in accordance with embodiments.
[0035] FIG. 22 shows a chromatogram of a healthy control and cancer patients, in accordance with embodiments.
[0036] FIG. 23 shows a chromatogram of a healthy control and a lab standard, in accordance with embodiments. [0037] FIG. 24 shows a chromatogram of blood and urine samples from kidney cancer patients, in accordance with embodiments.
[0038] FIG. 25 shows a computer control system that is programmed or otherwise configured to implement methods provided herein.
[0039] FIG. 26 is a diagram showing a method and system as disclosed herein.
[0040] FIG. 27A-B shows a gas chromatography (GC) device for testing a sample, in accordance with embodiments.
[0041] FIG. 28A-B shows the GC device testing a sample on a subject, in accordance with embodiments.
DETAILED DESCRIPTION
[0042] Disclosed herein are methods for treating or preventing a disease or condition. Also disclosed herein are methods of detecting a compound from a subject suspected of having a disease or condition. Also disclosed herein are pharmaceutical compositions for treating or preventing a disease or condition. The methods described herein can be used with a chromatography device such as one or more chromatography devices as described in PCT US2017/027523. PCT/US2017/027523 is incorporated herein by reference in its entirety. Such devices, systems and methods can be used to monitor the presence or absence of one or more compounds, such as volatile organic compounds. Also incorporated herein by reference in their entirety are US Appln. No. 12/669,965, US Appln. No. 12/742,455, US Appln. No. 12/994,177, and US Appln. No. 13/520,193.
[0043] A device as described herein can be portable, easy to operate, stand alone or in a cluster, low cost, fast, remote control capable, network connected or otherwise, gas chromatography device. The device can use ambient air as well as other gases as a carrier gas to sample a compound, such as one or more Volatile Organic Compounds (VOCs) emission from any bodily fluid such as blood or urine, as well as any body parts such as skin, hair, nails, cells or whole tumor. In some cases, a device or method disclosed herein can comprise or comprise the use of a chemical that is not a VOC, e.g., for analysis of a sample from a subject and/or in the diagnosis of a condition (e.g., disease). In some cases, a device or method disclosed herein can comprise the use of a chemical that is not organic, e.g., for analysis of a sample from a subject and/or in the diagnosis of a condition (e.g., disease). For example, a device or method disclosed herein can comprise nitric oxide (NO) and/or nitrogen dioxide (N02) or the use thereof, e.g., for analysis of a sample from a subject and/or in the diagnosis of a condition (e.g., disease). Data produced by the gas chromatography device can detect as well as separate individual VOCs as biomarkers, measure a presence or absence or concentration one or more compounds, such as VOCs. Data produced can be fed into a software comprising one or more algorithms capable of using these data to identify a presence or an absence of a disease such as Parkinson’s, infectious diseases, cancer or others.
The disease identification may be not limited to use a certain biomarker or biomarkers, but these data can also be used to construct patterns produced by the gas chromatography device.
In some cases, these patterns can be used for disease screening, diagnostics, companion diagnostics, prognostics, theranostics, or any combination thereof.
[0044] In some cases, a system can be for analyzing a gas mixture. In some cases, the system can comprise: a filter; a trap; a chromatographic column; a detector; and a pump. The trap and the pump can be fluidly connected to form a first gas flow path. The filter, the trap, the chromatographic column, the detector, and the pump, can be fluidly connected to form a second gas flow path. In some cases, the detector and the pump can be fluidly connected to form a third gas flow path. In some cases, the chromatographic column can be a gas-solid adsorption chromatographic column. In some cases, the chromatographic column can be a gas-liquid gas chromatography column. In some cases, the trap further can comprise an adsorbent material. In some cases, the filter can be an activated charcoal filter. In some cases, the detector can be selected from the group consisting of a photo ionization detector, a mass spectrometer, a spectrophotometer, and a thermal conductivity detector. In some cases, the detector can be a photo ionization detector. In some cases, the pump can provide negative pressure. In some cases, the system can further comprise a housing. In some cases, the housing can be no larger than 216 cubic inches. A method can comprise a method of analyzing a chemical compound in a gas mix. The method can comprise directing flow of the gas mix through a trap to concentrate at least a quantity of the chemical compound; redirecting flow of the gas mix through a filter to provide a filtered gas flow to the trap; releasing at least a quantity of the concentrated chemical compound into the filtered gas flow; and analyzing at least a quantity of the released concentrated chemical compound. In some cases, the chemical compound can be an organic compound. In some cases, the organic compound can be a volatile organic compound. In some cases, analysis of at least a quantity of the released concentrated chemical compound can comprise running at least a quantity of the released concentrated chemical compound through a gas chromatography column. In some cases, the gas chromatography column can be a gas-solid adsorption chromatographic column. In some cases, the chromatographic column can be a gas-liquid gas chromatography column. In some cases, analysis of at least a quantity of the released concentrated chemical compound can comprise identifying the organic compound by a method selected from the group consisting of photo ionization, mass spectrometry, spectrophotometry, and thermal conductivity. In some cases, analysis of at least a quantity of the released concentrated chemical compound can further comprise quantifying the chemical compound. In some cases, the gas mix can be an environmental gas mix. In another embodiment, the gas mix can comprise gases exhaled or otherwise emitted by a living subject. In some cases, the gas mix can be air.
[0045] Unless otherwise indicated, open terms for example “contain,” “containing,” “include,” “including,” and the like mean comprising.
[0046] The singular forms “a”, “an”, and “the” are used herein to include plural references unless the context clearly dictates otherwise. Accordingly, unless the contrary is indicated, the numerical parameters set forth in this application are approximations that can vary depending upon the desired properties sought to be obtained.
[0047] As used herein, the term “about” or “approximately” can mean within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, e.g ., the limitations of the measurement system. For example, “about” can mean plus or minus 10%, per the practice in the art. Alternatively, “about” can mean a range of plus or minus 20%, plus or minus 10%, plus or minus 5%, or plus or minus 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, within 5-fold, or within 2-fold, of a value. Where particular values are described in the application and claims, unless otherwise stated the term “about” meaning within an acceptable error range for the particular value should be assumed. Also, where ranges and/or subranges of values are provided, the ranges and/or subranges can include the endpoints of the ranges and/or subranges.
[0048] The term “substantially” as used herein can refer to a value approaching 100% of a given value. In some cases, the term can refer to an amount that can be at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.9%, or 99.99% of a total amount. In some cases, the term can refer to an amount that can be about 100% of a total amount.
[0049] The term “homology” can refer to a % identity of a sequence to a reference sequence. As a practical matter, whether any particular sequence can be at least 50%, 60%, 70%, 80%, 85%, 90%, 92%, 95%, 96%, 97%, 98% or 99% identical to any sequence described herein (which can correspond with a particular nucleic acid sequence described herein), such particular polypeptide sequence can be determined conventionally using known computer programs such the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, Wis. 53711). When using Bestfit or any other sequence alignment program to determine whether a particular sequence is, for instance, 95% identical to a reference sequence, the parameters can be set such that the percentage of identity is calculated over the full length of the reference sequence and that gaps in homology of up to 5% of the total reference sequence are allowed.
[0050] The terms “administer,” “administering”, “administration,” and the like, as used herein, can refer to methods that can be used to enable delivery of compounds or compositions described herein, to the desired site of biological action. In some cases, delivery can include injection, inhalation, catheterization, gastrostomy tube administration, intravenous administration, intraosseous administration, ocular administration, otic administration, topical administration, transdermal administration, oral administration, rectal administration, nasal administration, intravaginal administration, intracavemous administration, transurethral administration, buccal administration, sublingual administration, or a combination thereof. Delivery can include direct application to the affect tissue or region of the body. Delivery can include a parenchymal injection, an intra-thecal injection, an intra ventricular injection, or an intra-ci sternal injection. A composition provided herein can be administered by any method. A method of administration can be by inhalation, intraarterial injection, intracerebroventricular injection, intraci sternal injection, intramuscular injection, intraorbital injection, intraparenchymal injection, intraperitoneal injection, intraspinal injection, intrathecal injection, intravenous injection, intraventricular injection, stereotactic injection, subcutaneous injection, epidural, or any combination thereof. Delivery can include parenteral administration (including intravenous, subcutaneous, intrathecal, intraperitoneal, intramuscular, intravascular or infusion), oral administration, inhalation administration, intraduodenal administration, rectal administration. In some embodiments, delivery can comprise a nanoparticle, a viral vector, a viral-like particle, a liposome, an exosome, an extracellular vesicle, a microrobot, a microneedle, an implant, or a combination thereof. In some cases, the implant can partially or fully release a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, or a salt thereof. In some cases, delivery can be from a device. In some instances, delivery can be administered by a pump, an infusion pump or a combination thereof. In some cases, delivery can be by a contact lens, an eye drop, a nasal spray, an ear drop, or any combination thereof. Delivery can include, a pill, an enema, an eye drop, an ear drop, a gas, a mouthwash, a toothpaste, a film, a tea, an elixir, a osmotic delivery system, a dietary supplement, a food, a beverage, a chewing gum, an oil, a lozenge, a mouth spray, a lip balm, a shampoo, a dental implant, a dental ware (e.g. tooth night guard), a suppository, a liquid, a powder, a solid, a membrane, an aerosol, or any combination thereof. In some cases, delivery can comprise an inhaler, a humidifier, a diffuser, a vape device, a bronchodilator, a face mask, a nasal cannula, a tent, a container at least partially surrounding the subjects head, a nebulizer, a jet nebulizer, an ultrasonic nebulizer, or a combination thereof. In some cases, an inhaler can comprise a metered-dose inhaler, a dry powder inhaler or any combination thereof. Delivery can include topical administration (such as a lotion, a cream, a patch, a gel, a spray, a drip, a liquid formulation, an ointment) to an external surface of a surface, such as a skin. In some cases, a patch can comprise a membrane, a microneedle patch, a single-layer drug in adhesive, a multi-layer drug in adhesive, a reservoir system, a matrix system, a vapour patch or any combination thereof. In some instances, a subject can administer the composition in the absence of supervision. In some instances, a subject can administer the composition under the supervision of a medical professional (e.g., a physician, nurse, physician’s assistant, orderly, hospice worker, etc.). In some cases, a medical professional can administer the composition. In some cases, a cosmetic professional can administer the composition.
[0051] The term “subject,” “host,” “individual,” and “patient” are as used interchangeably here-in to refer to animals, typically mammalian animals. Any suitable mammal can be administered a composition as described herein (such as an engineered oligonucleotide) or treated by a method as described herein. Non-limiting examples of mammals include humans, non-human primates (e.g., apes, gibbons, chimpanzees, orangutans, mon-keys, macaques, and the like), domestic animals (e.g., dogs and cats), farm animals (e.g., horses, cows, goats, sheep, pigs) and experimental animals (e.g., mouse, rat, rabbit, guinea pig). Mammals can be any age or at any stage of development, for example a mammal can be neonatal, infant, adolescent, adult or in utero. In some embodiments a mammal can be a human. Humans can be more than about: 1, 2, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 years of age. Humans can be less than about: 1, 2, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 years of age. A mammal can be male or female. In some embodiments a subject can be a human. In some embodiments, a subject may be suspected of having a disease or condition. The subject can be a patient, such as a patient being treated for a condition or a disease, such as a cancer, an infectious disease, or a neurological condition. The subject may be predisposed to a risk of developing a condition or a disease such as cancer. The subject may be in remission from a condition or a disease, such as a cancer patient. The subject may be healthy.
[0052] A cell can refer to any mammalian cell, typically a human cell. In some embodiments the human cell can be a skin cell, a cervical cell, a prostate cell, a stem cell, a bone cell, a blood cell, a muscle cell, a fat cell, a nerve cell, an endothelial cell, sperm cell, egg cell, cancer cell, barrier cell, hormone-secreting cell, exocrine-secretory cell, epithelial cell, oral cell, sensory transducer cell, autonomic neuron cell, peripheral neuron cell, central nervous neuron cell, secretory cell, barrier cell, muscle cell, cardiac muscle cell, white blood cell, germ cell, nurse cell, kidney cell, or any combination thereof. In some cases, the cell will be a cancer cell.
[0053] As used herein, treating a disease or condition can include one or more of: reducing the frequency, the severity, or any combination thereof, of a symptom. In some cases, treating a disease can comprise elimination of a symptom, elimination of an underlying cause, and improvement or remediation of damage. For example, treatment of cancer can include, for example, relieving the pain experienced by a subject suffering from cancer, or causing the regression or disappearance of cancer. Treating may also include: reduced malaise, cessation of cancer side effects, abatement of tumors, or any combination thereof. Treating can include administering a compound of Table 1 As used herein, “treating” of a urogenital disease can include one or more of: reducing the frequency, the severity of symptoms, the elimination of symptoms, the elimination of a symptoms underlying cause, and improvement or remediation of damage. For example, treatment of urogenital disease can include, for example, relieving pain experienced by a subject suffering from a kidney stone, or causing the regression of kidney stones. Treating may also include: reduction of malaise, cessation of urinary side effects, abatement of kidney damage, or any combination thereof. As used herein, treating of an infectious disease can include one or more of: reducing the frequency or severity of a symptom, elimination of a symptom, elimination of a symptom’s underlying cause, elimination of the infectious disease, and improvement or remediation of damage. For example, treatment of a viral infection can include, for example, relieving the cough experienced by a subject suffering from a viral infection or causing the regression, the disappearance, or any combination thereof of a viral infection. Treating may also include: reduction of malaise, cessation of viral infection side effects, abatement of a viral infection, or any combination thereof. Treatment can include administration to the subject in need thereof the compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or a salt thereof, or a pharmaceutical composition as described herein. Treatment can include one or more than one of the compounds of Table 1, the homologs thereof, the derivatives thereof, the esters thereof, the enantiomers thereof, the diastereomers thereof, the racemates thereof, or the salts thereof or any combination thereof delivered in a single dose or in multiple doses. As used herein, preventing a disease or a condition can include inhibiting a disease from occurring in a subject. [0054] A diagnostic test can comprise an imaging procedure, a blood count analysis, a tissue pathology analysis, a biomarker analysis, a biopsy, a magnetic resonance image procedure, a physical examination, a urine test, an ultrasonography procedure, a genetic test, a liver function test, a positron emission tomography procedure, a X-ray, serology, an angiography procedure, an electrocardiography procedure, an endoscopy, a diagnostic polymerase chain reaction test (PCR), a pap smear, a hematocrit test, a skin allergy test, a urine test, a colonoscopy, an enzyme-linked immunosorbent assay (ELISA), microscopy analysis, bone marrow examination, rapid diagnostic test, pregnancy test, organ function test, toxicology test, infectious disease test, bodily fluids test, or any combination thereof. In some cases, a diagnostic test can be comprised of the unit described herein. In some cases, the unit described herein diagnostic test can comprise a chromatography column, an eluant, a detector, a mass spectrometer, a microelectromechanical system (MEMS), a sample collection system, a breath collection tube or any combination thereof.
[0055] In some embodiments, a biomolecule disclosed herein can be a polypeptide, a protein, a lipid, a nucleic acid, a carbohydrate, a saccharide, a polysaccharide, a modified protein, a glycosylated protein, a modified lipid, a lipoprotein or any combination thereof.
[0056] In some embodiments, a compound disclosed herein can be a small molecule. In some cases, a small molecule can be a low molecular weight organic compound. In some embodiments the small molecule can be less than about: 10 Daltons, 50 Daltons, 100 Daltons,
200 Daltons, 300 Daltons, 400 Daltons, 500 Daltons, 600 Daltons, 700 Daltons, 800 Daltons,
900 Daltons, 1000 Daltons, 1100 Daltons, 1200 Daltons, 1300 Daltons, 1400 Daltons, or
1500 Daltons. In some embodiments the small molecule can be more than about: 10 Daltons,
50 Daltons, 100 Daltons, 200 Daltons, 300 Daltons, 400 Daltons, 500 Daltons, 600 Daltons,
700 Daltons, 800 Daltons, 900 Daltons, 1000 Daltons, 1100 Daltons, 1200 Daltons, 1300
Daltons, 1400 Daltons, or 1500 Daltons. In some embodiments, the small molecule may not be a polymer. In some embodiments, a small molecule can comprise, a carbon, a hydrogen, a nitrogen, an oxygen, a phosphorus, a sulfur, an organic atom, an inorganic atom, a plurality of molecular atoms, or any combination thereof. In some embodiments, a small molecule can bind to a biological molecule such as a protein, a nucleic acid, a polysaccharide, a lipid, or any combination thereof. In some cases, a small molecule can be an enzyme inhibitor, an enzyme activator, a cell signaling molecule, metabolite, a hormone, a drug, a pesticide, or any combination thereof. In some cases, a small molecule can bind to a biological channel, receptor, or any combination thereof. A small molecule can be a natural product (i.e. a sugar, a lipid, an amino acid, a fatty acid, a phenolic compound, an alkaloid or any combination thereof). In some cases, a small molecule can be a synthetic product. In some embodiments, a small molecule can be chemically stable, thermally stable, biologically stable or any combination thereof. In some embodiments a small molecule can bind to a single molecule.
In some instances, a small molecule may bind to multiple molecules. In some embodiments, a small molecule can have a half-life from about: 5 seconds to 10 seconds, 10 seconds to 30 seconds, 30 seconds to 60 seconds, 1 minute to 5 minutes, 5 minutes to 10 minutes, 10 minutes to 30 minutes, 30 minutes to 60 minutes, 1 hour to 2 hours, 2 hours to 4 hours, 4 hours to 8 hours, 8 hours to 12 hours, 12 hours to 16 hours, 16 hours to 24 hours, 1 day to 2 days, 2 days to 4 days, 4 days to 7 days, 1 week to 2 weeks, 2 weeks to 3 weeks, 3 weeks to 4 weeks, 1 month to 2 months, 2 months to 4 months, 4 months to 6 months, 6 months to 12 months or 1 year to about 2 years.
[0057] In some embodiments, the compounds disclosed in here can be an isomer. In some cases, an isomer can be a compound having the same molecular formula but a different structure. Isomers which may differ in configuration, conformation, or a combination thereof can be referred to as stereoisomers. An isomer can also be used to refer to an enantiomer. An enantiomer can be used to describe one of a pair of molecular isomers which can be a mirror image of each other and non-superimposable. A compound disclosed herein can have an asymmetric center and can exist in different enantiomeric and diastereomeric forms. The compounds disclosed herein can relate to the use of all optical isomers and stereoisomers of a compound disclosed herein, a mixture thereof, a pharmaceutical composition, and to a method of treatment that can contain them. In some cases, a compound disclosed herein can exist as a racemate. In some cases, a racemate can be a racemic mixture that can contain both enantiomers of a compound in equal amounts. In some cases, a compound disclosed herein can exist as a tautomer. In some cases, a compound disclosed herein can relate to the use of a tautomer, a mixture thereof, a pharmaceutical composition, and to a method of treatment that can contain a tautomer.
[0058] A homolog of a compound disclosed herein, can refer to a structure that can be similar to the parent structure. In some cases, a homolog can refer to similarity with respect to a structure, a function, a chemical composition or any combination thereof. In some cases, the homolog may not be readily derived synthetically from the parent structure. In some cases, the homolog can be derived synthetically from the parent structure. A derivative of a compound disclosed herein, can refer to a chemical substance related structurally a compound disclosed herein. A derivative can be made from the structurally-related parent compound in one or more steps. The general physical and chemical properties of a derivative can be similar to a parent compound.
[0059] In some embodiments, certain functional groups contained within the compounds of the present invention can be substituted for bioisosteric groups. In some cases, bioisosteric groups can have similar spatial or electronic requirements to the parent group but can exhibit differing or improved physicochemical or other properties. For example, some moieties are described in Patini et al., Chem. Rev, 1996, 96, 3147-3176.
[0060] In some embodiments, the compounds disclosed herein can be an ester. In some cases, an ester can be derived from a carboxylic acid. A carboxylic acid can contain the — COOH group, and in an ester the hydrogen in this group can be replaced by a hydrocarbon group.
The hydrocarbon group can be an alkyl group (e.g. methyl or ethyl, or one comprising a benzene ring.) In some cases, an ester can refer to a chemical moiety with formula — COOR, where R can represent an alkyl, a cycloalkyl, an aryl, a heteroaryl, or a heteroalicyclic. In some instances, an ester can be an alkoxycarbonyl group appended to the parent molecule on an available carbon atom. In some cases, an ester can be an alkoxycarbonyl group appended to the parent molecule on one or more available aryl groups, cycloalkyl groups, heterocycle groups, or any combination thereof. In some cases, a compound containing an ester can be a compound considered to be prodrug. In some embodiments, an ester group can comprise, pivaloyloxymethyl, acetoxymethyl, phthalidyl, indanyl and methoxymethyl, or any combination thereof.
[0061] A “salt” can include a pharmaceutically acceptable salt. Examples of pharmaceutically acceptable salts can include those salts prepared by reaction of a compound disclosed herein with a mineral, organic acid or inorganic base, such salts including, acetate, acrylate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, bisulfite, bitartrate, bromide, butyrate, butyn-l,4-dioate, camphorate, camphorsulfonate, caproate, caprylate, chlorobenzoate, chloride, citrate, cyclopentanepropionate, decanoate, digluconate, dihydrogenphosphate, dinitrobenzoate, dodecyl sulfate, ethanesulfonate, formate, fumarate, glucoheptanoate, glycerophosphate, glycolate, hemisulfate, heptanoate, hexanoate, hexyne- 1,6-dioate, hydroxybenzoate, g-hydroxybutyrate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, iodide, isobutyrate, lactate, maleate, malonate, methanesulfonate, mandelate. metaphosphate, methanesulfonate, methoxybenzoate, methylbenzoate, monohydrogenphosphate, 1-napthalenesulfonate, 2-napthalenesulfonate, nicotinate, nitrate, palmoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, pyrosulfate, pyrophosphate, propiolate, phthalate, phenyl acetate, phenylbutyrate, propanesulfonate, salicylate, succinate, sulfate, sulfite, succinate, suberate, sebacate, sulfonate, tartrate, thiocyanate, tosylate, undeconate, and xylenesulfonate. Further, a compound disclosed herein can be prepared as pharmaceutically acceptable salts formed by reacting the free base form of the compound with a pharmaceutically acceptable inorganic or organic acid, including, but not limited to, inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid metaphosphoric acid, and the like; and organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, Q-toluenesulfonic acid, tartaric acid, trifluoroacetic acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, arylsulfonic acid, methanesulfonic acid, ethanesulfonic acid, 1,2- ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 2- naphthalenesulfonic acid, 4-methylbicyclo-[2.2.2]oct-2-ene-l-carboxylic acid, glucoheptonic acid, 4,4’-methylenebis-(3-hydroxy-2-ene-l -carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid and muconic acid. Other acids, such as oxalic, while not in themselves pharmaceutically acceptable, can be employed in the preparation of salts useful as intermediates in obtaining a compound and/or a pharmaceutically acceptable acid addition salt. In some embodiments, a compound disclosed herein which can comprise a free acid group reacts with a suitable base, such as the hydroxide, carbonate, bicarbonate, sulfate, of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary or tertiary amine. Representative alkali or alkaline earth salts can include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts and the like. Illustrative examples of bases can include sodium hydroxide, potassium hydroxide, choline hydroxide, sodium carbonate, N+(Cl-4 alkyl)4, and the like. Representative organic amines useful for the formation of base addition salts can include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like. It may be understood that a compound disclosed herein can also include the quaternization of any basic nitrogen-containing groups they contain. In some embodiments, water or oil-soluble or dispersible products can be obtained by such quatemization. A compound disclosed herein can be prepared as pharmaceutically acceptable salts formed when an acidic proton present in the parent compound either can be replaced by a metal ion, for example an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base. In some embodiments, base addition salts can be also prepared by reacting the free acid form of a compound disclosed herein with a pharmaceutically acceptable inorganic or organic base, including, but not limited to organic bases such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like and inorganic bases such as aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like. In addition, the salt forms of the disclosed compounds can be prepared using salts of the starting materials or intermediates.
[0062] As used herein a sample can be a bodily fluid, a body odor, a compound extruded from the body, a bodily part, a blood sample, a plasma sample, a urine sample, a sweat sample, a breath sample, a skin sample, a buccal sample, a hair sample, a fecal sample, a tear sample, a nasal sample, a nasal mucus sample, a biopsy sample, a saliva sample, a sputum sample, or any combination thereof. In some cases, the compound extruded from the body can be a metabolite. In some cases, the compound extruded from the body can be an ester, a salt, or a combination thereof of the compound. In some cases, the compound extruded from the body can be a volatile chemical. In some cases, a sample can come from a body fluid. In some embodiments a body fluid can be, amniotic fluid, aqueous humour, vitreous humour, bile, whole blood, blood derivative, breast milk, cerebrospinal fluid, cerumen (earwax), chyle, chyme, endolymph and perilymph, exudates, nipple discharge. In some embodiments, a sample can be from a body odor. In some cases, a chemical extruded from the body can be a breath, a flatus, a burp, an odor of the body, recovered from an item that contacted a body, or any combination thereof. In some cases, a sample can be from a body part, an organ, a cell or any combination thereof. In some embodiments, a sample can be a live cell, or dead cell, a hair, a nail, or any combination thereof. In some cases, a sample can comprise, an additive, a preservative, an anticoagulant, a membrane, an antibacterial compound, an antifungal compound, an antiviral, a chemical reagent, an antimicrobial compound, a diluent, or any combination thereof. In some cases, a sample can be treated with an electromagnetic wave, an ultrasound, a light wave, a radiation, a laser, adjusting a temperature of the sample, or any combination thereof. In some cases, the sample can be heated. In some cases, the sample can be cooled (e.g. placed in a refrigerator, a freezer, or a combination thereof for a period of time.)
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000054_0002
Figure imgf000055_0001
[0063] Pharmaceutical composition [0064] In some embodiments, a pharmaceutical composition can comprise a small molecule. In some embodiments, a pharmaceutical composition may not comprise a small molecule. In some cases, a pharmaceutical composition can comprise a biomolecule. In some cases, a pharmaceutical composition can comprise both a small molecule and a biomolecule. In some embodiments a small molecule can be a compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof. In some cases, a pharmaceutical composition can comprise a molecule from Table 2. In some cases, a pharmaceutical composition can comprise a molecule from Table 2 and a compound of Table 1 a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof as described herein. A pharmaceutical composition can comprise a first active ingredient. The first active ingredient can comprise a compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof as described herein. A pharmaceutical composition can comprise more than about: 1 active ingredient, 2 active ingredients, 3 active ingredients, 4 active ingredients, 5 active ingredients, 6 active ingredients, 7 active ingredients, 8 active ingredients, 9 active ingredients, 10 active ingredients, 11 active ingredients ,12 active ingredients, 13 active ingredients, 14 active ingredients, 15 active ingredients, 20 active ingredients, 25 active ingredients, 30 active ingredients, 35 active ingredients, 40 active ingredients, 45 active ingredients, 50 active ingredients, 60 active ingredients, 70 active ingredients, 80 active ingredients ,90 active ingredients, 100 active ingredients, 125 active ingredients, 150 active ingredients, 175 active ingredients, 200 active ingredients, 250 active ingredients, 300 active ingredients, 400 active ingredients, 500 active ingredients, 1000 active ingredients, 5000 active ingredients, or 10,000 active ingredients. The pharmaceutical composition can be formulated in unit dose form. In some cases, a pharmaceutical composition can comprise 1 to 734 compounds of the compounds of Table 1, the homologs thereof, the derivatives thereof, the esters thereof, the enantiomers thereof, the diastereomers thereof, the racemates thereof, the salts thereof or any combination thereof. In some cases the pharmaceutical composition can comprise at least about 1, 2, 3, 4, 5, 6, 7, 8, 9,
10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34,
35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59,
60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84,
85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100 of the biomolecules in Table
2 or Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof. In some cases, the pharmaceutical composition can comprise at least 1 to about 734 molecules of Table 1, 50 to about 600 molecules of Table 1, 100 to about 400 molecules of Table 1, 10 to about 100 molecules of Table 1, 20 to about 60 molecules of Table 1, 15 to about 45 molecules of Table 1 or any combination thereof. The pharmaceutical composition can comprise a pharmaceutically acceptable excipient, diluent, or carrier. A pharmaceutical composition can comprise albuterol, beclomethasone, budesonide, formoterol, ciclesonide, fluticasone, fluticasone propionate, fluticasone furoate, fluticasone propionate, salmeterol, fluticasone furonate, umeclidinium, vilanterol, mometasone furoate, memetasone, a steroid, a bronchodilator. In some cases, a pharmaceutical composition can comprise a solid, a liquid, an aerosol, a gas, or a combination thereof.
[0065] A composition described herein can compromise an excipient. An excipient can comprise a pH agent (to minimize oxidation or degradation of a component of the composition), a stabilizing agent (to prevent modification or degradation of a component of the composition), a buffering agent (to enhance temperature stability), a solubilizing agent (to increase protein solubility), or any combination thereof. An excipient can comprise a surfactant, a sugar, an amino acid, an antioxidant, a salt, a non-ionic surfactant, a solubilizer, a triglyceride, an alcohol, or any combination thereof. An excipient can comprise sodium carbonate, acetate, citrate, phosphate, poly-ethylene glycol (PEG), human serum albumin
(HSA), sorbitol, sucrose, trehalose, polysorbate 80, sodium phosphate, sucrose, disodium phosphate, mannitol, polysorbate 20, histidine, citrate, albumin, sodium hydroxide, glycine, sodium citrate, trehalose, arginine, sodium acetate, acetate, HC1, disodium edetate, lecithin, glycerin, xanthan rubber, soy isoflavones, polysorbate 80, ethyl alcohol, water, teprenone, or any combination thereof. An excipient can be an excipient described in the Handbook of
Pharmaceutical Excipients, American Pharmaceutical Association (1986).
[0066] A carrier can refer to reagents, cells, compounds, materials, compositions, dosage forms, or any combination thereof that can be compatible with a small molecule or other agents that can be administered therapeutically. In some cases, a carrier can be suitable for use in contact with a tissue of a subject. In some cases, a carrier may not have a toxicity, an irritation, an allergic response, or any combination thereof. A carrier that may be suitable for use can include a liquid, an aerosol, a semi-solid (e.g., a gel), and a solid material (e.g., a pill, a patch, or an implant). In some cases, a carrier can be designed to resist degradation within the body (non-biodegradable) or they may be designed to degrade within the body
(biodegradable). A biodegradable material can further be bioresorbable or bioabsorbable. In some cases, a biodegradable material can be degraded and eliminated from the body by conversion into other materials or breakdown and elimination through natural pathways.
[0067] In some cases, a pharmaceutical composition can comprise isoprene, decanal, 4- methylbenzaldehyde, 3,7-dimethyloctan-3-ol, l-methyl-4(l -methyl -2 -propenyl)-benzene, 1- ethenyl-3 -ethyl-benzene, l-ethenyl-4-ethyl-benzene, 1-phenylbut-l-ene, octan-2-one, heptan-
3-one, hexan-3-one 5-methyl, butanoic acid 2-methyl-ethyl ester, propionic acid, 2- m ethoxy ethanol, acetaldehyde, propanal, n-propyl acetate, methyl methacrylate, styrene, 1,1- dipropoxypropane, a homolog of any of these, a derivative of any of these, an ester of any of these, a enantiomer of any of these, a diastereomer of any of these, a racemate of any of these, a salt of any of these, and any combination of any of these.
[0068] A pharmaceutical composition can comprise a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or a salt thereof. The pharmaceutical composition can be inhaled. The pharmaceutical composition can be encapsulated. The pharmaceutical composition can be formulated as a liquid, an aerosol or as a semi-solid, such as a gel, or a solid. The pharmaceutical composition can be formulated at a solution. The pharmaceutical composition can be formulated as an injectable. The pharmaceutical composition can be formulated as a subdermal implant. The pharmaceutical composition can be implantable. The pharmaceutical composition can be formulated for oral delivery. The pharmaceutical composition can be delivered by any method.
[0069] Administration
[0070] Administration or application of a composition disclosed herein can be performed for a treatment duration of at least about at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14,
15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39,
40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64,
65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89,
90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100 days consecutive or nonconsecutive days. In some cases, a treatment duration can be from about 1 to about 30 days, from about 2 to about 30 days, from about 3 to about 30 days, from about 4 to about 30 days, from about 5 to about 30 days, from about 6 to about 30 days, from about 7 to about 30 days, from about 8 to about 30 days, from about 9 to about 30 days, from about 10 to about 30 days, from about 11 to about 30 days, from about 12 to about 30 days, from about 13 to about 30 days, from about 14 to about 30 days, from about 15 to about 30 days, from about 16 to about 30 days, from about 17 to about 30 days, from about 18 to about 30 days, from about 19 to about 30 days, from about 20 to about 30 days, from about 21 to about 30 days, from about 22 to about 30 days, from about 23 to about 30 days, from about 24 to about 30 days, from about 25 to about
30 days, from about 26 to about 30 days, from about 27 to about 30 days, from about 28 to about 30 days, or from about 29 to about 30 days.
[0071] Administration or application of a composition disclosed herein can be performed for a treatment duration of at least about 1 week, at least about 1 month, at least about 1 year, at least about 2 years, at least about 3 years, at least about 4 years, at least about 5 years, at least about 6 years, at least about 7 years, at least about 8 years, at least about 9 years, at least about 10 years, at least about 15 years, at least about 20 years, or for life. Administration can be performed repeatedly over a lifetime of a subject, such as once a month or once a year for the lifetime of a subject. Administration can be performed repeatedly over a substantial portion of a subject’s life, such as once a month or once a year for at least about 1 year, 5 years, 10 years, 15 years, 20 years, 25 years, 30 years, or more.
[0072] Administration or application of composition disclosed herein can be performed at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 times a day. In some cases, administration or application of a composition disclosed herein can be performed continuously throughout an entire day, for example, when an implant can be used for administration. In some cases, an implant can release a composition continuously for about: 5 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 8 hours, 12 hours, 16 hours, 20 hours, 1 day, 2 days, 4 days, 7 days, 2 weeks, 4 weeks, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year or more. In some cases, an implant can have noncontinuous administration. In some cases, administration or application of composition disclosed herein can be performed at least 1, 2,
3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 21 times a week. In some cases, administration or application of composition disclosed herein can be performed at least 1, 2,
3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79,
80, 81, 82, 83, 84, 85, 86, 87, 88, 89, or 90 times a month.
[0073] In some cases, a composition can be administered/applied as a single dose or as divided doses. In some cases, the compositions described herein can be administered at a first time point and a second time point. In some cases, a composition can be administered such that a first administration can be administered before the other with a difference in administration time of 1 hour, 2 hours, 4 hours, 8 hours, 12 hours, 16 hours, 20 hours, 1 day,
2 days, 4 days, 7 days, 2 weeks, 4 weeks, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year or more.
[0074] In some cases, a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or a salt thereof when stored in a closed container placed in a room for a time period will remain at least about 80% of an initial amount of the compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or a salt thereof. In some cases, the compound of Table 1 will remain at least about 70% the initial amount. In some cases, the compound of Table 1 will remain at least about 90% the initial amount. In some cases, the compound of Table 1 will remain at least about: 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%. In some cases, the compound of Table 1 can be at least about 60% to about at least 80%. In some cases, the compound of Table 1 can be at least about 80% to at least about 99%. In some cases, the time period of storage can be at least 1 month. In some cases, the time period of storage can be at least about 3 months. In some cases, the time period of storage can be at least about 1 year. In some cases, the time period of storage can be at least about 1, 2, 4, 6, 8, 12, 18, 24, 36, 48 or 60 months. In some cases, the time period of storage can be at least about 1 month to about at least 1 year. In some cases, the time period of storage can be at least about 6 months to at least about 2 years. In some cases, the time period of storage can be at least about 1 month to at least about 5 years. [0075] The term “tissue” as used herein, can be any tissue sample. A tissue can be a tissue suspected or confirmed of having a disease or condition. A tissue can be a sample that may be substantially healthy, substantially benign, or otherwise substantially free of a disease or a condition. A tissue can be a tissue removed from a subject, such as a tissue biopsy, a tissue resection, an aspirate (such as a fine needle aspirate), a tissue washing, a cytology specimen, a bodily fluid, or any combination thereof. A tissue can comprise cancerous cells, tumor cells, non-cancerous cells, or a combination thereof. A tissue can comprise a blood sample (such as a cell-free DNA sample). A tissue can be a sample that may be genetically modified.
[0076] Diseases and conditions
[0077] Methods as described herein can be utilized for preventing, diagnosing and treating one or more diseases described herein. Methods can be utilized for preventing one or more diseases. Methods can be utilized for diagnosing one or more diseases. Methods can be utilized for treating, such as at least partially treating or substantially reducing one or more symptoms of one or more diseases.
[0078] In some embodiments, a disease can comprise a cancer, a tumor, an infectious disease, a skeletal disease, a urogenital disease, a digestive disease, a muscular disease, a neurological disease, an autoimmune disease, a respiratory disease, a cardiac disease, an endocrine disease, a liver disease, a hereditary disease, or any combination thereof.
[0079] In some cases, a disease or condition can comprise a cancer. In some cases, a cancer may comprise, a sarcoma, a carcinoma, a melanoma, a lymphoma, leukemia, blastoma, germ cell tumor, myeloma, or any combination thereof. In some cases, cancer may comprise a thyroid cancer, adrenal cortical cancer, anal cancer, aplastic anemia, bile duct cancer, bladder cancer, bone cancer, bone metastasis, central nervous system (CNS) cancers, peripheral nervous system (PNS) cancers, breast cancer, Castleman's disease, cervical cancer, childhood Non-Hodgkin's lymphoma, lymphoma, colon and rectum cancer, endometrial cancer, esophagus cancer, Ewing's family of tumors (e.g. Ewing's sarcoma), eye cancer, gallbladder cancer, gastrointestinal carcinoid tumors, gastrointestinal stromal tumors, gestational trophoblastic disease, hairy cell leukemia, Hodgkin's disease, Kaposi's sarcoma, kidney cancer, laryngeal and hypopharyngeal cancer, acute lymphocytic leukemia, acute myeloid leukemia, children's leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, liver cancer, lung cancer, lung carcinoid tumors, Non-Hodgkin's lymphoma, male breast cancer, malignant mesothelioma, multiple myeloma, myelodysplastic syndrome, myeloproliferative disorders, nasal cavity and paranasal cancer, nasopharyngeal cancer, neuroblastoma, oral cavity and oropharyngeal cancer, osteosarcoma, ovarian cancer, pancreatic cancer, penile cancer, pituitary tumor, prostate cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sarcoma (adult soft tissue cancer), melanoma skin cancer, non-melanoma skin cancer, stomach cancer, testicular cancer, thymus cancer, uterine cancer (e.g. uterine sarcoma), vaginal cancer, vulvar cancer, or Waldenstrom's macroglobulinemia.
[0080] A condition or a disease, as disclosed herein, can include hyperproliferative disorders. Malignant hyperproliferative disorders can be stratified into risk groups, such as a low risk group and a medium-to-high risk group. Hyperproliferative disorders can include but may not be limited to cancers, hyperplasia, or neoplasia. In some cases, the hyperproliferative cancer can be breast cancer such as a ductal carcinoma in duct tissue of a mammary gland, medullary carcinomas, colloid carcinomas, tubular carcinomas, and inflammatory breast cancer; ovarian cancer, including epithelial ovarian tumors such as adenocarcinoma in the ovary and an adenocarcinoma that has migrated from the ovary into the abdominal cavity; uterine cancer; cervical cancer such as adenocarcinoma in the cervix epithelial including squamous cell carcinoma and adenocarcinomas; prostate cancer, such as a prostate cancer selected from the following: an adenocarcinoma or an adenocarcinoma that has mi-grated to the bone; pancreatic cancer such as epithelioid carcinoma in the pancreatic duct tissue and an adenocarcinoma in a pancreatic duct; bladder cancer such as a transitional cell carcinoma in urinary bladder, urothelial carcinomas (transitional cell carcinomas), tumors in the urothelial cells that line the bladder, squamous cell carcinomas, adenocarcinomas, and small cell cancers; leukemia such as acute myeloid leukemia (AML), acute lymphocytic leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, hairy cell leukemia, myelodysplasia, myeloproliferative disorders, acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), mastocytosis, chronic lymphocytic leukemia (CLL), multiple myeloma (MM), and myelodysplastic syndrome (MDS); bone cancer; lung cancer such as non-small cell lung cancer (NSCLC), which may be divided into squamous cell carcinomas, adenocarcinomas, and large cell undifferentiated carcinomas, and small cell lung cancer; skin cancer such as basal cell carcinoma, melanoma, squamous cell carcinoma and actinic keratosis, which may be a skin condition that sometimes develops into squamous cell carcinoma; eye retinoblastoma; cutaneous or intraocular (eye) melanoma; primary liver cancer (cancer that begins in the liver); kidney cancer; autoimmune deficiency syndrome
(AIDS)-related lympho-ma such as diffuse large B-cell lymphoma, B-cell immunoblastic lymphoma and small non-cleaved cell lymphoma; Kaposi's Sarcoma; viral-induced cancers including hepatitis B virus (HBV), hepatitis C virus (HCV),and hepatocellular carcinoma; human lymphotropic virus-type 1 (HTLV-1) and adult T-cell leukemia/lymphoma; and human papilloma virus (HPV) and cervical cancer; central nervous system (CNS) cancers such as primary brain tumor, which includes gliomas (astrocytoma, anaplastic astrocytoma, or glioblastoma multiforme), oligodendrogliomas, ependymomas, meningiomas, lymphomas, schwannomas, and medulloblastomas; peripheral nervous system (PNS) cancers such as acoustic neuromas and malignant peripheral nerve sheath tumors (MPNST) including neurofibromas and schwannomas, malignant fibrous cytomas, malignant fibrous histiocytomas, malignant meningiomas, malignant mesotheliomas, and malignant mixed
Miillerian tumors; oral cavity and oropharyngeal cancer such as hypopharyngeal cancer, laryngeal cancer, nasopharyngeal cancer, and oropharyngeal cancer; stomach cancer such as lymphomas, gastric stromal tumors, and carcinoid tumors; testicular cancer such as germ cell tumors (GCTs), which include seminomas and nonseminomas, and gonadal stromal tumors, which include Leydig cell tumors and Sertoli cell tumors; thymus cancer such as to thymomas, thymic carcinomas, Hodgkin disease, non-Hodgkin lymphomas carcinoids or carcinoid tumors; rectal cancer; and colon cancer. In some cases, the diseases stratified, classified, characterized, or diagnosed by the methods of the present disclosure include but may not be limited to thyroid disorders such as for example benign thyroid disorders including but not limited to follicular adenomas, Hurthle cell adenomas, lymphocytic thyroiditis, and thyroid hyperplasia. In some cases, the diseases stratified, classified, characterized, or diagnosed by the methods of the present disclosure include but may not be limited to malignant thyroid disorders such as for example follicular carcinomas, follicular variant of papillary thyroid carcinomas, medullary carcinomas, and papillary carcinomas.
[0081] In some cases, urogenital disease can comprise benign prostate hyperplasia, urinary incontinence, kidney stones, kidney disease, erectile dysfunction, interstitial cystitis, prostatitis, overactive bladder, endometriosis, hypospadias, interstitial cystitis, posterior urethral valve, undescended testes, ureteropelvic junction obstruction, vesicoureteral reflux, or any combination thereof.
[0082] In some embodiments, a digestive disease can comprise irritable bowel syndrome, celiac disease, gastroesophageal reflux disease, Crohn’s disease, gastroparesis, gas, lactose intolerance, diarrhea, acid reflux, constipation, Hirschsprung disease, celiac disease, gallstones, abdominal adhesions, Barrett’s esophagus, bowl control problems, colon polyps, diverticulosis, food poisoning, gastritis, gastroparesis, GI bleeding, hemorrhoids, indigestion, hernia, microscopic colitis, pancreatitis, peptic ulcers, proctitis, short bowel syndrome, ulcerative colitis, Whipple disease, Zollinger-Ellison syndrome, or any combination thereof. [0083] In some embodiments a respiratory disease can comprise asthma, chronic obstructive pulmonary disease, chronic bronchitis, emphysema, cystic fibrosis, plural effusion, pneumonia, obstructive condition, alpha- 1 antitrypsin deficiency, bronchiectasis, pulmonary fibrosis, sarcoidosis, or any combination thereof.
[0084] In some embodiments, a cardiac disease can comprise congenital heart disease, arrhythmia, coronary artery disease, cardiac arrest, peripheral artery disease, dilated cardiomyopathy, myocardial infarction, stroke, structural heart disease, congestive heart failure, high blood pressure, mitral regurgitation, pulmonary stenosis, or any combination thereof.
[0085] In some embodiments, an endocrine disease can comprise type 2 diabetes, hypothyroidism, hyperthyroidism, low testosterone, obesity, Addison’s disease, Cushing’s syndrome, Graves’ disease, thyroiditis, prolactinoma, type 1 diabetes, type 2 diabetes, osteoporosis, thyroid cancer, gestational diabetes, growth hormone deficiency, Hashimoto’s thyroiditis, hyperglycemia, hyperparathyroidism, hypoglycemia, hypoparathyroidism, hypothyroidism, menopause, obesity, pre-diabetes, thyroid nodules, or any combination thereof.
[0086] In some embodiments, a liver disease can comprise cirrhosis, fibrosis, hemochromatosis, Wilson’s disease, Alagille syndrome, alcohol -related liver disease, alpha-1 antitrypsin deficiency, autoimmune hepatitis, benign liver tumor, biliary atresia, Crigler- Najjar Syndrome, Galactosemia, Gilbert Syndrome, Hemochromatosis, Hepatic Encephalopathy, Hepatitis A, Hepatitis B, Hepatitis C, hepatorenal syndrome, intrahepatic cholestasis of pregnancy, lysosomal acid lipase deficiency, liver cysts, newborn jaundice, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, primary biliary cholangitis, primary sclerosing cholangitis, progressive familial intrahepatic cholestasis, Reye syndrome, type 1 glycogen storage disease or any combination thereof.
[0087] In some cases, a disease or condition can comprise a skeletal disease can comprise osteoarthritis, osteoporosis, Pagets disease of bone, back pain, bursitis, fibromyalgia, fibrous dysplasia, growth plate injuries, heritable disorders of connective tissue, osteonecrosis, scoliosis, shoulder problems, hip problems, knee problems, joint problems, spinal stenosis, tendinitis, bone breaks, bone spurs, osteopetrosis, or any combination thereof.
[0088] In some cases, a disease or condition can comprise a neurological disorder. In some cases, a neurological disorder may comprise Acquired Epileptiform Aphasia, Acute
Disseminated Encephalomyelitis, Adrenoleukodystrophy, Agenesis of the corpus callosum,
Agnosia, Aicardi syndrome, Alexander disease, Alpers' disease, Alternating hemiplegia,
Alzheimer's disease, Amyotrophic lateral sclerosis (see Motor Neuron Disease),
Anencephaly, Angelman syndrome, Angiomatosis, Anoxia, Aphasia, Apraxia, Arachnoid cysts, Arachnoiditis, Arnold-Chiari malformation, Arteriovenous mal-formation, Asperger's syndrome, Ataxia Telangiectasia, Attention Deficit Hyperactivity Disorder, Autism, Auditory processing disorder, Autonomic Dysfunction, , Back Pain, Batten disease, Bechet’s disease,
Bell's palsy, Benign Essential Blepharospasm, Benign Focal Amyotrophy, Benign
Intracranial Hypertension, Bilateral frontoparietal polymicrogyria, Binswanger's disease,
Blepharo-spasm, Bloch-Sulzberger syndrome, Brachial plexus injury, Brain abscess, Brain damage, Brain in-jury, Brain tumor, Brown-Sequard syndrome, Canavan disease, Carpal tunnel syndrome (CTS), Causalgia, Central pain syndrome, Central pontine myelinolysis,
Centronuclear myopathy, Cephalic disorder, Cerebral aneurysm, Cerebral arteriosclerosis,
Cerebral atrophy, Cerebral gigantism, Cerebral palsy, Charcot-Marie-Tooth disease, Chiari malformation, Chorea, Chronic inflammatory de-myelinating polyneuropathy (CIDP),
Chronic pain, Chronic regional pain syndrome, Coffin Lowry syndrome, Coma, including
Persistent Vegetative State, Congenital facial diplegia, Corticobasal degeneration, Cranial arteritis, Craniosynostosis, Creutzfeldt-Jakob disease, Cumulative trauma disorders,
Cushing's syndrome, Cytomegalic inclusion body disease (CIBD), Cytomegalovirus
Infection, , Dandy-Walker syndrome, Dawson disease, De Morsier's syndrome, Dejerine-
Klumpke palsy, Dejerine-Sottas disease, Delayed sleep phase syndrome, Dementia,
Dermatomyositis, Neurological Dyspraxia, Diabetic neuropathy, Diffuse sclerosis, Dysautonomia, Dyscalculia, Dysgraphia, Dyslexia, Dystonia, , Early infantile epileptic encephalopathy, Empty sella syndrome, Encephalitis, Encephalocele, Encephalotrigeminal angiomatosis, Encopresis, Epilepsy, Erb's palsy, Erythromelalgia, Essential tremor, Fabry's disease, Fahr's syndrome, Fainting, Familial spastic paralysis, Febrile seizures, Fisher syndrome, Friedreich's ataxia, FART Syndrome, Gaucher's disease, Gerstmann's syndrome,
Giant cell arteritis, Giant cell inclusion disease, Globoid cell Leukodystrophy, Gray matter heterotopia, Guillain-Barre syndrome, HTLV-1 associated myelopathy, Hallervorden-Spatz disease, Head injury, Headache, Hemifacial Spasm, Hereditary Spastic Paraplegia,
Heredopathia atactica polyneuritiformis, Herpes zoster oticus, Herpes zoster, Hirayama syndrome, Holoprosencephaly, Huntington's disease, Hydranencephaly, Hydrocephalus,
Hypercortisolism, Hypoxia, Immune-Mediated encephalomyelitis, Inclusion body myositis,
Incontinentia pigmenti, Infantile phytanic acid storage disease, Infantile Refsum disease,
Infantile spasms, Inflammatory myopathy, Intracranial cyst, Intracranial hypertension,
Joubert syndrome, Kearns-Sayre syndrome, Kennedy disease, Kinsboume syndrome, Klippel
Feil syndrome, Krabbe disease, Kugelberg-Welander disease, Kuru, Lafora disease, Lambert-
Eaton myasthenic syndrome, Landau-Kleffner syndrome, Lateral medullary (Wallenberg) syndrome, Learning disabilities, Leigh's disease, Lennox-Gastaut syndrome, Lesch-Nyhan syndrome, Leukodystrophy, Lewy body dementia, Lissencephaly, Locked-In syn-drome, Lou
Gehrig's disease, Lumbar disc disease, Lyme disease - Neurological Sequelae, Macha-do-
Joseph disease (Spinocerebellar ataxia type 3), Macrencephaly, Maple Syrup Urine Disease,
Megalencephaly, Melkersson-Rosenthal syndrome, Menieres disease, Meningitis, Menkes disease, Metachromatic leukodystrophy, Microcephaly, Migraine, Miller Fisher syndrome,
Mini-Strokes, Mitochondrial Myopathies, Mobius syndrome, Monomelic amyotrophy, Motor
Neuron Disease, Motor skills disorder, Moyamoya disease, Mucopolysaccharidoses, Multi-
Infarct Dementia, Multi-focal motor neuropathy, Multiple sclerosis, Multiple system atrophy,
Muscular dystrophy, Myalgic encephalomyelitis, Myasthenia gravis, Myelinoclastic diffuse sclerosis, Myoclonic Encephalopathy of infants, Myoclonus, Myopathy, Myotubular myopathy, Myotonia congenita, Narcolepsy, Neurofibromatosis, Neuroleptic malignant syndrome, Neurological manifestations of AIDS, Neurological sequelae of lupus,
Neuromyotonia, Neuronal ceroid lipofuscinosis, Neuronal migration disorders, Niemann-Pick disease, Non 24-hour sleep-wake syndrome, Nonverbal learning disorder, O'Sullivan
McLeod syndrome, Occipital Neuralgia, Occult Spinal Dysraphism Sequence, Ohtahara syndrome, Olivopontocerebellar atrophy, Opsoclonus myoclonus syndrome, Optic neuritis,
Orthostatic Hypotension, Overuse syndrome, Palinopsia, Paresthesia, Parkinson's disease,
Paramyotonia Con-genita, Paraneoplastic diseases, Paroxysmal attacks, Parry-Romberg syndrome, Rombergs Syndrome, Pelizaeus-Merzbacher disease, Periodic Paralyses,
Peripheral neuropathy, Persistent Vegetative State, Pervasive neurological disorders, Photic sneeze reflex, Phytanic Acid Storage disease, Pick's disease, Pinched Nerve, Pituitary
Tumors, PMG, Polio, Polymicrogyria, Polymyositis, Porencephaly, Post-Polio syndrome,
Postherpetic Neuralgia (PHN), Postinfectious Encephalomyelitis, Postural Hypotension,
Prader-Willi syndrome, Primary Lateral Sclerosis, Prion diseases, Progressive Hemifacial
Atrophy also known as Rombergs Syndrome, Progressive multifocal leukoencephalopathy,
Progressive Sclerosing Poliodystrophy, Progressive Supranuclear Palsy, Pseudotumor cerebri, Ramsay-Hunt syndrome (Type I and Type II), Rasmussen's encephalitis, Reflex sympathetic dystrophy syndrome, Refsum disease, Repetitive motion disorders, Repetitive stress injury, Restless legs syndrome, Retrovirus-associated myelopathy, Rett syndrome,
Reye's syndrome, Rombergs Syndrome, Rabies, Saint Vitus dance, Sandhoff disease,
Schytsophrenia, Schilder's disease, Schizencephaly, Sensory Integration Dysfunction,
Septooptic dysplasia, Shaken baby syndrome, Shingles, Shy-Drager syndrome, Sjogren's syndrome, Sleep apnea, Sleeping sickness, Snatiation, Sotos syndrome, Spasticity, Spina bifida, Spinal cord injury, Spinal cord tumors, Spinal muscular atrophy, Spinal stenosis,
Steele-Richardson-Olszewski syndrome, see Progressive Supranuclear Palsy, Spinocerebellar ataxia, Stiff-person syndrome, Stroke, Sturge-Weber syndrome, Subacute sclerosing panencephalitis, Subcortical arteriosclerotic encephalopathy, Superficial siderosis, Syden ham's chorea, Syncope, Synesthesia, Syringomyelia, Tardive dyskinesia, Tay-Sachs disease, Temporal arteritis, Tethered spinal cord syndrome, Thomsen disease, Thoracic outlet syndrome, Tic Douloureux, Todd's paralysis, Tourette syndrome, Transient ischemic attack, Transmissible spongiform encephalopathies, Transverse myelitis, Traumatic brain injury, Tremor, Trigeminal neuralgia, Tropical spastic paraparesis, Trypanosomiasis, Tuberous sclerosis, Vasculitis including temporal arteritis, Von Hippel-Lindau disease (VHL), Viliuisk Encephalomyelitis (VE), Wallenberg's syn-drome, Werdnig-Hoffman disease, West syndrome, Whiplash, Williams syndrome, Wilson's disease, X-Linked Spinal and Bulbar Muscular Atrophy, and Zellweger syndrome. Neurological conditions can comprise movement disorders, for example multiple system atrophy (MSA).
[0089] In some cases, a disease or condition may comprise an autoimmune disease. In some cases, an autoimmune disease may comprise acute disseminated encephalomyelitis (ADEM), acute necrotizing hemorrhagic leukoencephalitis, Addison's disease, agammaglobulinemia, allergic asthma, allergic rhinitis, alopecia areata, amyloidosis, ankylosing spondylitis, anti- GBM/anti-TBM nephritis, antiphospholipid syndrome (APS), autoimmune aplastic anemia, autoimmune dysautonomia, autoimmune hepatitis, autoimmune hyperlipidemia, autoimmune immunodeficiency, autoimmune inner ear disease (AIED), autoimmune myocarditis, autoimmune pancreatitis, autoimmune retinopathy, autoimmune thrombocytopenic purpura (ATP), autoimmune thyroid disease, axonal & neuronal neuropathies, Balo disease, Bechet’s disease, bullous pemphigoid, cardiomyopathy, Castlemen disease, celiac sprue (non-tropical), Chagas disease, chronic fatigue syndrome, chronic inflammatory demyelinating polyneuropathy (CIDP), chronic recurrent multifocal ostomyelitis (CRMO), Churg-Strauss syndrome, cicatricial pemphigoid/benign mucosal pemphigoid, Crohn's disease, Cogan's syndrome, cold agglutinin disease, congenital heart block, coxsackie myocarditis, CREST disease, essential mixed cryoglobulinemia, demyelinating neuropathies, dermatomyositis,
Devic's disease (neuromyelitis optica), discoid lupus, Dressler's syndrome, endometriosis, eosinophillic fasciitis, erythema nodosum, experimental allergic encephalomyelitis, Evan's syndrome, fibromyalgia, fibrosing alveolitis, giant cell arteritis (temporal arteritis), glomerulonephritis, Goodpasture's syn-drome, Grave's disease, Guillain-Barre syndrome,
Hashimoto's encephalitis, Hashimoto's thyroiditis, hemolytic anemia, Henock-Schoniein purpura, herpes gestationis, hypogammaglobulinemia, idiopathic thrombocytopenic purpura
(ITP), IgA nephropathy, immunoregulatory lipoproteins, inclusion body myositis, insulin- dependent diabetes (type 1), interstitial cystitis, juvenile arthritis, juvenile diabetes, Kawasaki syndrome, Lambert-Eaton syndrome, leukocytoclastic vasculitis, lichen planus, lichen sclerosis, ligneous conjunctivitis, linear IgA disease (LAD), Lupus (SLE), Lyme dis-ease,
Meniere's disease, microscopic polyangitis, mixed connective tissue disease (MCTD),
Mooren's ulcer, Mucha-Habermann disease, multiple sclerosis, myasthenia gravis, myositis, narcolepsy, neuromyelitis optica (Devic's), neutropenia, ocular cicatricial pemphigoid, optic neuritis, palindromic rheumatism, PANDAS (Pediatric Autoimmune Neuropsychiatric
Disorders Associated with Streptococcus), paraneoplastic cerebellar degeneration, paroxysmal nocturnal hemoglobinuria (PNH), Parry Romberg syndrome, Parsonnage-Turner syndrome, pars plantis (peripheral uveitis), pemphigus, peripheral neuropathy, perivenous encephalomyelitis, pernicious anemia, POEMS syndrome, polyarteritis nodosa, type I, II &
III autoimmune polyglandular syndromes, polymyalgia rheumatic, polymyositis, postmyocardial infarction syndrome, postpericardiotomy syndrome, progesterone dermatitis, primary biliary cirrhosis, primary sclerosing cholangitis, psoriasis, psoriatic arthritis, idiopathic pulmonary fibrosis, pyoderma gangrenosum, pure red cell aplasis, Raynaud's phenomena, reflex sympathetic dystrophy, Reiter's syndrome, relapsing polychondritis, restless legs syndrome, retroperitoneal fibrosis, rheumatic fever, rheumatoid arthritis, sarcoidosis, Schmidt syn-drome, scleritis, scleroderma, Slogren's syndrome, sperm and testicular autoimmunity, stiff person syndrome, subacute bacterial endocarditis (SBE), sympathetic ophthalmia, Takayasu's arteritis, temporal arteritis/giant cell arteries, thrombocytopenic purpura (TPP), Tolosa-Hunt syndrome, transverse myelitis, ulcerative colitis, undifferentiated connective tissue disease (UCTD), uveitis, vasculitis, vesiculobullous dermatosis, vitiligo or Wegener's granulomatosis or , chronic active hepatitis, primary biliary cirrhosis, cadilated cardiomyopathy, myocarditis, autoimmune polyendocrine syndrome type
I (APS-I), cystic fibrosis vasculitides, acquired hypoparathyroidism, coronary artery disease, pemphigus foliaceus, pemphigus vulgaris, Rasmussen encephalitis, autoimmune gastritis, insulin hypoglycemic syndrome (Hirata disease), Type B insulin resistance, acanthosis, systemic lupus erythematosus (SLE), pernicious anemia, treatment-resistant Lyme arthritis, polyneuropathy, demyelinating diseases, atopic dermatitis, autoimmune hypothyroidism, vitiligo, thyroid associated ophthalmopathy, autoimmune coeliac disease, ACTH deficiency, dermatomyositis, Sjogren syndrome, systemic sclerosis, progressive systemic sclerosis, morphea, primary antiphospholipid syndrome, chronic idiopathic urticaria, connective tissue syndromes, necrotizing and crescentic glomerulonephritis (NCGN), systemic vasculitis,
Raynaud syndrome, chronic liver disease, visceral leishmaniasis, autoimmune Cl deficiency, membrane proliferative glomerulonephritis (MPGN), prolonged coagulation time, immunodeficiency, atherosclerosis, neuronopathy, paraneoplastic pemphigus, paraneoplastic stiff man syndrome, paraneoplastic encephalomyelitis, subacute autonomic neuropathy, cancer-associated retinopathy, paraneoplastic opsoclonus myoclonus ataxia, lower motor neuron syndrome and Lambert-Eaton myasthenic syndrome.
[0090] In some cases, a disease or a condition may comprise an infectious disease. In some cases, an infectious disease can comprise a viral infection, a bacterial infection, a parasitic infection, a fungal infection or any combination thereof. In some cases, an infectious disease can comprise, AIDS, anthrax, botulism, brucellosis, chancroid, chlamydial infection, cholera, coccidioidomycosis, cryptosporidiosis, cyclosporiasis, dipheheria, ehrlichiosis, arboviral encephalitis, enterohemorrhagic Helicobacter pylori, Staphylococcus aureus, Mycobacterium tuberculosis, Escherichia coli, Campylobacter jejuni, Salmonella, Klebsiella pneumonia,
Streptococcus pyogenes, Neisseria gonorrhoeae, Treponema pallidum, Borrelia burgdorferi,
Neisseria meningitidis, Chlamydia trachomatis, Enterobacteriaceae, Vibrio sp., giardiasis,
Enterobius vermicularis, Ancylostoma duodenale, Necator americanus, Entamoeba histolytica, Trypanosoma cruzi, Cyclospora cayetanenensis, Haemophilus influenza, Hansen's disease (Leprosy), trichomoniasis, ascariasis, taenia infection, lymphatic filariasis, African sleeping sickness, onchocerciasis, leishmaniasis, fungal nail infections, vaginal candidiasis, ringworm, Candida infections, blastomycosis, coccidioidomycosis, Cryptococcus gattii,
Candida auris, Histoplasmosis, paracoccidioidomycosis, aspergillosis, candidiasis,
Cryptococcus neoformans, pneumoncystis pneumona, talaromycosis, mucormycosis, fungal eye infection, mycetoma, sporotrichosis, Tinea versicolor, ringworm, hantavirus pulmonary syndrome, hemolytic uremic syndrome, hepatitis A, hepatitis B, hepatitis C, human immunodeficiency virus (HIV), legionellosis, listeriosis, Lyme disease, malaria, measles.
Meningococcal disease, mumps, pertussis (whooping cough), plague, paralytic poliomyelitis, psittacosis, Q fever, rabies, rocky mountain spotted fever, rubella, congenital rubella syndrome , shigellosis, smallpox, rotavirus, norovirus, streptococcal disease (invasive group
A), streptococcal toxic shock syndrome, streptococcus pneumonia, syphilis, tetanus, toxic shock syndrome, trichinosis, tuberculosis, tularemia, typhoid fever, Chikungunya virus, varicella, yellow fever, variant Creutzfeldt-Jakob disease (vCJD), Ebola hemorrhagic fever,
Echinococcosis, enterovirus, herpes simplex 1, herpes simplex 2, varicella zoster virus, dengue virus, Hendra virus, Nipah virus, human monkey-pox, influenza, influenza A, influenza B, H5N1, lassa fever, Margurg hemorrhagic fever, Nipah virus, O'nyong fever, Rift valley fever, Herpes, SARS-CoV-2 (COVID-19), SARS-CoV (SARS), MERS-CoV (MERS),
229E coronavirus, NL63 coronavirus, OC43 coronavirus, CoV-HKUl (HKU1), alpha coronavirus, beta coronavirus, Venezuelan equine encephalitis and West Nile virus. In some cases, an infectious disease can be a drug resistant infection. In some cases, an infectious disease can be a multi-drug resistant infection.
[0091] Conditions or diseases of the present disclosure can include a genetic disorder. In some embodiments, a genetic disorder can be a hereditary disease. In some cases, a heredity disease can be inherited mendelian disorders, peroxisomal disorders, mitochondrial disorders, cystic fibrosis, sickle-cell anemia, Marfan syndrome, Huntington’s disease, kidney disease, Tay-Sachs disease, phenylketonuria, hemophilia, or any combination thereof. A genetic disorder may be an illness caused by abnormalities in genes or chromosomes. Genetic disorders can be grouped into two categories: single gene disorders and multifactorial and polygenic (complex) disorders. A single gene disorder can be the result of a single mutated gene. Inheriting a single gene disorder can include but not be limited to autosomal dominant, autosomal recessive, X-linked dominant, X-linked recessive, Y-linked and mitochondrial inheritance. In some cases, one mutated copy of the gene can be necessary for a person to be affected by an autosomal dominant disorder. Examples of autosomal dominant type of disorder can include but are not be limited to Huntington's disease, Neurofibromatosis 1, Marfan Syndrome, Hereditary nonpolyposis colorectal cancer, or Hereditary multiple exostoses. In autosomal recessive disorders, two copies of the gene can be mutated for a subject to be affected by an autosomal recessive disorder. Examples of this type of disorder can include but may not be limited to cystic fibrosis, sickle-cell disease (also partial sickle cell disease), Tay-Sachs disease, Niemann-Pick disease, or spinal muscular atrophy. X-linked dominant disorders are caused by mutations in genes on the X chromosome such as X-linked hypophosphatemic rickets. Some X-linked dominant conditions such as Rett syndrome, Incontinentia Pigmenti type 2 and Aicardi Syndrome can be fatal. X-linked recessive disorders are also caused by mutations in genes on the X chromosome. Examples of this type of disorder can include but are not limited to Hemophilia A, Duchenne muscular dystrophy, red-green colorblindness, muscular dystrophy and Androgenetic alopecia. Y-linked disorders are caused by mutations on the Y chromosome. Examples can include but are not limited to
Male Infertility and hypertrichosis pinnae. The genetic disorder of mitochondrial inheritance, also known as maternal inheritance, can apply to genes in mitochondrial DNA such as in
Leber's Hereditary Optic Neuropathy.
[0092] Genetic disorders may also be complex, multifactorial or polygenic. Polygenic genetic disorders can be associated with the effects of multiple genes in combination with lifestyle and environmental factors. Although complex genetic disorders can cluster in families, they do not have a clear-cut pattern of inheritance. Multifactorial or polygenic disorders can include heart disease, diabetes, asthma, autism, autoimmune diseases such as multiple sclerosis, cancers, ciliopathies, cleft palate, hypertension, inflammatory bowel disease, mental retardation or obesity.
[0093] Other genetic disorders can include but may not be limited to lp36 deletion syndrome, 21 -hydroxylase deficiency, 22qll.2 deletion syndrome, aceruloplasminemia, achondrogenesis, type II, achondroplasia, acute intermittent porphyria, adenylosuccinate lyase deficiency, Adrenoleu-kodystrophy, Alexander disease, alkaptonuria, alpha- 1 antitrypsin deficiency, Alstrom syndrome, Alzheimer's disease (type 1, 2, 3, and 4), Amelogenesis Imperfecta, amyotrophic lateral sclerosis, Amyotrophic lateral sclerosis type 2, Amyotrophic lateral sclerosis type 4, amyotrophic lateral sclerosis type 4, androgen insensitivity syndrome, Anemia, Angelman syndrome, Apert syndrome, ataxia-telangiectasia, Beare-Stevenson cutis gyrata syndrome, Benjamin syndrome, beta thalassemia, biotimidase deficiency, Birt-Hogg-Dube syndrome, bladder cancer, Bloom syndrome, Bone diseases, breast cancer, Camptomelic dysplasia, Canavan disease, Cancer, Celiac Disease, Chronic Granulomatous Disorder (CGD), Charcot-Marie-Tooth disease, Charcot-Marie-Tooth disease Type 1, Charcot-Marie-Tooth disease Type 4, Charcot-Marie-Tooth disease Type 2, Charcot- Marie-Tooth disease Type 4, Cockayne syndrome, Coffm-Lowry syndrome, collagenopathy types II and XI, Colorectal Cancer, Congenital absence of the vas deferens, congenital bilateral absence of vas deferens, congenital diabetes, congenital erythropoietic porphyria,
Congenital heart disease, congenital hypothyroidism, Connective tissue disease, Cowden syndrome, Cri du chat syndrome, Crohn's disease, fibrostenosing, Crouzon syndrome,
Crouzonodermoskeletal syndrome, cystic fibrosis, De Grouchy Syndrome, Degenerative nerve diseases, Dent's disease, developmental disabilities, Di-George syndrome, Distal spinal muscular atrophy type V, Down syndrome, Dwarfism, Ehlers-Danlos syndrome, Ehlers-
Danlos syndrome arthrochalasia type, Ehlers-Danlos syndrome classical type, Ehlers-Danlos syndrome dermatosparaxis type, Ehlers-Danlos syndrome kyphoscoliosis type, vascular type, erythropoietic protoporphyria, Fabry's disease, Facial injuries and disorders, factor V Leiden thrombophilia, familial adenomatous polyposis, familial dysautonomia, fanconi anemia, FG syndrome, fragile X syndrome, Friedreich ataxia, Friedreich's ataxia, G6PD deficiency, galactosemia, Gaucher's disease (type 1, 2, and 3), Genetic brain disorders, Glycine encephalopathy, Haemochromatosis type 2, Haemochromatosis type 4, Harlequin Ichthyosis,
Head and brain malformations, Hearing disorders and deafness, Hearing problems in children, hemochromatosis (neonatal, type 2 and type 3), hemophilia, hepatoerythropoietic porphyria, hereditary coproporphyria, Hereditary Multiple Exostoses, hereditary neuropathy with liability to pressure palsies, hereditary non-polyposis colorectal cancer, homocystinuria,
Huntington's disease, Hutchinson Gilford Progeria Syndrome, hyperoxaluria, primary, hyperphenylalaninemia, hypochondrogenesis, hypochondroplasia, idicl 5, incontinentia pigmenti, Infantile Gaucher disease, infantile-onset ascending hereditary spastic paralysis,
Infertility, Jackson-Weiss syndrome, Joubert syndrome, Juvenile Primary Lateral Sclerosis,
Kennedy disease, Klinefelter syndrome, Kniest dysplasia, Krabbe disease, Learning disability, Lesch-Nyhan syndrome, Leukodystrophies, Li-Fraumeni syndrome, lipoprotein lipase deficiency, familial, Male genital disorders, Marfan syndrome, McCune- Albright syndrome, McLeod syndrome, Mediterranean fever, familial, Menkes disease, Menkes syndrome, Metabolic disorders, methemoglobinemia beta-globin type, Methemoglobinemia congenital methaemoglobinaemia, methylmalonic acidemia, Micro syndrome, Microcephaly,
Movement disorders, Mowat-Wilson syndrome, Mucopolysaccharidosis (MPS I), Muenke syndrome, Muscular dystrophy, Muscular dystrophy, Duchenne and Becker type, muscular dystrophy, Duchenne and Becker types, myotonic dystrophy, Myotonic dystrophy type 1 and type 2, Neonatal hemochromatosis, neurofibromatosis, neurofibromatosis 1, neurofibromatosis 2, Neurofibromatosis type I, neurofibromatosis type II, Neurologic diseases, Neuromuscular disorders, Niemann-Pick disease, Nonketotic hyperglycinemia, nonsyndromic deafness, Nonsyndromic deafness autosomal recessive, Noonan syn-drome, osteogenesis imperfecta (type I and type III), otospondylomegaepiphyseal dysplasia, pantothenate kinase-associated neurodegeneration, Patau Syndrome (Trisomy 13), Pendred syndrome, Peutz-Jeghers syndrome, Pfeiffer syndrome, phenylketonuria, porphyria, porphyria cutanea tarda, Prader-Willi syndrome, primary pulmonary hypertension, prion disease, Progeria, propionic acidemia, protein C deficiency, protein S deficiency, pseudo-
Gaucher disease, pseudoxanthoma elasticum, Retinal disorders, retinoblastoma, retinoblastoma FA — Friedreich ataxia, Rett syndrome, Rubinstein-Taybi syndrome, Sandhoff disease, sensory and autonomic neuropathy type III, sickle cell anemia, skeletal muscle regeneration, Skin pigmentation disorders, Smith Lemli Opitz Syn-drome, Speech and communication disorders, spinal muscular atrophy, spinal-bulbar muscular atrophy, spinocerebellar ataxia, spondyloepimetaphyseal dysplasia, Strudwick type, spondyloepiphyseal dysplasia congenita, Stickler syndrome, Stickler syndrome COL2A1,
Tay-Sachs disease, tetrahydrobiopterin deficiency, thanatophoric dysplasia, thiamine- responsive megaloblastic anemia with diabetes mellitus and sensorineural deafness, Thyroid disease, Tourette's Syndrome, Treacher Collins syndrome, triple X syndrome, tuberous sclerosis, Turner syndrome, Usher syndrome, variegate porphyria, von Hippel-Lindau disease, Waardenburg syndrome, Weissenbacher-Zweymiiller syndrome, Wilson disease, Wolf-Hirschhom syndrome, Xeroderma Pigmentosum, X-linked severe combined immunodeficiency, X-linked sideroblastic anemia, or X-linked spinal-bulbar muscle atrophy.
[0094] Compositions and methods disclosed herein for treating or preventing a disease or a condition can comprise administering to a subject a therapeutically effective amount of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some cases, a portion of a plant, an extract thereof, an herb, a portion of an herb, an extract thereof, or any combination thereof can be administered with a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some cases, a plant or an herb can comprise ginseng, ginkgo, mushrooms, wolfberry, dang gui, astragalus, atractylodes, bupleurum, cinnamon, coptis chinensis, ginger, licorice, ephedra, peon, rehmania, rubarb, salvia, cannabis, mint, velvet leaf, Chinese goldthread, poppy, prgining croton, lilac daphne, devil’s trumpet, jimson weed, noble dendrobium, blue evergreen, kudzu, snakeroot, foxglove, stephania root, skullcap, pagoda tree, Indian stringbush or any combination thereof.
[0095] In some cases, a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can be increased or decreased in a subject by altering a composition of a food, a beverage, a nutrient, or any combination thereof. In some cases, the increase or decrease can achieved by altering the diet of a subject, administering a supplement, or by adding exercise routine.
[0096] A method of detecting a compound
[0097] In some embodiments, a method can comprise detecting a presence and a level of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof, from a sample of a subject may have or may be suspected of having a disease or condition and comparing the level of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof, to a reference level; or detecting the presence, the levels, the elution times, of a plurality of compounds of
Table 1, homologs thereof, derivatives thereof, esters thereof, enantiomers thereof, diastereomers thereof, racemates thereof, salts thereof, or any combination thereof and comparing a pattern thereof to a reference pattern. In some cases, the presence and the level can be a concentration. In some cases, the elution time can be in seconds, in minutes, in hours or a combination thereof. In some cases, the reference pattern can be compiled from a single patient result or a plurality of patient results. In some cases, the reference pattern can be obtained from a lab sample, a clinical sample, or a reference sample. In some cases, the reference pattern can be one compound or a plurality of compounds. In some cases, a level can indicate a viral infection, a bacterial infection, a fungal infection, a parasitic infection, a cancer, a tumor, an infectious disease, a skeletal disease, a urogenital disease, a digestive disease, a muscular disease, a neurological disease, an autoimmune disease, a respiratory disease, a cardiac disease, an endocrine disease, a liver disease, a hereditary disease or any combination thereof.
[0098] In some embodiments, the method can be a method of diagnosing a disease or a condition and further can comprise selecting a therapeutic regiment for treating the disease or the condition. In some cases, the therapeutic regiment can comprise selecting a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof to treat the disease or condition in a subject.
[0099] In some cases, the method can comprise detecting a presence and a level of a first compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, a biomolecule of Table 2 or any combination thereof; a presence and a level of a second compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, a biomolecule of Table 2 or any combination thereof; a presence and a level of a third compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, a biomolecule of Table 2 or any combination thereof; and comparing the first level to a first reference level, the second level to a second reference level, and the third level to a third reference level. In some cases, the method can comprise detecting the presence and level of more than the three compounds. In some cases, the method can comprise detecting the presence and the level of more than about: 1, 2, 3, 4, 5, 6,
7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40
45, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 compounds, biomolecules or any combination thereof. In some cases, the method can comprise detecting the presence and level of less than about: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17,
18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 200, 300
400, 500, 600, 700, 800, 900, or 1000 compounds, biomolecules or any combination thereof. [00100] In some cases, a method can be conducted at least in part on a diagnostic device, a screening device, or a diagnostic and a screening device comprising at least one of; a chromatography column, an eluant, a detector, and a mass spectrometer. In some cases, a chromatography column can comprise a gas chromatography column, a liquid chromatography column or any combination thereof and the eluant can comprise, a gas, a liquid or any combination thereof. In some cases, a gas can be helium, hydrogen, or nitrogen. In some instances, the chromatograph column can comprise a paper, a silica gel, a cellulose, an aluminum oxide, or any combination thereof. In some cases, a diagnostic device, a screening device, or a diagnostic and a screening device can comprise a microelectromechanical system (MEMS), a nanoelectromechanical systems (NEMS), or any combination thereof. In some cases, a MEMS and NEMS device can comprise a central unit to process data, a microsensor, and an energy source. In some cases, a diagnostic device, a screening device, or a diagnostic and a screening device can comprise a breath collection tube, a sample collection port, or any combination thereof. In some cases, the breath collection tube can be more than about: 0.1 meters, 0.2 meters, 0.3 meters, 0.4 meters, 0.5 meters, 0.6 meters, 0.7 meters, 0.8 meters, 0.9 meters, 1.0 meters, 1.5 meters, 2 meters. 2.5 meters. 3 meters. 3.5 meters or 4 meters. In some cases, the breath collection tube can be less than about: 0.1 meters, 0.2 meters, 0.3 meters, 0.4 meters, 0.5 meters, 0.6 meters, 0.7 meters,
0.8 meters, 0.9 meters, 1.0 meters, 1.5 meters, 2 meters. 2.5 meters. 3 meters. 3.5 meters or 4 meters.
[00101] In some embodiments, a diagnostic device, a screening device, or a diagnostic and a screening device may have: a positive volume of less than about 16 cubic feet, a method of transport, wherein the device can be moved by a robot, a drone, or a remote-control transport machine; a self-contained power supply, or any combination thereof. In some cases a diagnostic device, a screening device, or a diagnostic and a screening device may have a volume of less than about: 25 cubic feet, 24 cubic feet, 23 cubic feet, 22 cubic feet, 21 cubic feet, 20 cubic feet, 19 cubic feet, 18 cubic feet, 17 cubic feet, 16 cubic feet, 15 cubic feet, 14 cubic feet, 13 cubic feet, 12 cubic feet, 11 cubic feet, 10 cubic feet, 9 cubic feet, 8 cubic feet, 7 cubic feet, 6 cubic feet, 5 cubic feet, 4 cubic feet, 3 cubic feet, 2 cubic feet, 1 cubic foot, or 0.5 cubic foot. In some cases, a robot, a drone, or a remote-control transport machine can be controlled autonomously. In some embodiments, a diagnostic device, a screening device, or a diagnostic and screening device can link wirelessly or directly to a processing station. In some cases, a wireless connection can be a Wi-Fi connection, a broadband cellular connection, a Bluetooth® connection, or any combination thereof. A processing station can analyze the data remotely. In some cases, the device can have a length of about: 65 cm, 64 cm, 63 cm, 62 cm, 61 cm, 60 cm, 59 cm, 58 cm, 57 cm, 56 cm, 55 cm, 54 cm, 53 cm, 52 cm,
51 cm, 50 cm, 49 cm, 48 cm, 47 cm, 46 cm, 45 cm, 44 cm, 43 cm, 42 cm, 41 cm, or 40 cm. In some cases, the device can have a width of about: 45 cm, 44 cm, 43 cm, 42 cm, 41 cm, 40 cm, 39 cm, 38 cm, 37 cm, 36 cm, 35 cm, 34 cm, 33 cm, 32 cm, 31 cm, 30 cm, 29 cm, 28 cm, 27 cm, 26 cm, 25 cm, 24 cm, 23 cm, 22 cm, 21 cm, or 20 cm. In some cases, the device can have a height of about: 40 cm, 39 cm, 38 cm, 37 cm, 36 cm, 35 cm, 34 cm, 33 cm, 32 cm, 31 cm, 30 cm, 29 cm, 28 cm, 27 cm, 26 cm, 25 cm, 24 cm, 23 cm, 22 cm, 21 cm, or 20 cm, 19 cm, 18 cm, 17 cm, 16 cm, or 15 cm. In some cases, a device can have a length of less than about: 16 cm, 15, cm, 14 cm, 13 cm, 12 cm, 11 cm, 10 cm, 9 cm, 8 cm, 7 cm, or 6 cm. In some cases, a device can have a width of less than about: 11 cm, 10 cm, 9 cm, 8 cm, 7 cm, 6 cm, 5 cm, or 4 cm. In some cases, a device can have a height of less than about: 6 cm, 5 cm, 4 cm, 3 cm, 2 cm, 1 cm or 0.5 cm.
[00102] In some embodiments, the method can be used to identify a disease or condition, wherein the sample can be taken from a subject and applied to a device. In some cases, a computer can be used to at least partially compare a chromatogram in a subject having a disease to a chromatogram of a subject not having the disease, and determining independently the difference in the presence, the absence, the level, or any combination thereof of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, a biomolecule from Table 2 or any combination thereof. In some embodiments, the method can be used to identify a disease or condition, wherein the sample can be taken from a subject and applied to a device. In some cases, a computer can be used to at least partially compare data (e.g., measured or recorded data that may or may not be normalized to a control, e.g., in the form of a chromatogram, a heat map, or a scalogram) in a subject having a disease to data (e.g., measured or recorded data that may or may not be normalized to a control, e.g., in the form of a chromatogram, a heat map, or scalogram) from a subject not having the disease, and determining independently the difference in the presence, the absence, the level, or any combination thereof of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, a biomolecule from Table 2 or any combination thereof. For instance, a heat map may be generated during analysis of data produced from embodiments described herein, e.g., which can comprise the use of MEMS. In some cases, a heat map can be used for analysis of data measured or recorded from embodiments described herein, e.g., which can comprise the use of MEMS, for example, to determine the presence of a condition such as a cancer in a subject, the risk of a condition such as cancer in a subject, or a prognosis of a subject.. In some cases, a chromatogram or scalogram can be generated during analysis of data produced from embodiments described herein, e.g., which can comprise the use of GCMS or MEMS.
In some cases, a chromatogram or scalogram can be used for analysis of data measured or recorded from embodiments described herein, e.g., which can comprise the use of GCMS or
MEMS, for example, to determine the presence of a condition such as a cancer in a subject, the risk of a condition such as cancer in a subject, or a prognosis of a subject. In some cases, the chromatogram can comprise a signature of a disease or a condition. In some cases, the chromatogram can comprise a signature of more than one disease or condition. In some cases, the signature can be compiled from a single patient result or a plurality of patient results. In some cases, the signature can be obtained from a lab sample, a clinical sample, or a reference sample. In some cases, the signature can be a pattern. In some cases, the pattern can be one compound or a plurality of compounds. In some cases, the pattern can be one biomolecule or a plurality of biomolecules. In some cases, the pattern can comprise both biomolecules and compounds.
[00103] In some cases, the method may include using an algorithm. The algorithm may be employed to perform one or more comparisons of the method, such as comparing one or more chromatograms. The algorithm may be a supervised learning algorithm, a trained algorithm, a machine learning algorithm or other. In some cases, the algorithm may comprise unsupervised learning algorithm. In some cases, the algorithm may comprise a hierarchical clustering. Examples of algorithms may include support vector machine (SVM) algorithm, a random forest algorithm, a logistical regression algorithm, a neural network, a decision tree algorithm, a naive bayes algorithm, nearest neighbor algorithm, or any combination thereof.
In some cases, the method may not comprise use of an algorithm. In some cases, a portion of the method may employ use of an algorithm. In some cases, an algorithm can use a processor or a microprocessor.
[00104] In some cases, a computer can comprise a memory, one or more executable instructions, one or more processors and a graphical user interface. In some cases, a result can be communicated via a communication medium. In some instances, a communication medium can be a screen, an audio description, a physical readout, or any combination thereof. In some cases, a communication medium can be the internet, a private server, a cell phone, a computer, a television, a hologram, a smart device, a speaker, a printer, or any combination thereof.
In some cases, the compound of Table 1 identified by the method, or the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof can be selected as a therapeutic In some cases a biomolecule of Table 2 can be identified by the method and can be selected as a therapeutic. In some cases a compound from Table 1 and a biomolecule from Table 2 can be identified by the method. In some cases, the method can include diagnosing a presence or an absence of a disease or condition in a subject. In some cases, the method can include a prognosis of a disease or condition in a subject, such as a prognosis prior to, during, or following administration of one or more pharmaceutical compositions. In some cases, the method can include a theranostic method, such as administration of a first component to a subject to diagnose a disease or condition and administration of a second component to the subject to at least partially treat or reduce one or more symptom of the disease or condition.
In some cases, the method can include measuring a health level information, such as a heart rate, a blood pressure, a body temperature, an oxygen saturation, a protein, a chemokine or a hormone level, a medical history, or any combination thereof. The subject may be symptomatic for the disease or condition. The subject may be asymptomatic for the disease or condition. The subject may have received a previous diagnosis for the disease or condition.
The subject may be suspected of having the disease or condition.
[00105] In some instances, the subject can display an altered profile, a level of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, the biomolecule of Table 2 or any combination thereof may be at least partially increased or at least partially decreased. The altered profile can be visually represented as chromatogram. In some cases, a measurement and/or an analysis (e.g., comprising a measurement and/or analysis of a healthy subject’s metabolic profile or a metabolic profile of a subject having a condition such as cancer, wherein either profile can include VOC) can be made one or more times over a period of 1 minute, 2 minutes, 3 minutes, 5 minutes, 10 minutes, 15 minutes, 20 minutes, 30 minutes, 1 hour, 2 hours, 6 hours, 12 hours, 24 hours, 2 days, 5 days, 1 week, 2 weeks, 1 month, 2 months, 6 months, 1 year, 2 years, 3 years, 5 years, 10 years, 20 years, less than 1 minute, from 1 minute to 2 minutes, from 2 minutes to 3 minutes, from 3 minutes to 5 minutes, from 5 minutes to 10 minutes, from 10 minutes to 15 minutes, from 15 minutes to
20 minutes, from 20 minutes to 30 minutes, from 30 minutes to 1 hour, from 1 hour to 2 hours, from 2 hours to 6 hours, from 6 hours to 12 hours, from 12 hours to 24 hours, from 1 day to 2 days, from 2 days to 5 days, from 2 days to 1 week, from 1 week to 2 weeks, from 2 weeks to 1 month, from 1 month to 2 months, from 2 months to 6 months, from 6 months to 1 year, from 1 year to 2 years, from 2 years to 3 years, from 3 years to 5 years, from 5 years to
10 years, from 10 years to 20 years, at least 1 minute, at least 2 minutes, at least 3 minutes, at least 5 minutes, at least 10 minutes, at least 15 minutes, at least 20 minutes, at least 30 minutes, at least 1 hour, at least 2 hours, at least 6 hours, at least 12 hours, at least 24 hours, at least 2 days, at least 5 days, at least 1 week, at least 2 weeks, at least 1 month, at least 2 months, at least 6 months, at least 1 year, at least 2 years, at least 3 years, at least 5 years, at least 10 years, at least 20 years, at most 1 minute, at most 2 minutes, at most 3 minutes, at most 5 minutes, at most 10 minutes, at most 15 minutes, at most 20 minutes, at most 30 minutes, at most 1 hour, at most 2 hours, at most 6 hours, at most 12 hours, at most 24 hours, at most 2 days, at most 5 days, at most 1 week, at most 2 weeks, at most 1 month, at most 2 months, at most 6 months, at most 1 year, at most 2 years, at most 3 years, at most 5 years, at most 10 years, at most 20 years. In some cases, a healthy subject’s profile can be recorded for at least about: 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months 9 months, 10 months, 11 months, 1 year, 2 years, 3 years, 4 years, 5 years, 6 years, 7 years, 8 ears, 9 years, 10 years, 11 years, 12 years, 13 years, 14 years, 15 years, 20 years, 25 years, 30 years, 35 years, 40 years, 50 years, 60 years, 70 years, 80 years, 90 years or 100 years. In some cases, a healthy subject’s metabolic profile can be recorded from about: 1 day to about 100 days, 1 day to about 50 days, 1 day to about 25 days, 1 day to about 15 days, 1 day to about 13 days, 1 day to about 12 days, 1 day to about 11 days, 1 day to about 10 days,
1 day to about 9 days, 1 day to about 8 days, 1 day to about 7 days, 1 day to about 6 days, 1 day to about 5 days, 1 day to about 4 days, 1 day to about 3 days, or 1 day to about 2 days. In some cases, a subject’s profile can be recorded once over a time period. In some cases, a subjects profile can be recorded at least about: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15,
16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40,
41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65,
66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90 or more times. In some embodiments, a metabolic profile of the subject can be unknown. In some cases, the healthy metabolic profile can be determined from factors comprising a subject’s genetic profile, a diseased metabolic profile, a healthy population profile, a diseased population profile, a reference profile, or any combination thereof.
[00106] In some cases, a method can comprise performing a diagnostic test and administering a therapeutic comprising the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof. In some cases, the diagnostic test can comprise subjecting a sample or at least a portion of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof to a device comprising: a chromatography column, an eluant, a detector, and a mass spectrometer.
[00107] In some cases, a method can be promoting or maintain health in a subject. Promoting or maintaining health in a subject can comprise promoting or maintaining: ear, nose and throat health, eye health, cardiovascular health, respiratory health, skeletal health, skin health, urogenital health, digestive health, muscular health, neural health, immune system health, liver health, mental health, sexual health, circulatory system health, joint health, dermatological health, lung health, dental health, bone health, central nervous system health, peripheral nervous system health, spine health, kidney health, heart health, intestine health, podiatry health, endocrine health or any combination thereof. In some cases, health can include pediatric health, infant health, adult health or any combination thereof.
[00108] Altering compound levels in a subject
[00109] In some embodiments, a starting material can be administered to a subject and the starting material can be transformed into the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof. In some cases, the starting material can be a compound of a biological pathway, an intermediate in a biological pathway, a prodrug, or any combination thereof. In some cases, the transformation can be a phase 1 reaction, a phase 2 reaction, a microbial biotransformation, a digestive biproduct, a metabolic biproduct, a degradation biproduct or any combination thereof.
[00110] In some cases, a compound can be administered to a subject to alter the production of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, a enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof. In some cases, the production of a biomolecule in Table 2 can be altered. In some cases, the compound can be an inhibitor, an activator, a metabolite, a toxin, an intermediate compound of a biological pathway, or any combination thereof. In some cases, the compound can be an at least partial inhibitor or a complete inhibitor to a polypeptide or a biologically active fragment thereof. In some cases, the compound can have a direct or an indirect effect on a biological pathway. In some cases, the biological pathway can be a competing biological pathway. In some cases, the production of the compound from Table 1, a homolog thereof, a derivative thereof, an ester thereof, a enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof can increase or decrease. In some cases, the compound from Table 1 can be structurally modified.
[00111] In some embodiments, the compound of Table 1, homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof can be covalently linked to an antibody or fragment thereof. In some cases, the linkage can be a cleavable linkage. A non-cleavable linkage can comprise a thioether linker, a maleimide alkane linker, a maleimide cyclohexane linker, or any combination thereof. The cleavable linkage can comprise an acid-labile linker, a reducible linker, a disulfide-linker, a hydrazone linker, a peptide linker, or any combination thereof. [00112] In some cases, the method of treating or preventing a disease can include at least partially inducing, promoting, stimulating, or any combination thereof endocytosis from a cell in the subject. In some cases, the endocytosis can be of a polypeptide, a virus, a lipid, a polynucleotide, a molecule, a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or the salt thereof. In some cases, the endocytosis can be measured by intravital microscopy, fluorescence microscopy, and labeling endocytosed substrates. In some cases, the endocytosis can be measured by comparing endocytosis in untreated cells with cells that have been treated. In some cases, the cell can be an isolated cell from the subject. In some cases, the isolated cell can be manipulated in vitro. In some cases, the method can include at least partially inhibiting a cell growth, a cell communication, a cell metastasis or any combination thereof in the subject. In some cases, the cell growth, the cell communication, or the cell metastasis can be of a cancer cell, a tumor cell, a mutated cell, or any combination thereof. In some cases, the cell growth, the cell communication, or the cell metastasis can be inhibited by a direct or indirect contact with the pharmaceutical composition. In some cases, the cell growth, the cell communication, or the cell metastasis can be measured by a biochemical assay, intravital microscopy, fluorescence microscopy, tumor shrinkage, cancer cell depletion, cancer remission, cell death, or any combination thereof. In some cases, the cell growth, the cell communication or the cell metastasis can be measured by comparing cell growth, the cell communication or the cell metastasis in an untreated cell with the cell growth, the cell communication or the cell metastasis in a treated cell. In some cases, the cell can be an isolated cell from the subject. In some cases, the isolated cell can be manipulated in vitro and shown to be have inhibited cell growth, cellular communication, or cellular metastasis. In some cases, the method can include at least partially inducing, promoting, stimulating, or any combination thereof apoptosis or cell death in the subject. In some cases, the apoptosis or cell death can be of a cancer cell, a tumor cell, a mutated cell, or any combination thereof. In some cases, apoptosis or cell death can be stimulated by a direct or indirect contact with the pharmaceutical composition. In some cases, the cell death or apoptosis can be measured by a biochemical assay, intravital microscopy, fluorescence microscopy, tumor shrinkage, cancer cell depletion, cancer remission or any combination thereof. In some cases, the cell can be an isolated cell from the subject. In some cases, the isolated cell can be manipulated in vitro and shown to be have increased cell death or apoptosis. In some cases, the method can include at least partially inducing, promoting, stimulating or any combination thereof Reactive Oxygen Species (ROS) within the subject.
The ROS can be produced by a direct or indirect contact with the pharmaceutical composition. In some cases, the ROS production can occur in a cancer cell, a tumor cell, a mutated cell, or any combination thereof. In some cases, the ROS production can be measured by an electron paramagnetic resonance assay, a cytochrome c reduction assay, nitroblue tetrazolium assay, fluorescence analysis of dihydroethidium or hydroethidium, fluorescence analysis of hydrocyans, chemiluminescence assays, electrochemical detection, a probe, in vivo redox imaging using photon-electron double-resonance imagine, fluorescence microscopy, or any combination thereof. In some cases, the cell can be an isolated cell from the subject. In some cases, the ROS can be measured by comparing ROS production in an untreated cell with ROS production in a treated cell In some cases, the isolated cell can be manipulated in vitro and shown to have increased ROS production.
[00113] In some cases, the method can comprise at least partially activating or deactivating an immune cell. In some instances, the immune cell can be activated or deactivated by a direct or indirect contact with a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or the salt thereof. In some cases the immune cell can be a dendritic cell, a neutrophil, a granulocyte, a monocyte, a lymphocyte, a B-cell, a T-cell, a natural killer (NIC) cell, a neutrophil, a mast cell, a basophil, an eosinophil, a memory B-cell, a plasma cell, a memory T-cell, a CD4 T-cell, a CD8 T-cell, a Thl T-cell, a Thl7 T-cell, a Treg T-cell, a Th2 T-cell, a precursor cell type to any of these or any combination thereof. In some cases, an immune cell can be induced into maturation, senescence, differentiation or any combination thereof. For example, a myeloblast can be induced into a granulocyte or a CD4 T-cell can be induced into a TH17 T-cell. In some cases, a hyperactive immune cell can be induced into senescence. In some cases, an activated immune cell can indirectly suppress a tumor or a cancer in a subject (e.g. a CD4 T-Cell a CD8 T-cell, an NK cell or a combination thereof). In some cases, an immune cell can be deactivated and suppress a tumor, for example, in a lymphoma or a leukemia. In some cases, an immune cell can be activated and repress an infection, for example, activation of a CD8 T-Cell to suppress a viral infection or activation of a neutrophil to suppress a bacterial or fungal infection. In some cases, the immune cell can be activated to remove excess protein or other toxic molecules, for example, a NK cell can be activated to remove alpha-synuclein aggregates in a subject.
[00114] In some cases, the method can comprise at least partially disrupting a communication between one or more cells in the subject. In some cases, cell communication can be inhibited, enhanced, altered, or any combination thereof. In some cases, cell communication can be altered in one cell type, or multiple cell types. In some cases, disrupting cell communication can lead to cell death or apoptosis. In some cases, disrupting cell communication can increase or decrease an immune response. In some cases, disrupting cell communication can increase or decrease neural cell communication.
[00115] In some embodiments, the method of treating or preventing a disease can comprise directly or indirectly at least partially increasing in the subject a level, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
[00116] In some cases, the treating or preventing disease can comprise administering a genetically engineered cell to capture, to produce, or to chemically alter the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof. In some cases, the genetically engineered cell can be a prokaryote. In some cases, the prokaryote can be a bacterium (e.g. E. coli or a Lactobacillus species) or an Archaea. In some cases, the genetically engineered cell can be a eukaryotic cell. In some cases, the genetically engineered cell can be autologous or allogenic. In some cases, the genetically engineered cell can comprise a polynucleotide. In some cases, the polynucleotide encodes for a polypeptide or a biologically active fragment that can produce, alter, capture, or degrade the compound of
Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof.
[00117] In some embodiments, the method of treating or preventing a disease can comprise directly or indirectly at least partially reducing in the subject a level, an activity, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, a biomolecule of Table 2 or any combination thereof. In some cases, the treating or preventing disease can comprise editing a polynucleotide. In some cases, editing can be performed by a CRISPR Cas system or a similar nuclease. In some cases, the polynucleotide can encode for a polypeptide in a biochemical pathway for making the compound, or for degrading the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof. In some cases, the polynucleotide can comprise a ribose or a deoxyribose. In some cases, the edited polynucleotide can be comprised in an isolated cell and the isolated cell can be administered to the subject. In some cases, the isolated cell can be autologous or allogenic.
[00118] In some embodiments, the treating or preventing disease can comprise administering an at least partially interfering polynucleotide to bind to RNA or DNA to at least partially inhibit polypeptide production in a biochemical pathway for making the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the salt thereof, a biomolecule of Table 2 or any combination thereof. In some cases, the interfering polynucleotide can be a miRNA molecule or a siRNA molecule. In some embodiments, the treating or preventing disease can comprise administering a CRISPR system to edit a polynucleotide to at least partially inhibit polypeptide production in a biochemical pathway for making the compound. In some cases, the CRISPR system, can be a class 1 system, a class 2 system. In some cases the CRISPR system can comprise, a Cas3, a Cas8a, a Cas5, a
Cas8b, a Cas8c, a caslOd, a Csel, a Cse2, a Csyl, a Csy2, a Csy 3, a GSU0054, a CaslO, a
Csm2, a Cmr5, a CaslO, a Csxl 1, a CsxlO, a Csfl, a Cas9, a Csn2, a Cas4, a cpfl, a C2cl, a
Casl3a, a Casl3b, a Casl3c, a Casl3d or any combination thereof. In some cases, an edited polynucleotide can have a nonsense mutation, a missense mutation, a deletion or any combination thereof. In some cases, a polynucleotide can be engineered. In some cases, an engineered polynucleotide can comprise a plurality of nucleotides. In some cases, an engineered oligonucleotide can comprise an artificial nucleic acid analogue. In some cases, an engineered oligonucleotide can comprise DNA, can comprise cell-free DNA, cDNA, fetal
DNA, or maternal DNA. In some cases, an oligonucleotide can comprise miRNA, shRNA, or siRNA.
[00119] In some cases, the treating or preventing disease can comprise administering an antibody, an aptamer, or a fragment thereof to at least partially bind to the compound of Table 1 the homolog thereof, the derivative thereof, the ester thereof, or the salt thereof. In some cases, the antibody can be monoclonal or polyclonal. In some cases, the antibody or fragment thereof can be from an IgG, IgA, IgM. In some cases, the binding can decrease bioavailability, decrease half-life, inhibit the action of the compound, or any combination thereof.
[00120] In some cases, the treating or preventing disease can comprise administering a molecule, a salt thereof, or a formulation containing at least one of these to capture the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, or the salt thereof. In some cases, the molecule can contain functional groups that bind to the compound. In some cases, the binding can be a covalent bond an ionic bond or a combination thereof.
[00121] In some embodiments, the treating or preventing disease can comprise administering dialysis to a patient, wherein the dialysis filters at least some of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, or the salt thereof. In some cases, the dialysis can comprise hemodialysis, peritoneal dialysis, or any combination thereof. In some cases, the dialysis can be hemodialysis, peritoneal dialysis, or any combination thereof. In some cases, the compound can be filtered from the blood of a subject.
[00122] Second therapy
[00123] As used herein, a second therapy can include chemotherapy, radiation, bone marrow transplantation, a checkpoint inhibitor, CAR T-cell therapy, immunotherapy, hormone therapy, cryotherapy, surgical procedure (such as tumor resection) or any combination thereof. A second therapy can include administration of a pharmaceutical composition, such as a small molecule. A second therapy can include administration of a pharmaceutical composition, such as one or more antiviral drugs, for example, interferon, oseltamivir, ribavirin, daclatasvir, sofosbuvir, velpatasvir, voxilapresvir, remdesivir, indomethacin, or any combination thereof. A second therapy can include administration of a pharmaceutical composition, such as one or more antibiotics. A second therapy can comprise administration of a muscle relaxant, an anti-depressant, a steroid, an opioid, a cannabis-based therapeutic, acetaminophen, a non-steroidal anti-inflammatory, a neuropathic agent, a cannabis, a progestin, a progesterone, or any combination thereof. A neuropathic agent may comprise gabapentin. A non-steroidal anti-inflammatory can comprise naproxen, ibuprofen, a COX-2 inhibitor, or any combination thereof. A second therapy can comprise administration of a heart medication such as a high blood pressure medication, a beta blocker, an anticoagulant, an antiplatelet agent, an ACE inhibitor, an angiotensin II receptor blocker, an angiotensin receptor-neprilysin inhibitor, a calcium channel blocker, a cholesterol-lowering medication, a diuretic, a vasodilator or any combination thereof. A second therapy can comprise administration of a biologic agent, cellular therapy, regenerative medicine therapy, a tissue engineering approach, a stem cell transplantation or any combination thereof. A second therapy can comprise a medical procedure. A medical procedure can comprise an epidural injection (such as a steroid injection), acupuncture, exercise, physical therapy, an ultrasound, a surgical therapy, a chiropractic manipulation, an osteopathic manipulation, a chemonucleolysis, or any combination thereof. A second therapy can comprise use of a breathing assist device or a ventilator. A second therapy can comprise administration of a regenerative therapy or an immunotherapy such as a protein, a stem cell, a cord blood cell, an umbilical cord tissue, a tissue, or any combination thereof. A second therapy can comprise a biosimilar. In some cases, a second therapy, can alter the levels of compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof in a subject. In some cases, a second therapy, may not alter the levels of compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof in a subject. In some cases, the administering of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or a salt thereof and a second therapy can be concurrent.
In some cases, the administering of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, or a salt thereof and a second therapy can be sequential.
[00124] In some cases, the treating or preventing can comprise administering a polynucleotide to a subject, a genetically engineered virus, a vector, or any combination thereof to a subject to directly or indirectly produce the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, or the salt thereof or any combination thereof in a subject. In some cases, the genetically engineered virus, the vector or any combination thereof can comprise at least one polynucleotide. In some cases, a polynucleotide can encode for a polypeptide or biologically active fragment thereof in a biochemical pathway for making the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, or the salt thereof. In some cases, the polynucleotide can encode for a gene, a biologically active RNA, such as siRNA or miRNA, or any combination thereof. In some cases, the vector can be a plasmid. In some cases, a vector can comprise RNA, DNA or any combination thereof. In some cases, a vector can be used for drug delivery, in an in vitro setting, in vivo setting, or any combination thereof. The vector can comprise a recombinant vector. The vector can be a vector that can be modified from a naturally occurring vector. The vector can comprise at least a portion of a non-naturally occurring vector. Any vector can be utilized. In some cases, a vector can deliver the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof. In some cases, the vector can be targeted to but may not be limited to a subject, or a specific organ, or a specific cell, or any combination thereof. The vector can comprise any composition described herein. In some cases, the vector can be comprised of a liposome, a nanoparticle or any combination thereof.
The liposome can include but may not be limited to unilamellar liposome, multilamellar liposome, archaeosome, noisome, novasome, cryptosome, emulsome, vesosome, or a derivative of any of these, or any combination thereof. The nanoparticle can include but may not be limited to biopolymeric nanoparticle, alginate nanoparticle, xanthan gum nanoparticle, cellulose nanoparticle, dendrimer, polymeric micelle, polyplexed, inorganic nanoparticle, nanocrystal, metallic nanoparticle, quantum dot, protein nanoparticle, polysaccharide nanoparticle, or a derivative of any of these, or any combination thereof. In some cases, the vector can be an RNA viral vector which can include but may not be limited to a retrovirus, lentivirus, coronavirus, alphavirus, flavivirus, rhabdovirus, morbillivirus, picornavirus, coxsackievirus, or picornavirus or portions of any of these, or fragments of any of these, or any combination thereof. In some cases, the vector can be a DNA viral vector which can include but may not be limited to an adeno-associated viral (AAV) vector, adenovirus, hybrid adenoviral system, hepadnavirus, parvovirus, papillomavirus, polyomavirus, herpesvirus, poxvirus, a portion of any of these, or a fragment of any of these, or any combination thereof.
[00125] In some cases, a subject may not have been previously diagnosed with a disease or condition. In some cases, a subject may have been diagnosed with a disease or condition. In some cases, a subject may not have received a definitive diagnosis of a disease or condition.
In some cases, a subject may have previously had a disease or condition. A subject may be in remission. A subject may be at risk of developing a disease or condition (such as based at least in part on a previous condition, a lifestyle factor, a genetic variant, or any combination thereof). A subject may have received a diagnostic test. In some cases, a subject may receive a second diagnostic test in addition to the diagnostic device, the screening device, or the diagnostic and screening device described herein. A diagnostic test can include an imaging procedure, a blood count analysis, a tissue pathology analysis, a biomarker analysis, a rapid diagnostic test, a polymerase chain reaction diagnostic test, an enzyme-linked immunosorbent assay (ELISA), a biopsy, a colonoscopy, an electrocardiogram, an electroencephalogram, a gastroscopy, an eye test, an magnetic resonance imaging (MRI scan), a positron emission tomography (PET) scan, an ultra sound, an X-ray, a computed tomography (CT) scan, a physical examination, an organ function test, a pregnancy test, a toxicology test, a uranalysis, a thyroid function test, a pap smear, a gastric fluid analysis, a fecal sample analysis, a histology test, a genetic test, a mental function test, or any combination thereof. In some cases, the disease or condition may comprise a cancer, a tumor, an infectious disease, a skeletal disease, a urogenital disease, a digestive disease, a muscular disease, a neurological disease, an autoimmune disease, a respiratory disease, a cardiac disease, an endocrine disease, a liver disease, a hereditary disease, or any combination thereof. In some cases, the disease or condition may be a viral infection, such as a SARS-CoV infection, a SARS-CoV-2 infection, a MERS-CoV infection, a HIV infection, or any combination thereof. In some cases, the disease or condition may be cancer, such as a lymphoma, a carcinoma, a sarcoma, a skin cancer, a cervical cancer, a prostate cancer, or any combination thereof.
[00126] Methods can include in vivo or in vitro delivery methods. Methods can include contacting a cell, such as a cell in vivo with the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, or the salt thereof. Methods can include contacting a cell, such as an isolated and purified cell (such as a cell in vitro) with the compound of Table
1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof, or a pharmaceutical composition as described herein. Methods can include contacting a tissue, such as an in vivo tissue or an isolated in vitro tissue, with a compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the salt thereof, the nucleic acid construct, or the pharmaceutical composition as described herein.
[00127] Referring to FIG. 1, this figure shows a gas chromatography mass spectrometry
(GCMS) chromatogram of a patient with breast cancer, in accordance with embodiments.
[00128] Referring to FIG. 2 this figure shows a GCMS chromatogram of a patient with kidney cancer, in accordance with embodiments.
[00129] Referring to FIG. 3A-3D, these figures show GCMS chromatograms from gastric patients’ blood, in accordance with embodiments.
[00130] Referring to FIG. 4A-4D, these figures show this figure shows GCMS chromatograms from lung cancer patients’ blood.
[00131] Referring to FIG. 5A-5D, these figures show this figure shows GCMS chromatograms from gastric cancer patients’ urine, in accordance with embodiments.
[00132] Referring to FIG. 6A-6D, these figures show this figure shows GCMS chromatograms from kidney cancer patients’ urine, in accordance with embodiments. [00133] Referring to FIG. 7A-7D, these figures show this figure shows GCMS chromatograms from lung cancer patients’ urine, in accordance with embodiments.
[00134] Referring to FIG. 8A-8C, these figures show this figure shows GCMS chromatograms from health control’s blood, in accordance with embodiments.
[00135] Referring to FIG. 9A-B this figure shows chromatograms of breast cancer patients, in accordance with embodiments. FIG. 9A shows one breast cancer patient’s metabolite volatile organic compound (VOC) profile. FIG. 9B shows an overlay of six breast cancer patients’ metabolite VOC profile.
[00136] Referring to FIG. 10A-B this figure shows chromatograms of kidney cancer patients. FIG. 10A shows one kidney cancer patients metabolite VOC profile. FIG.10B shows an overlay of six kidney cancer patients’ metabolite VOC profiles.
[00137] Referring to FIG. 11A-B this figure shows chromatograms of lymphoma patients, in accordance with embodiments. FIG. 11A shows one lymphoma VOC profile. FIG. 11B shows an overlay of six lymphoma patients’ metabolite VOC profiles.
[00138] Referring to FIG. 12A-12G, these figures show this figure shows chromatograms of breast and kidney cancer patients’ blood, in accordance with embodiments.
[00139] Referring to FIG. 13 this figure shows chromatograms from lung cancer and liver cancer patients’ blood, in accordance with embodiments.
[00140] Referring to FIG. 14 this figure shows chromatograms from kidney cancer patients’ urine, in accordance with embodiments.
[00141] Referring to FIG. 15 this figure shows saliva chromatograms, in accordance with embodiments.
[00142] Referring to FIG. 16 this figure shows chromatograms from volunteers’ breath, in accordance with embodiments. Portable gas chromatography (GC) chromatograms in the top and middle rows (upper left, upper right, middle left and middle right) are from volunteers. The chromatogram at the lower left is a blank control chromatogram. [00143] Referring to FIG. 17 this figure shows a chromatogram from a cancer patient, in accordance with embodiments. The portable GC chromatogram is from the blood of a male lung cancer patient.
[00144] Referring to FIG. 18 this figure shows a chromatogram from a cancer patient, in accordance with embodiments. The portable GC chromatogram is from the blood of a female endometrial cancer patient.
[00145] Referring to FIG. 19 this figure shows a chromatogram from a lymphoma patient, in accordance with embodiments. The portable GC chromatogram is from the blood of a female lymphoma cancer patient.
[00146] Referring to FIG. 20 this figure shows a chromatogram from a lung cancer patient, in accordance with embodiments. The portable GC chromatogram is from the saliva of a female lung cancer patient.
[00147] Referring to FIG. 21 this figure shows a chromatogram from a prostate cancer patient, in accordance with embodiments. The portable GC chromatogram is from the blood of a male with prostate cancer.
[00148] Referring to FIG. 22 this figure shows a chromatogram of a healthy control and cancer patients, in accordance with embodiments. The portable GC chromatogram is an overlay of a healthy control patient, a breast cancer patient, a lung cancer patient, a kidney cancer patient.
[00149] Referring to FIG. 23 this figure shows a chromatogram of a healthy control and a lab standard, in accordance with embodiments. The portable GC chromatogram of a healthy controls urine and a lab made synthetic standard urine sample to identify VOCs.
[00150] Referring to FIG. 24 this figure shows a gas chromatography (GC) chromatogram analysis of heated blood and urine samples from kidney cancer patients. The portable GC chromatogram of heated blood and urine samples from kidney cancer patients against room air, which can comprise a confounding factor in some embodiments. [00151] Referring to FIG. 27A-B this figure shows a GC device for testing a sample.
FIG.27A shows the connected to a computer that displays the chromatogram. FIG. 27B shows the device connected to a breath sampling mask.
[00152] Referring FIG. 28A-B this figure shows a GC device testing a sample from a subject. FIG.28A shows the device connected to a PC which can transmit or present the data. A robotic drone is pictured moving the device. FIG. 28B shows a device that can transmit the data wirelessly via cellular connections or Wi-Fi. The device is autonomously moved by a robot.
Computer control systems
[00153] In some cases, as shown in FIG. 26, a sample 202 containing a blood sample can be obtained from a subject 201, such as a human subject. A sample 202 can be subjected to one or more methods as described herein, such as adding the sample to a chromatography column, an eluant, a detector, a mass spectrometer, or any combination thereof. In some cases, an assay can comprise obtaining a chromatograph. One or more results from a method can be input into a processor 204. One or more input parameters such as a sample identification, subject identification, sample type, a reference, or other information can be input into a processor 204. One or more metrics from an assay can be input into a processor 204 such that the processor can produce a result, such a diagnosis, a prognosis, a theranostic, of a viral infective disease or a recommendation for a treatment. A processor can send a result, an input parameter, a metric, a reference, or any combination thereof to a display 205, such as a visual display or graphical user interface. A processor 204 can (i) send a, an input parameter, a metric, or any combination thereof result wirelessly or directly to a server 207, (ii) receive a result, an input parameter, a metric, or any combination thereof from a server 207, (iii) or a combination thereof. In some cases, the output can be a chromatograph.
[00154] The present disclosure provides computer control systems that are programmed to implement methods of the disclosure. FIG. 25 shows a computer system 101 that is programmed or otherwise configured to perform gas chromatography on a blood sample. The computer system 101 can regulate various aspects of the present disclosure, such as, diagnosis, prognosis, a theranostic method, and a health level information method The computer system 101 can be an electronic device of a user or a computer system, a handheld system, a cell phone or any combination thereof that is remotely located with respect to the electronic device.
[00155] The computer system 101 includes a central processing unit (CPU, also “processor” and “computer processor” herein) 105, which can be a single core or multi core processor, or a plurality of processors for parallel processing. The computer system 101 also includes memory or memory location 110 (e.g., random-access memory, read-only memory, flash memory), electronic storage unit 115 (e.g., hard disk), communication interface 120 (e.g., network adapter) for communicating with one or more other systems, and peripheral devices 125, such as cache, other memory, data storage and/or electronic display adapters. The memory 110, storage unit 115, interface 120 and peripheral devices 125 are in communication with the CPU 105 through a communication bus (solid lines), such as a motherboard. The storage unit 115 can be a data storage unit (or data repository) for storing data. The computer system 101 can be operatively coupled to a computer network (“network”) 130 with the aid of the communication interface 120. The network 130 can be the Internet, an internet and/or extranet, or an intranet and/or extranet that is in communication with the Internet. The network 130 in some cases is a telecommunication and/or data network. The network 130 can include one or more computer servers, which can enable distributed computing, such as cloud computing. The network 130, in some cases with the aid of the computer system 101, can implement a peer-to-peer network, which may enable devices coupled to the computer system 101 to behave as a client or a server.
[00156] The CPU 1105 can execute a sequence of machine-readable instructions, which can be embodied in a program or software. The instructions may be stored in a memory location, such as the memory 110. The instructions can be directed to the CPU 105, which can subsequently program or otherwise configure the CPU 105 to implement methods of the present disclosure. Examples of operations performed by the CPU 105 can include fetch, decode, execute, and writeback.
[00157] The CPU 105 can be part of a circuit, such as an integrated circuit. One or more other components of the system 101 can be included in the circuit. In some cases, the circuit is an application specific integrated circuit (ASIC).
[00158] The storage unit 115 can store files, such as drivers, libraries and saved programs. The storage unit 115 can store user data, e.g., user preferences and user programs. The computer system 101 in some cases can include one or more additional data storage units that are external to the computer system 101, such as located on a remote server that is in communication with the computer system 101 through an intranet or the Internet. [00159] The computer system 101 can communicate with one or more remote computer systems through the network 130. For instance, the computer system 101 can communicate with a remote computer system of a user (e.g., a physician). Examples of remote computer systems include personal computers (e.g., portable PC), slate or tablet PC’s (e.g., Apple® iPad, Samsung® Galaxy Tab), telephones, Smart phones (e.g., Apple® iPhone, Android- enabled device, Blackberry®), or personal digital assistants. The user can access the computer system 101 via the network 130.
[00160] Methods as described herein can be implemented by way of machine (e.g., computer processor) executable code stored on an electronic storage location of the computer system 101, such as, for example, on the memory 110 or electronic storage unit 115. The machine executable or machine readable code can be provided in the form of software. During use, the code can be executed by the processor 105. In some cases, the code can be retrieved from the storage unit 1115 and stored on the memory 110 for ready access by the processor 105. In some situations, the electronic storage unit 115 can be precluded, and machine- executable instructions are stored on memory 110.
[00161] The code can be pre-compiled and configured for use with a machine having a processer adapted to execute the code, or can be compiled during runtime. The code can be supplied in a programming language that can be selected to enable the code to execute in a pre-compiled or as-compiled fashion.
[00162] Aspects of the systems and methods provided herein, such as the computer system 101, can be embodied in programming. Various aspects of the technology may be thought of as “products” or “articles of manufacture” typically in the form of machine (or processor) executable code and/or associated data that is carried on or embodied in a type of machine readable medium. Machine-executable code can be stored on an electronic storage unit, such as memory (e.g., read-only memory, random-access memory, flash memory) or a hard disk. “Storage” type media can include any or all of the tangible memory of the computers, processors or the like, or associated modules thereof, such as various semiconductor memories, tape drives, disk drives and the like, which may provide non-transitory storage at any time for the software programming. All or portions of the software may at times be communicated through the Internet or various other telecommunication networks. Such communications, for example, may enable loading of the software from one computer or processor into another, for example, from a management server or host computer into the computer platform of an application server. Thus, another type of media that may bear the software elements includes optical, electrical and electromagnetic waves, such as used across physical interfaces between local devices, through wired and optical landline networks and over various air-links. The physical elements that carry such waves, such as wired or wireless links, optical links or the like, also may be considered as media bearing the software. As used herein, unless restricted to non-transitory, tangible “storage” media, terms such as computer or machine “readable medium” refer to any medium that participates in providing instructions to a processor for execution.
[00163] Hence, a machine readable medium, such as computer-executable code, may take many forms, including but not limited to, a tangible storage medium, a carrier wave medium or physical transmission medium. Non-volatile storage media include, for example, optical or magnetic disks, such as any of the storage devices in any computer(s) or the like, such as may be used to implement the databases, etc. shown in the drawings. Volatile storage media include dynamic memory, such as main memory of such a computer platform. Tangible transmission media include coaxial cables; copper wire and fiber optics, including the wires that comprise a bus within a computer system. Carrier-wave transmission media may take the form of electric or electromagnetic signals, or acoustic or light waves such as those generated during radio frequency (RF) and infrared (IR) data communications. Common forms of computer-readable media therefore include for example: a floppy disk, a flexible disk, hard disk, magnetic tape, any other magnetic medium, a CD-ROM, DVD or DVD- ROM, any other optical medium, punch cards paper tape, any other physical storage medium with patterns of holes, a RAM, a ROM, a PROM and EPROM, a FLASH-EPROM, any other memory chip or cartridge, a carrier wave transporting data or instructions, cables or links transporting such a carrier wave, or any other medium from which a computer may read programming code and/or data. Many of these forms of computer readable media may be involved in carrying one or more sequences of one or more instructions to a processor for execution.
[00164] The computer system 101 can include or be in communication with an electronic display 135 that comprises a user interface (UI) 140 for providing, for example, a chromatogram of the subject. Examples of UFs include, without limitation, a graphical user interface (GUI) and web-based user interface. [00165] Methods and systems of the present disclosure can be implemented by way of one or more algorithms. An algorithm can be implemented by way of software upon execution by the central processing unit 105. The algorithm can, for example, determine if a subject’s chromatogram matches that of a healthy patient’s profile or that it matches that of a patient with a known disease.
[00166] Example 1
[00167] A patient is diagnosed with pancreatic cancer and the patient’s breath is tested with the gas chromatography device described herein. The patient’s chromatogram displays reduced levels of two volatile organic compounds from Table 1 as compared to a subject without the disease. The two compounds are administered to the patient who has pancreatic cancer by a nebulizer twice a day for six weeks and during this time the patient’s chromatogram is monitored for changes in the presence and concentration of volatile organic compounds. After one week, the levels of the two chemicals begin to rise in the patient. An imaging diagnostic test is administered, and cancer is shown to be decreasing in size as compared to before the treatment with the two compounds. A biopsy is taken of the pancreatic cancer. A portion of the cancer cells are tested in vitro, and administration of the compound to the cancer cells demonstrates that they are killed on contact by the two compounds.
[00168] Example 2
[00169] A patient is diagnosed with lymphatic cancer and the patient’s serum is tested with the gas chromatography device described herein. The patient’s chromatogram displays reduced levels of a volatile organic compound from Table 1 as compared to a subject without the disease. The compound is administered to the patient who has lymphatic cancer by intravenous administration each day for eight weeks. The patient’s volatile organic compound chromatogram is monitored every three days by the device for changes in the presence and concentration of volatile organic compounds. After two weeks, the levels of the compound begin to rise in the patient. A biopsy sample is administered at the end of treatment, and the lymphatic cancer cells are shown to be reduced in number as compared to before the treatment with the compound. A portion of the cancer cells are tested in vitro , and administration of the compound for two hours to the cancer cells kills the cancer cells.
[00170] Example 3
[00171] A patient is diagnosed with endometrial cancer and the patient’s serum is tested with the gas chromatography device described herein. The patient’s chromatogram displays increased levels of three volatile organic compound from Table 1 as compared to a subject without the disease. The compounds are removed by dialysis every four days for ten weeks. The patient’s volatile organic compound chromatogram is monitored every day by the device for changes in the presence and concentration of volatile organic compounds. After three weeks, the levels of the three chemicals begin to rise in the patient. An imaging diagnostic test and a blood diagnostic is administered at the end of treatment, and the cancer is shown to be in remission as compared to before the dialysis treatment that removed the three compounds.
[00172] Example 4
[00173] A patient arrives to a health care provider and is having trouble breathing. A patient’s breath and saliva are tested with the gas chromatography device described herein. The patient’s chromatogram displays reduced levels of two volatile organic compound from Table 1 as compared to a subject without the disease. This chromatogram pattern is associated with lung cancer. An imaging diagnostic test is performed and confirms the patient has lung cancer. The compounds are administered to the patient who has lung cancer by an inhaler each day for twenty weeks. The patient’s volatile organic compound chromatogram is monitored every two days by the device for changes in the presence and concentration of volatile organic compounds. After six weeks, the levels of the compounds begin to rise in the patient. An imaging diagnostic test and a biopsy sample is administered at the end of treatment. The diagnostic image displays a reduction in the size of the cancer. A portion of the cancer cells from the biopsy are tested in vitro , and administration of the two compounds inhibit cell growth of the cancer cells.
[00174] Example 5
[00175] A patient is diagnosed with a melanoma and the patient’s skin is tested with the gas chromatography device described herein. The patient’s chromatogram displays reduced levels of four volatile organic compounds from Table 1 as compared to a subject without melanoma. The four compounds are administered to the patient who has melanoma by a applying a patch that contains the four compounds directly to the site of the melanoma, each day for nine weeks. The patients skin sample is tested throughout the treatment for changes in the presence and concentration of the volatile organic compounds. After four days, the levels of the four chemicals begin to rise in the patient. An imaging diagnostic test is administered, and cancer is shown to be decreasing in size as compared to before the treatment with the four compounds. A biopsy is taken of the melanoma and the cancer cells are reduced. A portion of the patient’s cancer cells and the patient’s natural killer (NK) cells are cocultured in vitro, and administration of the four compounds to the coculture demonstrates the NK cells begin to kill the cancer cells after administration of the four compounds.
[00176] Example 6
[00177] A patient arrives to a health care provider and is having trouble breathing. A patient’s breath is tested with the gas chromatography device described herein. The patient’s chromatogram displays increased levels of two volatile organic compound from Table 1 as compared to a subject without the disease. This chromatogram pattern is associated with SARS-CoV-2. A treatment is administered for the SARS-CoV-2 infection and the patient’s volatile organic compound chromatogram is monitored every day by the device for changes in the presence and concentration of volatile organic compounds. After three days, the levels of the two compounds begin to rise in the patient. A polymerase chain reaction (PCR) diagnostic test is administered at the end of treatment and the patient displays a normal level of the two compounds. The diagnostic test shows the patient is no longer positive for the virus.
[00178] Example 7
[00179] A patient arrives to a health care provider and is diagnosed with dementia. A patient’s blood is tested with the gas chromatography device described herein. The patient’s chromatogram displays decreased levels of two volatile organic compound from Table 1 as compared to a subject without the disease. This chromatogram pattern is associated with dementia. The compounds are administered to the patient by a pill once a day for ten years. The patient’s volatile organic compound chromatogram is monitored every week by the device for changes in the presence and concentration of volatile organic compounds. After five weeks, the levels of the two compounds begin to rise in the patient. A physical diagnostic test is administered after four months of treatment. The test shows the dementia symptoms have not progressed. After 8 months of treatment the patient is displaying normal levels of the two compounds and the symptoms of dementia have diminished.
[00180] Example 8
[00181] A basal cell carcinoma cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
[00182] A squamous cell carcinoma cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
[00183] An adenocarcinoma cancer cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
[00184] An osteosarcoma cancer cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
[00185] A soft tissue sarcoma cancer cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound. [00186] A rhabdomyosarcoma cancer cell is treated with a volatile organic compound from
Table 1 and the cell is killed after contact with the organic compound.
[00187] A B-cell acute lymphoblastic leukemia cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
[00188] Example 9
[00189] A S. aureus cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
[00190] An E. coli cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
[00191] AM tuberculosis cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
[00192] AN gonorrhoeae cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
[00193] A C. trachomatic cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
[00194] A Plasmodium cell is treated with a volatile organic compound from Table 1 and the cell is killed after contact with the organic compound.
[00195] Example 10
[00196] The device is used to diagnose an infectious disease the SARS-CoV-2 virus. A new outbreak occurs and there is no current diagnostic available. The device disclosed herein, is immediately used to diagnose patients with a different pattern in their chromatogram. The chromatography profile is improved when more people are tested by algorithms. The device is linked to a database, so information is updated immediately from other testing sites. The metabolite profile pattern recognition algorithm is be updated over the air remotely and in real time. The disease can be diagnosed before antibodies appear. [00197] While exemplary embodiments have been shown and described herein, such embodiments are by way of example only. Numerous variations, changes, and substitutions can be performed on the exemplary embodiments. It should be understood that various alternatives to the embodiments described herein may be employed.

Claims

CLAIMS WHAT IS CLAIMED IS:
1. A method of treating or preventing a disease or condition comprising:
(i) administering to a subject a therapeutically effective amount of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof;
(ii) directly or indirectly at least partially increasing in the subject a level, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof;
(iii) directly or indirectly at least partially reducing in the subject a level, an activity, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof, or
(iv) any combination of i, ii, or iii.
2. A method comprising detecting a presence and a level of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof, from a sample of a subject having or suspected of having a disease or condition and comparing the level of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof, to a reference level; or detecting the presence, the levels, the elution times, of a plurality of compounds of Table 1, homologs thereof, derivatives thereof, esters thereof, enantiomers thereof, diastereomers thereof, racemates thereof, salts thereof, or any combination thereof and comparing a pattern thereof to a reference pattern.
3. The method of claim 2, wherein the method is a method of diagnosing a disease or a condition and further comprising at least one of the following: selecting a therapeutic regiment for treating the disease or the condition, selecting a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof to treat the disease or condition, treating the subject for the disease or condition, or any combination thereof.
4. The method of claim 2, comprising detecting:
(i) a presence and a level of a first compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof;
(ii) a presence and a level of a second compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof;
(iii) a presence and a level of a third compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof; and
(iv) comparing the first level to a first reference level, the second level to a second reference level, and the third level to a third reference level.
5. The method of any one of claims 2-4, wherein the method is conducted at least in part on a diagnostic device, a screening device, or a diagnostic and a screening device comprising at least one of; a chromatography column, an eluant, a detector, and a mass spectrometer.
6. The method of claim 5, wherein the chromatography column comprises a gas chromatography column and the eluant comprises a gas.
7. The method of claim 5, wherein the diagnostic device, the screening device, or the diagnostic and the screening device comprises a microelectromechanical system (MEMS).
8. The method of claim 5, wherein the diagnostic device, the screening device, or the diagnostic and the screening device comprises a breath collection tube.
9. The method of claim 5, wherein the diagnostic device, the screening device, or the diagnostic and the screening device has:
(i) a positive volume of less than about 16 cubic feet,
(ii) a method of transport, wherein the device is moved by a robot, a drone, or a remote-control transport machine;
(iii) a self-contained power supply, or
(iv) or any combination of (i)-(iii).
10. The method of claim 9, wherein the robot, the drone, or the remote-control transport machine is controlled autonomously.
11. The method of claim 9, wherein the diagnostic device, the screening device, or the diagnostic and screening device is linked wirelessly or directly to a processing station.
Ill
12. The method of claim 9, wherein the diagnostic device, the screening device, or the diagnostic and screening device is linked wirelessly by Wi-Fi, a broadband cellular network, or any combination thereof
13. The method of claim 11, wherein the processing station analyzes the data remotely.
14. The method of any one of claims 5-12, wherein the device is about: 50 cm long, 30 cm wide, and 25 cm tall.
15. The method of any one of claims 5-12, wherein the device is less than about: 12 cm long, 7 cm wide, and 3 cm tall.
16. The method of claim 5, wherein an additional diagnostic test is performed on the subject.
17. The method of any one of claims 2-5, wherein the sample comprises a bodily fluid, a compound extruded by a body, a bodily part, a blood sample, a plasma sample, a urine sample, a sweat sample, a breath sample, a skin sample, a buccal sample, a hair sample, a fecal sample, a tear sample, a nasal sample, a nasal mucus sample, a biopsy sample, a saliva sample, a sputum sample or any combination thereof.
18. The method of claim 17, wherein the compound extruded by the body comprises an ester, a salt, or a combination thereof of the compound.
19. The method of claim 17, wherein the compound extruded by the body comprises a volatile compound.
20. The method of claim 17, wherein the compound extruded by the body comprises a metabolite.
21. The method of claim 17, wherein the sample further comprises an additive, a preservative, an anticoagulant, a membrane, an antibacterial compound, an antifungal compound, an antiviral, a chemical reagent, an antimicrobial compound, a diluent, or any combination thereof.
22. The method of claim 17, wherein the sample is treated with an electromagnetic wave, an ultrasound, a light wave, a radiation, a laser, adjusting a temperature of the sample, or any combination thereof.
23. The method of any one of claims 4-21, wherein the method is used to identify a disease or condition, wherein the sample is taken from the subject and applied to a device, wherein a computer is used to at least partially compare a chromatogram in the subject having a disease to a chromatogram of a subject not having the disease, and determining independently the difference in the presence, the absence, the level, or any combination thereof of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof.
24. The method of claim 23, wherein the chromatogram comprises a signature of the disease or the condition.
25. The method of claim 23, wherein the results generated by the method are evaluated by an algorithm using a processor or microprocessor.
26. The method of claim 25, wherein the algorithm comprises a trained algorithm, a machine learning algorithm, or any combination thereof using a processor or microprocessor.
27. The method of claim 23, wherein the computer comprises a memory, one or more executable instructions, one or more processors, and a graphical user interface.
28. The method of any one of claims 23-27, wherein a result is communicated via a communication medium.
29. The method of any one of claims 2-28, wherein the compound of Table 1 identified by the method, or the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof is selected as a therapeutic.
30. The method of any one of claims 2-29, wherein the method comprises a method of diagnosis, of prognosis, a theranostic method, a health level information method, or any combination thereof.
31. The method of claim 30, wherein when the subject displays an altered profile, a level of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof is at least partially increased or at least partially decreased.
32. The method of claim 31, wherein a healthy subject’s metabolic profile is recorded for at least 2 weeks.
33. The method of claim 32, wherein the healthy subject’s profile is recorded a plurality of times over a period of at least two weeks.
34. The method of claim 31, wherein a metabolic profile of the subject is unknown, wherein a healthy metabolic profile is determined from factors comprising the subject’s genetic profile, the diseased metabolic profile, a healthy population profile, or any combination thereof.
35. The method of any one of claims 2-34, wherein the method is performing a diagnostic test and administering a therapeutic comprising the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof, wherein the diagnostic test comprises a diagnostic test, a prognostic test or a theranostic test, wherein running the diagnostic test comprises subjecting at least a portion of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof to a device comprising: a chromatography column, an eluant, a detector, and a mass spectrometer.
36. A method of promoting or maintaining health in a subject comprising:
(i) administering to the subject a therapeutically effective amount of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof;
(ii) directly or indirectly at least partially increasing in the subject a level, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof;
(iii) directly or indirectly at least partially reducing in the subject a level, an activity, a half-life, or any combination thereof of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof or any combination thereof, or
(iv) any combination of i, ii, or iii.
37. The method of claim 36, wherein the administering comprises administering an oral formulation.
38. The method of claim 37, wherein the oral formulation is a dietary supplement
39. The method of claim 36, wherein the promoting or maintaining health comprises promoting or maintaining: cardiovascular health, respiratory health, skeletal health, urogenital health, digestive health, muscular health, neural health, immune system health, liver health, endocrine health or any combination thereof
40. The method of claim 1, comprising (i).
41. The method of claim 40, wherein the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof is comprised in a pharmaceutical composition comprising an excipient, a diluent, or a carrier.
42. The method of claim 41, wherein the pharmaceutical composition comprises a first compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof; and a second compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof
43. The method of claim 41, wherein the pharmaceutical composition is in unit dose form.
44. The method of any one of claims 40-43, wherein the pharmaceutical composition comprises a molecule selected from the group consisting of: isoprene, decanal, 4- methylbenzaldehyde, 3,7-dimethyloctan-3-ol, l-methyl-4(l -methyl -2 -propenyl)- benzene, l-ethenyl-3 -ethyl-benzene, l-ethenyl-4-ethyl-benzene, 1-phenylbut-l-ene, octan-2-one, heptan-3-one, hexan-3-one 5-methyl, butanoic acid 2-methyl-ethyl ester, propionic acid, 2-methoxyethanol, acetaldehyde, propanal, n-propyl acetate, methyl methacrylate, styrene, 1,1-dipropoxypropane, a homolog of any of these, a derivative of any of these, an ester of any of these, a enantiomer of any of these, a diastereomer of any of these, a racemate of any of these, a salt of any of these, and any combination of any of these.
45. The method of claim 41, wherein the pharmaceutical composition further comprises albuterol or a salt thereof.
46. The method of claim 45, wherein the pharmaceutical composition is administered with an inhaler, a nebulizer, a vape device, a bronchodilator, a face mask, a nasal cannula, a diffuser, a tent, a container at least partially surrounding the subjects head, or any combination thereof.
47. The method of claim 1, wherein the method further comprises administering a plant, a portion of a plant, an extract thereof, an herb, a portion of an herb, an extract thereof, or any combination thereof.
48. The method of claim 1, comprising (i), wherein the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof or any combination thereof is comprised in an implant, wherein the implant at least partially releases the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, or the salt thereof, or any combination thereof over a time when at least partially implanted in the subject.
49. The method of claim 1, comprising (i), wherein a starting material is administered to a subject and the starting material is transformed into the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof.
50. The method of claim 1, comprising (i), wherein the compound of Table 1, homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof is covalently linked to an antibody or fragment thereof.
51. The method of claim 1, comprising (i), wherein the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof is covalently linked by a cleavable linker to an antibody or fragment thereof.
52. The method of claim 1, comprising (i), wherein a cell endocytosis is at least partially promoted in the subject.
53. The method of claim 52, wherein the cell endocytosis is of a polypeptide, a virus, a lipid, a polynucleotide, a molecule, or any combination thereof.
54. The method of claim 1, comprising (i), wherein the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof is administered and at least partially crosses the blood brain barrier in the subject.
55. The method of claim 1, comprising (i), wherein an apoptosis or a cell death is at least partially induced in the subject.
56. The method of claim 55, wherein the induction is at least in part a selective induction.
57. A method comprising; contacting an isolated cell with a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof wherein after the contact the isolated cell undergoes apoptosis or cell death.
58. The method of claim 1, comprising (i), wherein a cell growth, a cell communication, a cell metastasis, or any combination thereof is at least partially inhibited in the subject.
59. The method of claim 58, wherein the inhibition is at least in part a selective inhibition.
60. The method of claim 1, comprising (i), wherein a Reactive Oxygen Species (ROS) is at least partially induced within the subject.
61. A method comprising; contacting an isolated cell with a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof, wherein the isolated cell produces ROS.
62. The method of claim 61, wherein the method is a method of screening.
63. The method of any one of claims 55-61, wherein the cell is a cancer cell.
64. The method of claim 1, comprising (i), wherein an immune cell is at least partially deactivated in the subject.
65. The method of claim 64, wherein the deactivation is at least in part a selective deactivation.
66. The method of claim 1, comprising (i), wherein an immune cell is at least partially activated in the subject.
67. The method of claim 66, wherein the activation is at least in part a selective activation.
68. The method of claim 1, comprising (i), wherein an immune cell is at least partially induced into maturation, differentiation, or a combination thereof.
69. The method of claim 66, wherein the immune cell comprises a dendritic cell, a neutrophil, a granulocyte, a monocyte, a lymphocyte, or any combination thereof.
70. The method of claim 66, wherein the immune cell comprises a dendritic cell, a CD4 T-cell, a CD8 T-cell, or any combination thereof.
71. The method of claim 70, wherein the dendritic cell, the CD4 T-cell, the CD8 T-cell, or any combination thereof at least partially directly or at least partially indirectly suppresses a tumor in the subject.
72. The method of claim 66, wherein the immune cell is a natural killer (NK) cell.
73. The method of claim 72, wherein the NK cell at least partially removes alpha- synuclein aggregates in the subject.
74. The method of claim 1, comprising (i), wherein the treating or preventing disease comprises administering a pharmaceutical composition comprising an excipient, a carrier, a diluent or any combination of these and the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof.
75. The method of claim 1, comprising (ii), wherein the treating or preventing comprises administering a polynucleotide to a subject, wherein the polynucleotide encodes for a polypeptide or biologically active fragment thereof in a biochemical pathway for making the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof.
76. The method of claim 1, comprising (ii), wherein the treating or preventing disease comprises administering a genetically engineered virus comprising at least one polynucleotide coding for a polypeptide, or a biologically active fragment thereof, in a pathway to produce the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof.
77. The method of claim 1, comprising (ii), wherein the treating or preventing disease comprises administering a vector comprising at least one polynucleotide coding for a polypeptide, or a biologically active fragment thereof, in a pathway to produce the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof.
78. The method of claim 77, wherein the vector is a plasmid.
79. The method of claim 1, comprising (ii) or (iii), wherein the treating or preventing disease comprises administering a genetically engineered cell to capture, to produce, or to chemically alter the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof.
80. The method of claim 79, wherein the genetically engineered cell is a eukaryotic cell.
81. The method of claim 79, wherein the genetically engineered cell is a prokaryotic cell.
82. The method of claim 1, comprising (ii) or (iii), comprising altering a composition of a food, a beverage, a nutrient, or any combination thereof consumed by the subject.
83. The method of claim 1, comprising (ii) or (iii), wherein the treating or preventing disease comprises administering a compound to the subject, wherein the compound is an at least partial inhibitor, a salt thereof, or a formulation containing at least one of these, of a polypeptide or biologically active fragment thereof involved in a competing biological pathway, wherein the production of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof is altered.
84. The method of claim 1, comprising (ii) or (iii), wherein the treating or preventing disease comprises administering an intermediate compound, a salt thereof, or a formulation thereof in a biological pathway, wherein the production of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof is altered.
85. The method of claim 1, comprising (ii) or (iii), wherein the treating or preventing disease comprises at least partially disrupting a communication between one or more cells in the subject.
86. The method of claim 1, comprising (iii) wherein the treating or preventing disease comprises editing a polynucleotide, wherein the polynucleotide encodes for a polypeptide in a biochemical pathway for making the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, or the salt thereof.
87. The method of claim 86, wherein the polynucleotide comprises a ribose.
88. The method of claim 86, wherein the polynucleotide comprises a deoxyribose.
89. The method of claim 86, wherein the edited polynucleotide is comprised in an isolated cell and the isolated cell is administered to the subject.
90. The method of claim 89, wherein the isolated cell is autologous.
91. The method of claim 89, wherein the isolated cell is allogenic.
92. The method of claim 1, comprising (iii), wherein the treating or preventing disease comprises administering an at least partial inhibitor, a salt thereof, or a formulation containing at least one of these: of a polypeptide or biologically active fragment thereof, involved in the production of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof.
93. The method of claim 1, comprising (iii), wherein the treating or preventing disease comprises administering an at least partially interfering polynucleotide to bind to RNA or DNA to at least partially inhibit polypeptide production in a biochemical pathway for making the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof or any combination thereof in the subject.
94. The method of claim 93, wherein the polynucleotide comprises a ribose.
95. The method of claim 93, wherein the polynucleotide comprises a deoxyribose.
96. The method of claim 1, comprising (iii), wherein the treating or preventing disease comprises administering a CRISPR system to edit a polynucleotide to at least partially inhibit polypeptide production in a biochemical pathway for making the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof in the subject.
97. The method of claim 93, wherein the polynucleotide comprises a ribose.
98. The method of claim 93, wherein the polynucleotide comprises a deoxyribose.
99. The method of claim 96, wherein the edited polynucleotide comprises a nonsense mutation, a missense mutation, a deletion, or any combination thereof.
100. The method of claim 1, comprising (iii), wherein the treating or preventing disease comprises administering an antibody, an aptamer, or a fragment thereof to at least partially bind to the compound of Table 1 the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof in the subject.
101. The method of claim 1, comprising (iii) wherein the treating or preventing disease comprises administering a molecule, a salt thereof, or a formulation containing at least one of these to capture the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof or any combination thereof in the subject.
102. The method of claim 1, comprising (iii), wherein the treating or preventing disease comprises administering dialysis to a patient, wherein the dialysis filters at least some of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof from the patient.
103. The method of claim 1, wherein the disease comprises a cancer, a tumor, an infectious disease, a skeletal disease, a urogenital disease, a digestive disease, a muscular disease, a neurological disease, an autoimmune disease, a respiratory disease, a cardiac disease, an endocrine disease, a liver disease, a hereditary disease, or any combination thereof.
104. The method of claim 103, wherein the disease comprises a cancer, wherein the cancer comprises a sarcoma, a carcinoma, a melanoma, a lymphoma, leukemia, blastoma, germ cell tumor, myeloma, or any combination thereof.
105. The method of claim 103, wherein the disease comprises an infectious disease, wherein the infectious disease comprises a viral infection.
106. The method of claim 105, wherein the viral infection comprises, influenza A, influenza B, SARS-CoV-2 (COVID-19), SARS-COV, MERS-CoV, HKU1 coronavirus, OC43 coronavirus, NL63 coronavirus, 229E coronavirus, hepatitis A, hepatitis B, hepatitis C, norovirus, rotavims, measles, rubella, HIV, enterovirus, herpes simplex 1, herpes simplex 2, varicella zoster virus, ebola virus, dengue virus or any combination thereof.
107. The method of claim 103, wherein the disease comprises an infectious disease, wherein the infectious disease comprises a bacterial infection.
108. The method of claim 107, wherein the bacterial infection comprises Helicobacter pylori, Staphylococcus aureus, Mycobacterium tuberculosis, Escherichia coli, Campylobacter jejuni, Klebsiella pneumonia, Streptococcus pyogenes, Neisseria gonorrhoeae, Treponema pallidum, Borrelia burgdorferi, Neisseria meningitidis, Chlamydia trachomatis, Salmonella, Vibrio, Enterobacteriaceae, or any combination thereof.
109. The method of claim 103, wherein the disease comprises the infectious disease, wherein the infectious disease comprises a fungal, a parasitic infection, or any combination thereof.
110. The method of any one of claims 105-109, wherein the infection is a drug resistant infection.
111. The method of claim 110, wherein the infection is a multi-drug resistant infection.
112. The method of claim 103, wherein the disease comprises the skeletal disease, wherein the skeletal disease comprises osteoarthritis, osteoporosis, Paget disease of bone, or any combination thereof.
113. The method of claim 103, wherein the disease comprises the urogenital disease, wherein the urogenital disease comprises benign prostate hyperplasia, urinary incontinence, kidney stones, kidney disease, erectile dysfunction, interstitial cystitis, prostatitis, overactive bladder, endometriosis, or any combination thereof.
114. The method of claim 103, wherein the disease comprises the digestive disease, wherein the digestive disease comprises irritable bowel syndrome, celiac disease, gastroesophageal reflux disease, Chron’s disease, gastroparesis, or any combination thereof.
115. The method of claim 103, wherein the disease comprises the muscular disease, wherein the muscular disease comprises muscular dystrophy, amyotrophic lateral sclerosis, Charcot-Marie-Tooth disease, myasthenia gravis, myopathy, myositis, peripheral neuropathy, or any combination thereof.
116. The method of claim 103, wherein the disease comprises the neurological disease, wherein the neurological disease comprises psychiatric disease, spinal cord injury, epilepsy, headache, migraine, dementia, vertigo, seizures, dizziness, sleep disorder, aneurysm, chronic fatigue syndrome, cerebral palsy, amyotrophic lateral sclerosis, neuralgia, neuropathy, or any combination thereof
117. The method of claim 103, wherein the disease comprises the autoimmune disease, wherein the autoimmune disease comprises type 1 diabetes, rheumatoid arthritis, psoriasis, multiple sclerosis, systemic lupus erythematosus, Sjogren’s syndrome, an allergy, or any combination thereof.
118. The method of claim 103, wherein the disease comprises the respiratory disease, wherein the respiratory disease comprises asthma, chronic obstructive pulmonary disease, chronic bronchitis, emphysema, cystic fibrosis, plural effusion, or any combination thereof.
119. The method of claim 103, wherein the disease comprises the cardiac disease, wherein the cardiac disease comprises congenital heart disease, arrhythmia, coronary artery disease, dilated cardiomyopathy, myocardial infarction, congestive heart failure, high blood pressure, mitral regurgitation, pulmonary stenosis, or any combination thereof.
120. The method of claim 103, wherein the disease comprises the endocrine disease, wherein the endocrine disease comprises type 2 diabetes, hypothyroidism, hyperthyroidism, low testosterone, obesity, Addison’s disease, Cushing’s syndrome, Graves’ disease, thyroiditis, prolactinoma, or any combination thereof.
121. The method of claim 103, wherein the disease comprises the liver disease, wherein the liver disease is cirrhosis, fibrosis, hemochromatosis, Wilson’s disease, or any combination thereof.
122. The method of claim 103, wherein the disease comprises the hereditary disease, wherein the hereditary disease comprises, an inherited mendelian disorder, a peroxisomal disorder, a mitochondrial disorder, cystic fibrosis, sickle-cell anemia, Marfan syndrome, Huntington’s disease, kidney disease, Tay-Sachs disease, phenylketonuria, hemophilia, or any combination thereof.
123. The method of claim 1, wherein the treating or preventing the disease, comprises administering a second therapy.
124. The method of claim 123, wherein the second therapy for cancer comprises surgery, chemotherapy, radiation therapy, immunotherapy, hormone therapy, a checkpoint inhibitor, targeted drug therapy, CAR T-cell therapy, or a combination thereof.
125. The method of claim 123, wherein the administering of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof and the second administering are concurrent.
126. The method of claim 123, wherein the administering of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof and the second administering are sequential.
127. The method of claim 1, comprising (i), wherein the method of treating or preventing disease is administered by: injection, inhalation, catheterization, gastrostomy tube administration, intravenous administration, intraosseous administration, ocular administration, otic administration, topical administration, transdermal administration, oral administration, rectal administration, nasal administration, intravaginal administration, intracavemous administration, transurethral administration, buccal administration, sublingual administration, or a combination thereof.
128. The method of claim 127, comprising (i), wherein the administration comprises administering a nanoparticle, a viral vector, a viral-like particle, a liposome, an exosome, an extracellular vesicle, a microrobot, a microneedle, an implant, or a combination thereof.
129. The method of claim 127, comprising (i), wherein the administration comprises a topical administration, wherein the topical administration comprises administering a patch, a lotion, a cream, a gel, a spray, an ointment, a liquid formulation, or a combination thereof.
130. The method of claim 129, wherein a patch comprises a microneedle patch, a single-layer drug in adhesive, a multi-layer drug in adhesive, a reservoir system, a matrix system, a vapour patch, or any combination thereof.
131. The method of claim 127, comprising (i), wherein the administration comprises an injection, wherein the injection comprises administering an injector, intraarterial injection, intracerebroventricular injection, intraci sternal injection, intramuscular injection, intraorbital injection, intraparenchymal injection, intraperitoneal injection, intraspinal injection, intrathecal injection, intravenous injection, intraventricular injection, stereotactic injection, subcutaneous injection, or any combination thereof.
132. The method of claim 131, wherein the injection is administered directly to the site of the disease.
133. The method of claim 127, comprising (i), wherein the administration comprises an oral or rectal administration, wherein the oral or rectal administration comprises administering a pill formulation, a liquid formulation, a powder formulation, or any combination thereof.
134. The method of claim 127, comprising (i), wherein the administration comprises an inhalation administration, wherein the inhalation administration comprises administering an inhaler, a nebulizer, a vape device, a bronchodilator, a face mask, nasal cannula, a diffuser, a tent, a container at least partially surrounding the subjects head, an aerosol formulation, a powder formulation, a liquid formulation, a gas formulation, or any combination thereof.
135. The method of claim 1, comprising (i), wherein the administering is at least about once per day, twice per day, three times per day, or 4 times per day.
136. The method of claim 135, wherein the administering is continuous.
137. The method of claim 136, wherein the administering is by a pump or an implant.
138. The method of claim 1, comprising (i), wherein the time of administration comprises for at least about: 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months 11 months, 1 year, 2 years, 3 years, 4 years, 5 years, or for life.
139. The method of claim 1, comprising (i) wherein the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof can have an identical or a different dosage.
140. The method of claim 1, comprising (i), wherein a compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof can have an identical or a different route of administration.
141. A pharmaceutical composition comprising an excipient, a carrier, a diluent and a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
142. The composition of claim 141, wherein the pharmaceutical composition is in unit dose form.
143. The composition of claim 141, wherein the pharmaceutical composition comprises a molecule selected from the group consisting of: isoprene, decanal, 4- methylbenzaldehyde, 3,7-dimethyloctan-3-ol, l-methyl-4(l -methyl -2 -propenyl)- benzene, l-ethenyl-3 -ethyl-benzene, l-ethenyl-4-ethyl-benzene, 1-phenylbut-l-ene, octan-2-one, heptan-3-one, hexan-3-one 5-methyl, butanoic acid 2-methyl-ethyl ester, propionic acid, 2-methoxyethanol, acetaldehyde, propanal, n-propyl acetate, methyl methacrylate, styrene, 1,1-dipropoxypropane, a homolog of any of these, a derivative of any of these, an ester of any of these, a salt of any of these, and any combination of any of these.
144. The composition of claim 141, wherein the pharmaceutical composition further comprises albuterol a salt thereof, or any combination thereof.
145. The composition of claim 144, wherein the pharmaceutical composition is administered with an inhaler, a nebulizer, a vape device, a bronchodilator, a face mask, nasal cannula, a diffuser, a tent, a container at least partially surrounding the subject’s head, or any combination thereof.
146. A pharmaceutical composition in unit dose form comprising, a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof, wherein when the composition in unit dose form is placed in a closed container and placed in a room at about 23 degrees Celsius with a relative atmospheric humidity of about 50% for at least about 1 month, wherein at the end of the time period at least about 80% of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof remains.
147. The composition of claim 146, wherein the time period is about 1 month to about 2 years
148. A method of treating or preventing a disease or condition comprising:
(i) detecting a level of a compound of Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof, from a sample of a subject having or suspected of having a disease or condition and comparing the level of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof, to a reference level and
(ii) administering to a subject a therapeutically effective amount of a compound of
Table 1, a homolog thereof, a derivative thereof, an ester thereof, an enantiomer thereof, a diastereomer thereof, a racemate thereof, a salt thereof, or any combination thereof.
149. The method of claim 148, wherein the method is conducted at least in part on a diagnostic device, a screening device, or a diagnostic and a screening device comprising at least one of; a chromatography column, an eluant, a detector, and a mass spectrometer.
150. The method of claim 149, wherein the chromatography column comprises a gas chromatography column and the eluant comprises a gas.
151. The method of claim 149, wherein the diagnostic device, the screening device, or the diagnostic and the screening device comprises a microelectromechanical system (MEMS).
152. The method of claim 149, wherein the diagnostic device, the screening device, or the diagnostic and the screening device comprises a breath collection tube.
153. The method of claim 149, wherein an increase or decrease in concentration of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof, indicates a treatment is at least partially decreasing the presence of the disease, disease symptoms, or a combination thereof.
154. The method of claim 149, wherein the increase or decrease in concentration of the compound of Table 1, the homolog thereof, the derivative thereof, the ester thereof, the enantiomer thereof, the diastereomer thereof, the racemate thereof, the salt thereof, or any combination thereof indicates a viral infection in the subject.
155. The method of claim 149, wherein the diagnostic device is operably linked to a database or is not linked to a database.
156. The method of claim 149, wherein the database comprises a profile pattern of different diseases.
157. The method of claim 149, wherein the device compares a patient’s profile pattern with the database’s profile to identify a disease.
PCT/US2021/026650 2020-04-09 2021-04-09 Small molecule drugs as targeted therapeutics WO2021207647A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063007889P 2020-04-09 2020-04-09
US63/007,889 2020-04-09

Publications (1)

Publication Number Publication Date
WO2021207647A1 true WO2021207647A1 (en) 2021-10-14

Family

ID=78023690

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/026650 WO2021207647A1 (en) 2020-04-09 2021-04-09 Small molecule drugs as targeted therapeutics

Country Status (1)

Country Link
WO (1) WO2021207647A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4241766A1 (en) * 2022-03-07 2023-09-13 Fondazione Per L'Istituto Oncologico Di Ricerca (IOR) Use of olfactory receptor or13a1 ligands in the treatment of lymphomas

Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5219740A (en) * 1987-02-13 1993-06-15 Fred Hutchinson Cancer Research Center Retroviral gene transfer into diploid fibroblasts for gene therapy
US20030176804A1 (en) * 2002-01-22 2003-09-18 Melker Richard J. Method and apparatus for monitoring respiratory gases during anesthesia
US20040127808A1 (en) * 2001-04-17 2004-07-01 Vaughan John W. Device and method for assessing asthma and other diseases
US20080306152A1 (en) * 2003-07-18 2008-12-11 Idexx Lboratories, Inc. Compositions containing prodrugs of florfenicol and methods of use
US20100143241A1 (en) * 2008-10-22 2010-06-10 Johnson G Allan Method and apparatus for delivery of agents across the blood brain barrier
US20120294876A1 (en) * 2011-05-19 2012-11-22 C-Lock Inc. Vaccine and health-related applications for ruminant breath monitoring system
WO2014165818A2 (en) * 2013-04-05 2014-10-09 T Cell Therapeutics, Inc. Compositions and methods for preventing and treating prostate cancer
US20160375189A1 (en) * 2013-03-14 2016-12-29 Baxter International Inc. System and method for performing alternative and sequential blood and peritoneal dialysis modalities
US20170065631A1 (en) * 2013-03-07 2017-03-09 Advanced Inhalation Therapies (Ait) Ltd. Inhalation of nitric oxide for treating respiratory diseases
US20180169154A1 (en) * 2015-05-13 2018-06-21 Synlogic, Inc. Bacteria engineered to treat a disease or disorder
US20180214490A1 (en) * 2012-07-11 2018-08-02 Sangamo Therapeutics, Inc. Methods of treating lysosomal storage diseases
US20180235752A1 (en) * 2015-08-26 2018-08-23 Medtronic Xomed, Inc. Sinus and nasal stent
US20180242859A1 (en) * 2006-12-19 2018-08-30 Valencell, Inc. Physiological monitoring methods
US20190300598A1 (en) * 2007-02-23 2019-10-03 Prothena Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
US20190374560A1 (en) * 2013-04-22 2019-12-12 Avelas Biosciences, Inc. Selective drug delivery compositions and methods of use

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5219740A (en) * 1987-02-13 1993-06-15 Fred Hutchinson Cancer Research Center Retroviral gene transfer into diploid fibroblasts for gene therapy
US20040127808A1 (en) * 2001-04-17 2004-07-01 Vaughan John W. Device and method for assessing asthma and other diseases
US20030176804A1 (en) * 2002-01-22 2003-09-18 Melker Richard J. Method and apparatus for monitoring respiratory gases during anesthesia
US20080306152A1 (en) * 2003-07-18 2008-12-11 Idexx Lboratories, Inc. Compositions containing prodrugs of florfenicol and methods of use
US20180242859A1 (en) * 2006-12-19 2018-08-30 Valencell, Inc. Physiological monitoring methods
US20190300598A1 (en) * 2007-02-23 2019-10-03 Prothena Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
US20100143241A1 (en) * 2008-10-22 2010-06-10 Johnson G Allan Method and apparatus for delivery of agents across the blood brain barrier
US20120294876A1 (en) * 2011-05-19 2012-11-22 C-Lock Inc. Vaccine and health-related applications for ruminant breath monitoring system
US20180214490A1 (en) * 2012-07-11 2018-08-02 Sangamo Therapeutics, Inc. Methods of treating lysosomal storage diseases
US20170065631A1 (en) * 2013-03-07 2017-03-09 Advanced Inhalation Therapies (Ait) Ltd. Inhalation of nitric oxide for treating respiratory diseases
US20160375189A1 (en) * 2013-03-14 2016-12-29 Baxter International Inc. System and method for performing alternative and sequential blood and peritoneal dialysis modalities
WO2014165818A2 (en) * 2013-04-05 2014-10-09 T Cell Therapeutics, Inc. Compositions and methods for preventing and treating prostate cancer
US20190374560A1 (en) * 2013-04-22 2019-12-12 Avelas Biosciences, Inc. Selective drug delivery compositions and methods of use
US20180169154A1 (en) * 2015-05-13 2018-06-21 Synlogic, Inc. Bacteria engineered to treat a disease or disorder
US20180235752A1 (en) * 2015-08-26 2018-08-23 Medtronic Xomed, Inc. Sinus and nasal stent

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
GUNTNER ET AL.: "Breath Sensors for Health Monitoring", ACS SENSORS, vol. 4, no. 2, 9 January 2019 (2019-01-09), pages 268 - 80, XP055677521, [retrieved on 20190109], DOI: 10.1021/acssensors.8b00937 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4241766A1 (en) * 2022-03-07 2023-09-13 Fondazione Per L'Istituto Oncologico Di Ricerca (IOR) Use of olfactory receptor or13a1 ligands in the treatment of lymphomas
WO2023169996A1 (en) 2022-03-07 2023-09-14 Fondazione Per L'istituto Oncologico Di Ricerca (Ior) Use of olfactory receptor or13a1 antagonists in the treatment of lymphomas

Similar Documents

Publication Publication Date Title
Wu et al. Solubility of Folic Acid in Water at pH Values between 0 and 7 at Temperatures (298.15, 303.15, and 313.15) K
Simile et al. Targeted therapies in cholangiocarcinoma: emerging evidence from clinical trials
Pawar et al. Interaction of repaglinide with bovine serum albumin: Spectroscopic and molecular docking approaches
Eckford et al. Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) Potentiator VX-770 (Ivacaftor) Opens the Defective Channel Gate of Mutant CFTR in a Phosphorylation-dependent but ATP-independent Manner*♦
Uchiyama et al. Proton pump inhibitors and oncologic treatment efficacy: a practical review of the literature for oncologists
Schneider et al. Drug–drug plasma protein binding interactions of ivacaftor
Mertens et al. Next generation gold drugs and probes: chemistry and biomedical applications
Petruk et al. Interaction between proteins and Ir based CO releasing molecules: Mechanism of adduct formation and CO release
Mellion et al. Phase 1 clinical trial of losmapimod in facioscapulohumeral dystrophy: Safety, tolerability, pharmacokinetics, and target engagement
WO2021207647A1 (en) Small molecule drugs as targeted therapeutics
Bartos et al. Investigation of absorption routes of meloxicam and its salt form from intranasal delivery systems
Lin et al. Combined computational and experimental studies of molecular interactions of albuterol sulfate with bovine serum albumin for pulmonary drug nanoparticles
Izdebska et al. Involvement of actin in autophagy and autophagy-dependent multidrug resistance in cancer
JP2023022190A (en) cancer treatment
Karmacharya et al. Novel pyridine bioisostere of cabozantinib as a potent c-met kinase inhibitor: synthesis and anti-tumor activity against hepatocellular carcinoma
Leonardi et al. Heme oxygenase 1 impairs glucocorticoid receptor activity in prostate cancer
Martínez-Alonso et al. Preclinical characterization of antioxidant quinolyl nitrone QN23 as a new candidate for the treatment of ischemic stroke
US20170311597A1 (en) Inhibitors of grb2-associated binding protein 1 (gab1) and methods of treating cancer using the same
Russo et al. Synthesis and structure-activity relationships of amino acid conjugates of cholanic acid as antagonists of the EphA2 receptor
Perez et al. Effects of lysozyme on the activity of ionic of fluoroquinolone species
CN106031710A (en) Vonoprazan fumarate injection and preparation method thereof
US20160101194A1 (en) SMART® Medication Adherence Formulation, Method, Device and System for Topical, Vaginal or Rectal Routes of Administration
Torensma The dilemma of cure and damage in oligodendroglioma: Ways to tip the balance away from the damage
Zimmermann et al. Population pharmacokinetic modeling to establish the role of P-glycoprotein on ciprofloxacin distribution to lung and prostate following intravenous and intratracheal administration to Wistar rats
Hsiao et al. Effective perturbations of the amplitude, gating, and hysteresis of IK (DR) caused by PT-2385, an HIF-2α inhibitor

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: 21784156

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: 21784156

Country of ref document: EP

Kind code of ref document: A1