US20180340945A1 - Detection of biomarkers on vesicles for the diagnosis and prognosis of diseases and disorders - Google Patents

Detection of biomarkers on vesicles for the diagnosis and prognosis of diseases and disorders Download PDF

Info

Publication number
US20180340945A1
US20180340945A1 US15/989,120 US201815989120A US2018340945A1 US 20180340945 A1 US20180340945 A1 US 20180340945A1 US 201815989120 A US201815989120 A US 201815989120A US 2018340945 A1 US2018340945 A1 US 2018340945A1
Authority
US
United States
Prior art keywords
vesicles
protein
antibody
cancer
biomarker
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/989,120
Other languages
English (en)
Inventor
Masato Mitsuhashi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nanosomix Inc
Original Assignee
Nanosomix Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nanosomix Inc filed Critical Nanosomix Inc
Priority to US15/989,120 priority Critical patent/US20180340945A1/en
Publication of US20180340945A1 publication Critical patent/US20180340945A1/en
Assigned to NANOSOMIX, INC. reassignment NANOSOMIX, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MITSUHASHI, MASATO
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54313Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals the carrier being characterised by its particulate form
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/2814Dementia; Cognitive disorders
    • G01N2800/2821Alzheimer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/2835Movement disorders, e.g. Parkinson, Huntington, Tourette
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the present invention relates to methods, compositions, and kits for detecting and quantitating biomarkers on vesicles without lysis or permeabilization of the vesicles and the use of biomarkers identified on vesicles in diagnostic and prognostic methods for various diseases and disorders.
  • Disease and disorders of the present invention include neurological disorders, immunological disorders, placental diseases, and cancer.
  • Exosomes in biological fluids are potentially useful diagnostically for various diseases, because exosomes carry physiological and pathological materials (proteins, metabolites, RNAs, small molecules, etc.) of the mother cells from which they originate and the microenvironment near their mother cells.
  • physiological and pathological materials proteins, metabolites, RNAs, small molecules, etc.
  • exosomes are recognized as valuable resources for diagnostics, current analytical methods of analyzing exosomes are too complicated and expensive for routine use in diagnostic testing.
  • Alzheimer's disease the most common form of dementia.
  • Currently, the only definitive way to diagnose Alzheimer's disease is by direct examination of brain tissue after a patient dies. Doctors use brain imaging, evaluation of behavior, psychiatric tests, and other means to diagnose the disease in the patients suspected of having Alzheimer's disease, but none are highly accurate, and many are costly or not practical.
  • biomarkers and methods for diagnosing diseases and other disorders such as, for example, neurological disorders, cancer, immunological disorders, and placental disease.
  • compositions for detecting the biomarkers as well as compositions and methods useful for treating diseases and other disorders.
  • the present invention meets this need by providing accurate, noninvasive methods for diagnosing diseases and other disorders.
  • the present invention further provides novel methods, assays, biomarkers, kits, and compositions for diagnosing, prognosing, predicting, and treating various diseases and disorders.
  • the present invention relates to methods, compositions, and kits for detecting and quantitating biomarkers on vesicles.
  • the invention provides a method for selectively capturing vesicles carrying surface markers of interest on a solid support and detecting membrane-bound and adsorbed biomarkers on the captured vesicles without the need for lysis or permeabilization of the vesicles. Measurement of biomarkers on vesicles is useful in diagnostic and prognostic methods for various diseases.
  • the present invention provides methods of diagnosing or prognosing a disease or disorder in a subject, identifying a subject at risk of a disease or a disorder, or prescribing a therapeutic regimen or predicting benefit from therapy in a subject having a disease or a disorder.
  • the present invention provides methods of diagnosing or prognosing a neurological disorder in a subject, identifying a subject at risk of a neurological disorder, or prescribing a therapeutic regimen or predicting benefit from therapy in a subject having a neurological disorder.
  • the present invention provides methods of diagnosing or prognosing an immunological disorder in a subject, identifying a subject at risk of an immunological disorder, or prescribing a therapeutic regimen or predicting benefit from therapy in a subject having an immunological disorder.
  • the present invention provides methods of diagnosing or prognosing cancer in a subject, identifying a subject at risk of cancer, or prescribing a therapeutic regimen or predicting benefit from therapy in a subject having cancer.
  • the present invention provides methods of diagnosing or prognosing placental disease in a subject, identifying a subject at risk of a placental disease, or prescribing a therapeutic regimen or predicting benefit from therapy in a subject having a placental disease.
  • the invention provides a method comprising: a) providing a biological sample comprising vesicles; b) contacting a solid support comprising capture agents associated therewith with the biological sample under conditions wherein the capture agents selectively bind to a first biomarker, if present, on the vesicles, thereby capturing said vesicles having said first biomarker on the solid support; c) separating the solid support from the biological sample; and d) detecting a second biomarker on the vesicles captured on the solid support using a detection agent that selectively binds to the second biomarker, wherein the vesicles are not lysed or permeabilized.
  • the methods of the invention further comprise detecting a secretory protein on the vesicles captured on the solid support.
  • the secretory protein is selected from the group consisting of cytokines, growth factors, chemokines, and interleukins.
  • the cytokine is selected from the group consisting of IL1b, IL34, IL6, IL8, IL16, IL23A, IL32, IL33, CX3CL1, CCL2, CXCL12, TNFalpha, TNFSF10, IL12B, nociceptin, GnRH, FasL, and TNFSF13.
  • the vesicles are selected from the group consisting of exosomes, microparticles, microvesicles, nanosomes, extracellular vesicles, ectosomes, and apoptotic bodies.
  • the first and second biomarkers are membrane-bound proteins or adsorbed proteins on the vesicles.
  • the first or second biomarker may be an exosome surface marker (e.g., CD81, CD63, CD171), a neuron-specific protein (e.g., synaptosome associated protein 25 (SNAP25), neurogranin (NRGN), tau, phosphorylated tau, ⁇ -42, and synaptophysin), an astrocyte-specific protein (e.g., glial fibrillary acidic protein (GFAP) and excitatory amino acid transporter 1 (EAAT1)), a microglia-specific protein (CD11b), an oligodendrocyte-specific protein (e.g., myelin basic protein (MBP), an oligodendrocyte myelin glycoprotein (OMG), or a chemokine (CX3CL1) or cytokine (IL1b, IL34, FasL, or IL12B).
  • the first or second biomarker is a metabolite.
  • the method further comprises detecting a cytosolic protein (e.g., glyceraldehyde-3-phosphate dehydrogenase (GAPDH), alpha-synuclein (SNCA), tyrosine hydroxygenase (TH), cathepsin D (CTSD), AchE, LAMP1, REST, SYT, SYP, SYNPO, PSD95, SV2A, GYS, HSP70, BACE, SYMPO, NEFL, caspase, ubiquitin, PSEN1, GSK, PLAP, CSH1, PSG1, or FasL) on the vesicles captured on the solid support.
  • a cytosolic protein e.g., glyceraldehyde-3-phosphate dehydrogenase (GAPDH), alpha-synuclein (SNCA), tyrosine hydroxygenase (TH
  • the method further comprises detecting one or more additional biomarkers on the vesicles captured on the solid support.
  • the biomarker is selected from the group consisting of CD81, acetylcholinesterase (AchE), Lysosomal Associated Membrane Protein 1 (LAMP1), CTSD, RE1 Silencing Transcription Factor (REST), synaptotagmin (SYT), synaptophysin (SYP), synaptopodin (SYNPO), postsynaptic density protein 95 (PSD95), synaptic vesicle glycoprotein 2A (SV2A), NGRN, monocyte chemotactic protein-1 (CCL2), IL34, glycogen synthase (GYS), (OR), death receptor 6 (DR6), heat shock protein (HSP), IL12beta, alpha-beta (A ⁇ ), beta-secretase (BACE), dopamine receptors (D1 and D2), serotonin receptors
  • the first biomarker is SNAP25 and the second biomarker is selected from the group consisting of CD81, tau, NEFL, TNFa, and IL8.
  • the first biomarker is EAAT1 and the second biomarker is CD81 or GFAP.
  • the first biomarker is OMG and the second biomarker is CD81 or MBP. In some embodiments, the first biomarker is CD11b and the second biomarker is SYP. In some embodiments, the first biomarker is DAT and the second biomarker is CD81, SNCA, or TH. In some embodiments, the first biomarker is CD11b and the second biomarker is SYP.
  • the vesicles captured on the solid support are selected from the group consisting of neuron-derived exosomes, astrocyte-derived exosomes, oligodendrocyte-derived exosomes, and microglia-derived exosomes.
  • the solid support is a plate, a non-magnetic bead, a magnetic bead, a filter, a slide, a wafer, a rod, a particle, a strand, a disc, a membrane, or a surface of a tube, channel, column, flow cell device, or microfluidic device.
  • the solid support can comprise, for example, glass, quartz, silicon, metal, ceramic, plastic, nylon, polyacrylamide, a hydrogel, or a resin.
  • the solid support comprises more than one type of capture agent associated therewith, for example, at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or more different capture agents that selectively bind to different biomarkers on the vesicles.
  • detection of biomarker on vesicles captured on the solid support comprises using more than one type of detection agent, for example, at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or more different detection agents that selectively bind to different biomarkers on the vesicles.
  • the capture agents comprise antibodies, antibody fragments, antibody mimetics, or aptamers that specifically bind to the first biomarker on the vesicles.
  • the detection agents comprise antibodies, antibody fragments, antibody mimetics, or aptamers that specifically bind to the second biomarker on the vesicles.
  • Capture agents and detection agents may comprise monoclonal antibodies, polyclonal antibodies, chimeric antibodies, nanobodies, recombinant fragments of antibodies, Fab fragments, Fab′ fragments, F(ab′) 2 fragments, F v fragments, or scF v fragments.
  • the detection agent further comprises a detectable label, for example, a fluorescent, chemiluminescent, electrochemiluminescent, bioluminescent, isotopic, or radioactive label.
  • a detectable label for example, a fluorescent, chemiluminescent, electrochemiluminescent, bioluminescent, isotopic, or radioactive label.
  • detecting a biomarker on a vesicle comprises performing an immunoassay, such as an enzyme-linked immunosorbent assay (ELISA), an immunofluorescent assay (IFA), an immune-polymerase chain reaction assay, an electro-chemiluminescence immunoassay (ECLIA), and a radioimmunoassay (RIA).
  • an immunoassay such as an enzyme-linked immunosorbent assay (ELISA), an immunofluorescent assay (IFA), an immune-polymerase chain reaction assay, an electro-chemiluminescence immunoassay (ECLIA), and a radioimmunoassay (RIA).
  • the biological sample comprising vesicles may be from a subject or a cell culture supernatant.
  • the biological sample is selected from the group consisting of whole blood, serum, plasma, urine, interstitial fluid, peritoneal fluid, cervical swab, tears, saliva, buccal swab, skin, brain tissue, and cerebrospinal fluid.
  • a biological sample is obtained from a subject who has been diagnosed or is suspected of having a neurological disorder, such as Alzheimer's disease (AD), vascular disease dementia, frontotemporal dementia (FTD), corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), Lewy body dementia, tangle-predominant senile dementia, Pick's disease (PiD), argyrophilic grain disease, amyotrophic lateral sclerosis (ALS), other motor neuron diseases, Guam parkinsonism-dementia complex, FTDP-17, Lytico-Bodig disease, multiple sclerosis, traumatic brain injury (TBI), stroke, depression, bipolar disease, epilepsy, autism, schizophrenia, brain tumor, white matter disease, brain atrophy, mental retardation, cerebellar ataxia, multiple system atrophy, concussion, subconcussive impacts, or Parkinson's disease.
  • a neurological disorder such as Alzheimer's disease (AD), vascular disease dementia, frontotemporal
  • a biological sample is obtained from a subject who has been diagnosed or is suspected of having cancer, such as breast cancer, pancreatic cancer, lung cancer, colon cancer, colorectal cancer, rectal cancer, kidney cancer, bladder cancer, stomach cancer, prostate cancer, liver cancer, ovarian cancer, brain cancer, vaginal cancer, vulvar cancer, uterine cancer, oral cancer, penic cancer, testicular cancer, esophageal cancer, skin cancer, cancer of the fallopian tubes, head and neck cancer, gastrointestinal stromal cancer, adenocarcinoma, cutaneous or intraocular melanoma, cancer of the anal region, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, cancer of the urethra, cancer of the renal pelvis, cancer of the ureter, cancer of the endometrium, cancer of the cervix, cancer of the pituitary gland, neoplasms of the central
  • a biological sample is obtained from a subject who has been diagnosed or is suspected of having an immunological disorder. In other embodiments, a biological sample is obtained from a subject who has been diagnosed or is suspected of having a placental disease.
  • the invention provides a method of diagnosing and treating a disease or a disorder in a subject, the method comprising: a) obtaining a biological sample comprising exosomes from the subject; b) contacting a solid support comprising capture agents associated therewith with the biological sample under conditions wherein the capture agents selectively bind to a first biomarker on the exosomes, wherein exosomes are captured on the solid support; c) separating the solid support from the biological sample; d) measuring a level of a second biomarker on the exosomes captured on the solid support, wherein the exosomes are not lysed or permeabilized; e) diagnosing the subject with the disease or disorder by comparing the level of the second biomarker to a control level of the second biomarker; and f) treating the subject for the disease or disorder if the subject is diagnosed as having the disease or disorder.
  • the first biomarker is a neuron-specific protein, an astrocyte-specific protein, a microglia-specific protein, or an oligodendrocyte-specific protein.
  • the second biomarker is a cytosolic protein, a secretory protein, a receptor protein, a transporter protein, or a membrane protein.
  • the cytosolic protein is selected from the group consisting of GAPDH, CTSD, NRGN, MBP, GFAP, Tau, phosphorylated Tau (e.g., T181), synaptophysin, SNCA, ⁇ -42, AchE, LAMP1, REST, SYT, TH, SYP, SYNPO, PSD95, SV2A, GYS, HSP70, BACE, NEFL, caspase, ubiquitin, PSEN1, GSK, PLAP, CSH1, PSG1, or FasL.
  • the secretory protein is selected from the group consisting of cytokines, growth factors, chemokines, and interleukins.
  • the cytokine is selected from the group consisting of IL1b, IL34, IL6, IL8, IL16, IL23A, IL32, IL33, CX3CL1, CCL2, CXCL12, TNFalpha, TNFSF10, IL12B, nociceptin, GnRH, FasL and TNFSF13.
  • the neurotransmitter receptor is selected from the group consisting of dopamine receptors (D1 and D2), serotonin receptors (2A, 2C, and 3B), GABA receptors (1-6, 5.
  • the membrane protein is selected from the group consisting of EpCAM, PD-L1, ErbB2, CK19, TCR, CD16, CD28, CD32, CD79a, TREM2, and NCAM.
  • the transporter protein is a neurotransmitter transporter, a dopamine transporter (e.g., DAT), a serotonin transporter, a GABA transporter, a glutamate transporter, an insulin transporter, a tumor necrosis factor transporter, or a neuropeptide transporter.
  • the disease or disorder is a neurological disorder, an immunological disorder, a placental disease, or cancer.
  • the invention provides a method of diagnosing and treating a neurological disorder in a subject, the method comprising: a) obtaining a biological sample comprising exosomes from the subject; b) contacting a solid support comprising capture agents associated therewith with the biological sample under conditions wherein the capture agents selectively bind to a first biomarker on the exosomes selected from the group consisting of a neuron-specific protein, an astrocyte-specific protein, a microglia-specific protein, and an oligodendrocyte-specific protein, wherein neuron-derived exosomes, astrocyte-derived exosomes, oligodendrocyte-derived exosomes, or microglia-derived exosomes are captured on the solid support; c) separating the solid support from the biological sample; d) measuring a level of a second biomarker selected from the group consisting of a neuron-specific protein, an astrocyte-specific protein, a microglia-specific protein on the exosomes captured on
  • the present invention provides a method of prognosing or monitoring the progression of a disease or disorder in a subject, the method comprising: a) obtaining a biological sample comprising exosomes from the subject; b) contacting a solid support comprising capture agents associated therewith with the biological sample under conditions wherein the capture agents selectively bind to a first biomarker on the exosomes, wherein exosomes are captured on the solid support; c) separating the solid support from the biological sample; d) measuring a level of a second biomarker protein on the exosomes captured on the solid support, wherein the exosomes are not lysed or permeabilized, thereby prognosing or monitoring the progression of the disease or disorder in the subject.
  • the method further comprises obtaining a second biological sample comprising exosomes from the subject and measuring the level of a biomarker of the present invention and comparing the level of the biomarker in the second biological samples to the level of the biomarker in the first biological sample.
  • the first biomarker is a neuron-specific protein, an astrocyte-specific protein, a microglia-specific protein, or an oligodendrocyte-specific protein.
  • the second biomarker is a cytosolic protein, a secretory protein, a receptor protein, a transporter protein, or a membrane protein.
  • the cytosolic protein is selected from the group consisting of GAPDH, CTSD, NRGN, MBP, GFAP, Tau, phosphorylated Tau (e.g., T181), synaptophysin, SNCA, ⁇ -42, AchE, LAMP1, REST, SYT, TH, SYP, SYNPO, PSD-95, SV2A, GYS, HSP70, BACE, SYMPO, NEFL, caspase, ubiquitin, PSEN1, GSK, PLAP, CSH1, PSG1, or FasL.
  • the secretory protein is selected from the group consisting of cytokines, growth factors, chemokines, and interleukins.
  • the cytokine is selected from the group consisting of IL1b, IL34, IL6, IL8, IL16, IL23A, IL32, IL33, CX3CL1, CCL2, CXCL12, TNFalpha, TNFSF10, IL12B, nociceptin, GnRH, FasL and TNFSF13.
  • the neurotransmitter receptor is selected from the group consisting of dopamine receptors (D1 and D2), serotonin receptors (2A, 2C, and 3B), GABA receptors (1-6, 5.
  • the membrane protein is selected from the group consisting of EpCAM, PD-L1, ErbB2, CK19, TCR, CD16, CD28, CD32, CD79a, TREM2, and NCAM.
  • the transporter protein is a neurotransmitter transporter, a dopamine transporter, a serotonin transporter, a GABA transporter, a glutamate transporter, an insulin transporter, a tumor necrosis factor transporter, or a neuropeptide transporter.
  • the disease or disorder is a neurological disorder, an immunological disorder, a placental disease, or cancer.
  • the present invention provides a method of prognosing or monitoring the progression of a neurological disorder in a subject, the method comprising: a) obtaining a biological sample comprising exosomes from the subject; b) contacting a solid support comprising capture agents associated therewith with the biological sample under conditions wherein the capture agents selectively bind to a first biomarker on the exosomes selected from the group consisting of a neuron-specific protein, an astrocyte-specific protein, a microglia-specific protein, and an oligodendrocyte-specific protein, wherein neuron-derived exosomes, astrocyte-derived exosomes, oligodendrocyte-derived exosomes, or microglia-derived exosomes are captured on the solid support; c) separating the solid support from the biological sample; d) measuring a level of a second biomarker selected from the group consisting of a neuron-specific protein, an astrocyte-specific protein, a microglia-specific protein on
  • the method further comprises obtaining a second biological sample comprising exosomes from the subject and measuring the level of a biomarker of the present invention and comparing the level of the biomarker in the second biological samples to the level of the biomarker in the first biological sample.
  • the invention provides a method of diagnosing and treating a disease or disorder in a subject, the method comprising: a) obtaining a biological sample comprising exosomes from the subject; b) contacting a solid support comprising capture agents associated therewith with the biological sample under conditions wherein the capture agents selectively bind to a first biomarker on the exosomes, wherein exosomes are captured on the solid support; c) separating the solid support from the biological sample; d) detecting a second biomarker on the exosomes captured on the solid support, wherein the exosomes are not lysed or permeabilized; e) diagnosing the subject with the disease or disorder by comparing the level of the second biomarker to a control level of the second biomarker; and f) treating the subject for the disease or disorder if the subject is diagnosed as having the disease or disorder.
  • the first biomarker is a neuron-specific protein, an astrocyte-specific protein, a microglia-specific protein, or an oligodendrocyte-specific protein.
  • the second biomarker is a cytosolic protein, a secretory protein, a receptor protein, a transporter protein, or a membrane protein.
  • the cytosolic protein is selected from the group consisting of GAPDH, CTSD, NRGN, MBP, GFAP, Tau, phosphorylated Tau (e.g., T181), synaptophysin, SNCA, ⁇ -42, AchE, LAMP1, REST, SYT, TH, SYP, SYNPO, PSD95, SV2A, GYS, HSP70, BACE, SYMPO, NEFL, caspase, ubiquitin, PSEN1, GSK, PLAP, CSH1, PSG1, or FasL.
  • the secretory protein is selected from the group consisting of cytokines, growth factors, chemokines, and interleukins.
  • the cytokine is selected from the group consisting of IL1b, IL34, IL6, IL8, IL16, IL23A, IL32, IL33, CX3CL1, CCL2, CXCL12, TNFalpha, TNFSF10, IL12B, nociceptin, GnRH, FasL and TNFSF13.
  • the neurotransmitter receptor is selected from the group consisting of dopamine receptors (D1 and D2), serotonin receptors (2A, 2C, and 3B), GABA receptors (1-6, 5.
  • the membrane protein is selected from the group consisting of EpCAM, PD-L1, ErbB2, CK19, TCR, CD16, CD28, CD32, CD79a, TREM2, and NCAM.
  • the transporter protein is a neurotransmitter transporter, a dopamine transporter, a serotonin transporter, a GABA transporter, a glutamate transporter, an insulin transporter, a tumor necrosis factor transporter, or a neuropeptide transporter.
  • the disease or disorder is a neurological disorder, an immunological disorder, a placental disease, or cancer.
  • the invention provides a method of diagnosing and treating a neurological disorder in a subject, the method comprising: a) obtaining a biological sample comprising exosomes from the subject; b) contacting a solid support comprising capture agents associated therewith with the biological sample under conditions wherein the capture agents selectively bind to a first biomarker on the exosomes selected from the group consisting of a neuron-specific protein, an astrocyte-specific protein, a microglia-specific protein, and an oligodendrocyte-specific protein, wherein neuron-derived exosomes, astrocyte-derived exosomes, oligodendrocyte-derived exosomes, or microglia-derived exosomes are captured on the solid support; c) separating the solid support from the biological sample; d) detecting a second biomarker selected from the group consisting of a neuron-specific protein, an astrocyte-specific protein, a microglia-specific protein on the exosomes captured on the solid
  • the neurological disorder is selected from the group consisting of: Alzheimer's disease (AD), vascular disease dementia, frontotemporal dementia (FTD), corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), Lewy body dementia, tangle-predominant senile dementia, Pick's disease (PiD), argyrophilic grain disease, amyotrophic lateral sclerosis (ALS), other motor neuron diseases, Guam parkinsonism-dementia complex, FTDP-17, Lytico-Bodig disease, multiple sclerosis, traumatic brain injury (TBI), stroke, depression, bipolar disease, epilepsy, autism, schizophrenia, brain tumor, white matter disease, brain atrophy, mental retardation, cerebellar ataxia, multiple system atrophy, concussion, subconcussive impacts, and Parkinson's disease.
  • AD Alzheimer's disease
  • FDD frontotemporal dementia
  • CBD corticobasal degeneration
  • PSP progressive supranuclear pal
  • the first and second biomarkers are membrane-bound proteins or adsorbed proteins on the vesicles, including but not limited to, a neuron-specific protein selected from the group consisting of synaptosome associated protein 25 (SNAP25), neurogranin (NRGN), tau, phosphorylated tau, and synaptophysin, an astrocyte-specific proteins selected from the group consisting of glial fibrillary acidic protein (GFAP) and excitatory amino acid transporter 1 (EAAT1), a microglia-specific protein (CD11b), a cytokine selected from the group consisting of IL1b, IL34, FasL, or IL12B, a chemokine (CX3CL1), CD81, CD171, CTSD, CD63, 4-42, an oligodendrocyte-specific protein selected from the group consisting of myelin basic protein (MBP) and oligodendrocyte myelin glycoprotein (OMG).
  • a neuron-specific protein selected from the group
  • the biomarker is selected from the group consisting of CD81, acetylcholinesterase (AchE), Lysosomal Associated Membrane Protein 1 (LAMP1), CTSD, RE1 Silencing Transcription Factor (REST), synaptotagmin (SYT), NGRN, monocyte chemotactic protein-1 (CCL2), IL34, glycogen synthase (GYS), olfactory receptor (OR), death receptor 6 (DR6), heat shock protein (HSP), IL12beta, alpha-beta (A ⁇ ), beta-secretase (BACE), dopamine receptors (D1 and D2), serotonin receptors (2A, 2C, and 3B), GABA receptors (1-6, 5, B1, B2), glutamate receptors (1 and 2), insulin receptors, tumor necrosis factor receptors superfamily (TRAL, TNF receptor, death receptor 5 and 6), neuropeptide receptors (orexin receptor, opioid receptor KOR), Ep
  • the first biomarker is SNAP25 and the second biomarker is selected from the group consisting of CD81, tau, NEFL, TNFa, and IL8.
  • the first biomarker is EAAT1 and the second biomarker is CD81 or GFAP.
  • the first biomarker is OMG and the second biomarker is CD81 or MBP.
  • the first biomarker is CD11b and the second biomarker is SYP.
  • the level of one or more biomarkers on exosomes in the biological sample is compared to the level of one or more biomarkers in a control sample, wherein the level of the one or more biomarkers of the biological sample is elevated compared to the control sample. In some embodiments, the level of the one or more biomarkers in the biological sample is compared to the level of one or more biomarkers in a control sample, wherein the level of the one or more biomarkers of the biological sample is decreased compared to the control sample. In some embodiments, the biomarker level determines the disease or disorder status of the subject with at least 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% specificity.
  • the disease or disorder is a neurological disorder, an immunological disorder, a placental disease, or cancer.
  • the neurological disorder is selected from the group consisting of: Alzheimer's disease (AD), vascular disease dementia, frontotemporal dementia (FTD), corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), Lewy body dementia, tangle-predominant senile dementia, Pick's disease (PiD), argyrophilic grain disease, amyotrophic lateral sclerosis (ALS), other motor neuron diseases, Guam parkinsonism-dementia complex, FTDP-17, Lytico-Bodig disease, multiple sclerosis, traumatic brain injury (TBI), stroke, depression, bipolar disease, epilepsy, autism, schizophrenia, brain tumor, white matter disease, brain atrophy, mental retardation, cerebellar ataxia, multiple system atrophy, concussion, subconcussive impacts, and Parkinson's disease.
  • AD Alzheimer's disease
  • FDD
  • the biological sample is selected from the group consisting of whole blood, serum, plasma, urine, interstitial fluid, peritoneal fluid, cervical swab, tears, saliva, buccal swab, skin, brain tissue, and cerebrospinal fluid.
  • the cancer is breast cancer, pancreatic cancer, lung cancer, colon cancer, colorectal cancer, rectal cancer, kidney cancer, bladder cancer, stomach cancer, prostate cancer, liver cancer, ovarian cancer, brain cancer, vaginal cancer, vulvar cancer, uterine cancer, oral cancer, penic cancer, testicular cancer, esophageal cancer, skin cancer, cancer of the fallopian tubes, head and neck cancer, gastrointestinal stromal cancer, adenocarcinoma, cutaneous or intraocular melanoma, cancer of the anal region, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, cancer of the urethra, cancer of the renal pelvis, cancer of the ureter, cancer of the endometrium, cancer of the cervix, cancer of the pituitary gland, neoplasms of the central nervous system (CNS), primary CNS lymphoma, brain stem gliom
  • CNS
  • the invention includes a method of diagnosing and treating a disease or disorder in a subject, the method comprising: a) obtaining a biological sample comprising exosomes from the subject; b) contacting a solid support comprising capture agents associated therewith with the biological sample under conditions wherein the capture agents selectively bind to a membrane marker on the exosomes wherein the membrane marker is selected from the group consisting of SNAP25, OMG, CD11b, EAAT1 and CD171, wherein exosomes having the membrane marker are captured on the solid support; c) separating the solid support from the biological sample; d) measuring levels of one or more biomarkers selected from the group consisting of CD81, GAPDH, CTSD, NRGN, MBP, GFAP, Tau, phosphorylated Tau, CD63, ⁇ -42, CX3CL1, IL1b, and IL34 on the exosomes captured on the solid support, wherein the exosomes are not lysed or permeabilized; e
  • the biomarker is selected from the group consisting of CD81, acetylcholinesterase (AchE), Lysosomal Associated Membrane Protein 1 (LAMP1), CTSD, RE1 Silencing Transcription Factor (REST), synaptotagmin (SYT), NGRN, monocyte chemotactic protein-1 (CCL2), IL34, glycogen synthase (GYS), olfactory receptor (OR), death receptor 6 (DR6), heat shock protein (HSP), IL12beta, alpha-beta (A ⁇ ), beta-secretase (BACE), dopamine receptors (D1 and D2), serotonin receptors (2A, 2C, and 3B), GABA receptors (1-6, 5.
  • the disease or disorder is a neurological disorder, an immunological disorder, a placental disease, or cancer.
  • the invention includes a kit for detecting biomarkers on vesicles, the kit comprising: a) a solid support comprising capture agents associated therewith, wherein the capture agents selectively bind to a first biomarker on the surface of the vesicles; and b) at least one detection agent that selectively binds to a second biomarker on the surface of the vesicles.
  • the vesicles are selected from the group consisting of exosomes, microparticles, microvesicles, nanosomes, extracellular vesicles, and ectosomes.
  • the invention provides a kit for detecting biomarkers on vesicles, the kit comprising: a) a solid support comprising capture agents associated therewith, wherein at least one capture agent selectively binds to a neuron-specific protein, an astrocyte-specific protein, a microglia-specific protein, or an oligodendrocyte-specific protein on exosomes; and b) one or more detection agents, wherein the one or more detection agents selectively binds to cytosolic proteins, secretory proteins, neurotransmitter receptors or membrane proteins on the surface of the exosomes.
  • the cytosolic proteins are selected from the group consisting of GAPDH, CTSD, NRGN, MBP, GFAP, Tau, phosphorylated Tau (e.g., T181), synaptophysin, SNCA, ⁇ -42, AchE, LAMP1, REST, SYT, TH, SYP, SYNPO, PSD95, SV2A, GYS, HSP70, BACE, SYMPO, NEFL, caspase, ubiquitin, PSEN1, GSK, PLAP, CSH1, PSG1, or FasL.
  • the secretory protein is selected from the group consisting of cytokines, growth factors, chemokines, and interleukins.
  • the cytokine is selected from the group consisting of IL1b, IL34, IL6, IL8, IL16, IL23A, IL32, IL33, CX3CL1, CCL2, CXCL12, TNFalpha, TNFSF10, IL12B, nociceptin, GnRH, FasL and TNFSF13.
  • the neurotransmitter receptor is selected from the group consisting of dopamine receptors (D1 and D2), serotonin receptors (2A, 2C, and 3B), GABA receptors (1-6, 5.
  • the membrane protein is selected from the group consisting of EpCAM, PD-L1, ErbB2, CK19, TCR, CD16, CD28, CD32, CD79a, TREM2, and NCAM.
  • the capture agents and detection agents in the kit comprise antibodies, antibody fragments, antibody mimetics, or aptamers that specifically bind to the first biomarker on the vesicles.
  • the antibodies may include, for example, monoclonal antibodies, polyclonal antibodies, chimeric antibodies, nanobodies, recombinant fragments of antibodies, Fab fragments, Fab′ fragments, F(ab′) 2 fragments, F v fragments, and an scF v fragments.
  • the capture agents or detection agents comprise antibodies selected from the group consisting of an anti-CD171 antibody, an anti-synaptosome associated protein 25 (SNAP25) antibody, an anti-neurogranin (NRGN) antibody, an anti-tau antibody, an anti-synaptophysin antibody, and anti-CD63 antibody, an anti- ⁇ -42 antibody, an anti-CD81 antibody, an anti-CTD antibody, an anti-GAPDH antibody, an anti-IL1b antibody, an anti-IL34 antibody, an anti-CX3CL1 antibody, an anti-glial fibrillary acidic protein (GFAP) antibody, an anti-excitatory amino acid transporter 1 (EAAT1) antibody, an anti-myelin basic protein (MBP) antibody, an anti-SNCA antibody, an anti-TH antibody, an anti-CD11b antibody, and an anti-oligodendrocyte myelin glycoprotein (OMG) antibody.
  • an anti-CD171 antibody an anti-synaptosome associated protein 25 (SNAP
  • the at least one capture agent selectively binds to CD171, CD63, CD81, SNAP25, EAAT1, CD11b, or OMG on exosomes.
  • the one or more detection agents selectively binds to CD81, GAPDH, CTSD, NRGN, MBP, GFAP, Tau, phosphorylated Tau (e.g., T181), CD63, ⁇ -42, CX3CL1, IL1b, or IL34 on the surface of the exosomes.
  • the at least one or more detection agents selectively binds to CD81, acetylcholinesterase (AchE), Lysosomal Associated Membrane Protein 1 (LAMP1), CTSD, RE1 Silencing Transcription Factor (REST), synaptotagmin (SYT), NGRN, monocyte chemotactic protein-1 (CCL2), IL34, glycogen synthase (GYS), olfactory receptor (OR), death receptor 6 (DR6), heat shock protein (HSP), IL12beta, alpha-beta (A ⁇ ), beta-secretase (BACE), dopamine receptors (D1 and D2), serotonin receptors (2A, 2C, and 3B), GABA receptors (1-6, 5.
  • AchE acetylcholinesterase
  • LAMP1 Lysosomal Associated Membrane Protein 1
  • CTSD CTSD
  • REST RE1 Silencing Transcription Factor
  • SYT synaptotag
  • B1, B2 glutamate receptors (1 and 2), insulin receptors, tumor necrosis factor receptors superfamily (TRAL, TNF receptor, death receptor 5 and 6), neuropeptide receptors (orexin receptor, opioid receptor KOR), EpCAM, PD-L1, ErbB2, CK19, TCR, CD16, CD28, CD32, CD79a, TREM2, or NCAM.
  • the detection agent in the kit further comprises a detectable label, for example, a fluorescent, chemiluminescent, electrochemiluminescent, bioluminescent, isotopic, or radioactive label.
  • a detectable label for example, a fluorescent, chemiluminescent, electrochemiluminescent, bioluminescent, isotopic, or radioactive label.
  • the kit further comprises reagents for performing an immunoassay.
  • immunoassays include an enzyme-linked immunosorbent assay (ELISA), an immunofluorescent assay (IFA), an immune-polymerase chain reaction assay, an electro-chemiluminescence immunoassay (ECLIA), and a radioimmunoassay (RIA).
  • the invention includes a kit for diagnosing or prognosing a disease or disorder in a subject, identifying a subject at risk of a disease or disorder, or prescribing a therapeutic regimen or predicting benefit from therapy in a subject having a disease or disorder, the kit comprising: a) a solid support comprising capture agents associated therewith, wherein at least one capture agent selectively binds to a CD171 membrane marker on exosomes; and b) at least two detection agents, wherein at least one detection agent selectively binds to phosphorylated tau T181 and at least one detection agent selectively binds to neurogranin on the surface of the exosomes.
  • At least one capture agent or detection agent comprises an antibody, an antibody fragment, an antibody mimetic, or an aptamer that specifically binds to CD171, phosphorylated tau T181, or neurogranin.
  • the antibody is selected from the group consisting of a monoclonal antibody, a polyclonal antibody, a chimeric antibody, a nanobody, a recombinant fragment of an antibody, an Fab fragment, an Fab′ fragment, an F(ab′) 2 fragment, an F v fragment, and an scF v fragment.
  • the kit comprises an anti-neurogranin antibody, an anti-phosphorylated tau T181 antibody, and an anti-CD171 antibody.
  • the disease or disorder is a neurological disorder, an immunological disorder, a placental disease, or cancer.
  • the invention provides a method of prognosing or monitoring the progression of a disease or disorder in a subject, the method comprising: a) obtaining a biological sample comprising vesicles or exosomes from the subject; b) contacting a solid support comprising capture agents associated therewith with the biological sample under conditions wherein the capture agents selectively bind to a first biomarker on the vesicles or exosomes, wherein vesicles or exosomes are captured on the solid support; c) separating the solid support from the biological sample; d) measuring a level of a second biomarker protein on the vesicles or exosomes captured on the solid support, wherein the vesicles or exosomes are not lysed or permeabilized, thereby prognosing or monitoring the progression of the disease or disorder in the subject.
  • the method further comprises repeating (a), (b), (c), and (d) at a second time point with a second biological sample wherein an increase in the levels of the second biomarker relative to its level at the first time point indicates that the disease or disorder is progressing, and a decrease in the level of the second biomarker relative to its level at the first time point indicates that the disease or disorder is regressing.
  • the methods further comprise treating the subject for the disease or disorder if the disease or disorder is progressing.
  • the method further comprises reducing treatment or stopping treatment for the disease or disorder if the disease or disorder is regressing.
  • the disease or disorder is a neurological disorder, an immunological disorder, a placental disease, or cancer.
  • the neurological disorder is selected from the group consisting of: Alzheimer's disease (AD), vascular disease dementia, frontotemporal dementia (FTD), corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), Lewy body dementia, tangle-predominant senile dementia, Pick's disease (PiD), argyrophilic grain disease, amyotrophic lateral sclerosis (ALS), other motor neuron diseases, Guam parkinsonism-dementia complex, FTDP-17, Lytico-Bodig disease, multiple sclerosis, traumatic brain injury (TBI), stroke, depression, bipolar disease, epilepsy, autism, schizophrenia, brain tumor, white matter disease, brain atrophy, mental retardation, cerebellar ataxia, multiple system atrophy, concussion, subconcussive impacts, and Parkinson's disease.
  • AD Alzheimer's disease
  • FDD frontotemporal dementia
  • CBD corticobasal degeneration
  • PSP progressive supranuclear pal
  • FIGS. 1A and 1B shows methods for quantifying cell-specific biomarkers of exosomes.
  • FIG. 1A shows a schematic depicting the generation of exosomes containing proteins of the cell from which they originated (mother cell).
  • FIG. 1B shows methods of screening exosomes for membrane markers and adsorbed markers.
  • FIGS. 2A-2F show exemplary detection of cytosolic (non-membrane) proteins on the surface of exosomes.
  • Neuron-specific anti-SNAP25 FIGS. 2A-2C
  • control mouse IgG FIGS. 2D-2F
  • Various volumes (10, 2.5, and 0 uL) of plasma was suspended in PBS ( ⁇ ), 0.1% tween-20 PBS (o), or 0.1% triton-X100 PBS (I) in a final volume of 40 uL, and applied to the ELISA plates. After overnight incubation at 4° C., unbound materials were removed.
  • FIGS. 3A-3D show the specificity of marker proteins of the present invention.
  • Antibodies against ( FIG. 3A ) the exosome surface marker CD81, ( FIG. 3B ) neuron surface marker SNAP25, ( FIG. 3C ) astrocyte surface marker (EAAT1), or ( FIG. 3D ) oligodendrocyte surface marker (OMG) were immobilized onto white ELISA plates. Ten ⁇ L of plasma suspended in 40 ⁇ L of PBS were applied to the ELISA plates. After overnight incubation at 4° C., unbound materials were removed.
  • Biotin-labeled antibodies against the exosome surface marker CD81, general cytosolic marker GAPDH, cytosolic proteins in neuron (NRGN), oligodendrocyte (MBP), and astrocyte (GFAP) were applied to the ELISA plates.
  • a no antibody control (tPBS) was also included.
  • Conventional chemiluminescent ELISA was carried out, and the relative light unit (RLU) was determined.
  • FIGS. 4A-4D show detection and quantification of exosome biomarkers of the present invention.
  • Anti-SNAP25 FIG. 4A and FIG. 4B
  • anti-EAAT1 FIG. 4C
  • anti-OMG FIG. 4D
  • Various volume of standard plasma was suspended in PBS, and applied to ELISA wells in a final volume of 40 mL.
  • Antibodies against NRGN FIG. 4A
  • tau FIG. 4B
  • GFAP FIG. 4C
  • MBP FIG. 4D
  • FIGS. 5A-5C show screening of exosome adsorbed markers.
  • Control mouse IgG open columns
  • mouse anti-human CD81 light gray columns
  • mouse anti-human synaptosomal-associated protein 25 SNAP25
  • Detection antibodies were against not only exosome membrane proteins (CD81, CD171, CD63, and SNAP25), but also various non-membrane proteins (glyceraldehyde 3-phosphate dehydrogenase (GAPDH), cathepsin D (CTSD), neurogranin (NRGN), myelin basic protein (MBP), glial fibrillary acidic protein (GFAP), tau, microtubule associated protein tau (tau) and phosphorylated tau T181, and amyloid b 1-42 peptide (ABETA42).
  • GPDH glycoprotein dehydrogenase
  • CSD cathepsin D
  • NRGN neurogranin
  • MBP myelin basic protein
  • GFAP glial fibrillary acidic protein
  • tau microtubule associated protein tau
  • ABETA42 amyloid b 1-42 peptide
  • FIG. 5A shows the detection of the membrane proteins, CD81 and CD171, and the non-membrane proteins, GAPDH and CTSD.
  • FIG. 5B shows the detection of the membrane proteins, CD63, and SNAP25.
  • FIG. 5C shows the detection of the non-membrane proteins, NRGN, MBP, GFAP, tau, T181, and ABETA42.
  • FIG. 6A-6E shows clinical applications for diagnosis of Alzheimer's disease.
  • Anti-CD171 antibodies were immobilized on an ELISA plate as shown in the assay schematic in FIG. 6A .
  • Twelve EDTA plasma samples from subjects with Alzheimer's disease and an age, gender-matched control were applied to ELISA wells, and antibodies against ( FIG. 6B ) CD81, ( FIG. 6C ) CTSD.
  • FIG. 6D NRGN
  • FIG. 6E p-tau T181 were used as detection antibodies.
  • FIGS. 7A-7L show the stability of plasma levels of various exosome biomarkers of the present invention.
  • Anti-CD81, anti-CD171, anti-SNAP25, anti-EAAT1, and anti-OMG were immobilized to ELISA plate.
  • EDTA plasma was obtained from 7 control subjects every week for 2-3 weeks, and applied to ELISA wells.
  • FIG. 7A shows assay schematic for detecting exosome membrane targets.
  • FIGS. 7B-7F show detection with ( FIG. 7B ) the anti-CD81 probe on the CD81 plate for total exosome (TE), ( FIG. 7C ) the anti-CD171 probe on the CD81 plate for CD171-based NDE (cNDE), ( FIG.
  • FIG. 7D shows the anti-SNAP25 probe on the CD81 plate for SNAP25-based NDE (sNDE), ( FIG. 7E ) the anti-CD81 probe on the EAAT1 plate (ADE), and ( FIG. 7F ) the anti-CD81 probe on the OMG plate for ODE.
  • FIG. 7G shows assay schematic for exosome surface protein targets.
  • FIGS. 7H-7L show detection with ( FIG. 7H ) the GFAP probe on the EAAT1 plate, ( FIG. 7I ) the MBP probe on the OMG plate, ( FIG. 7J ) the NRGN probe on the SNAP25 plate, ( FIG. 7K ) the tau probe on the SNAP25 plate, and ( FIG. 7L ) the NRGN probe on the CD171 plate, respectively.
  • FIGS. 8A and 8B show detection and quantification of exosome biomarkers of the present invention in plasma using an anti-NRGN-immobilized ELISA plate.
  • FIG. 8A shows an exemplary assay schematic.
  • FIG. 8B shows the assay results.
  • Anti-NRGN medium gray and dark gray columns
  • control mouse IgG black and light gray columns
  • EDTA plasma light gray and dark gray columns
  • PBS alone black and medium gray columns
  • FIGS. 9A-9D show microglia targeting of exosome biomarkers of the present invention.
  • FIG. 9A shows an assay schematic.
  • FIGS. 9B-9D show the assay results.
  • Anti-CD11b medium gray and dark gray columns
  • control mouse IgG black and light gray columns
  • EDTA plasma from 7 different donors (IR1-IR7) (light gray and dark gray columns) or PBS alone (black and medium gray columns) were applied to ELISA wells.
  • Antibodies against GFAP FIG. 9B
  • MBP FIG. 9C
  • NRGN FIG. 9D
  • FIGS. 10A-10F set forth data showing detection of secretory proteins (cytokines (IL1b and IL34) and chemokines (CX3CL1)) on the surface of brain-derived exosomes.
  • cytokines IL1b and IL34
  • CX3CL1 chemokines
  • FIG. 11 sets forth data showing dilution of standard plasma as an ELISA quantification standard.
  • FIG. 12 sets forth data showing duplicate variation for plasma levels of 16 exosomal biomarkers of the present invention.
  • FIG. 13 sets forth data showing standard curves for plasma levels of 16 exosomal biomarkers of the present invention.
  • FIG. 14 sets forth data showing quantification of target exosomal surface protein biomarkers in plasma.
  • FIG. 15 sets forth data showing plasma exosomal biomarker levels normalized to CD81.
  • FIG. 16 sets forth data showing plasma exosomal biomarker levels normalized to SYT.
  • FIG. 17 sets forth data showing the principle of an exemplary assay of the present invention.
  • NDE, ADE, and ODE are specifically captured on ELISA plates, where anti-SNAP25 (neuron marker), anti-EAAT1 (astrocyte marker), and anti-OMG (oligodedrocyte marker) are immobilized. Captured exosomes were further probed with biotinylated anti-CD81 (exosome common marker).
  • FIGS. 18A-18C set forth data showing the specificity of an exemplary assay of the present invention.
  • Anti-SNAP25 500 ng/mL
  • anti-EAAT1 830 ng/mL
  • anti-OMG 830 ng/mL
  • control mouse IgG 500 ng/mL
  • FIGS. 19A-19C set forth data showing intra-assay precision of an exemplary assay of the present invention.
  • NDE (A), ADE (B), and ODE (C) were enumerated in 52 samples (15 each of control, PD, MSA, and PSP) in duplicate, and each replicate was analyzed in x-y plot to show the intra-assay variation. Dotted lines are 45° line and solid lines are regression lines.
  • FIGS. 20A-20C set forth data showing inter-assay precision for an exemplary assay of the present invention.
  • NDE (A), ADE (B), and ODE (C) were enumerated in 3 different control plasma samples 9 separate times.
  • 7 dilutions of standard plasma (arbitrary assigned 100 units/mL) and buffer control were analyzed simultaneously, then ELISA readings (RLU) were converted to units/mL.
  • Average ⁇ standard deviation (CV %) of control plasma 1 ( ⁇ ), 2 ( ⁇ ), and 3 (•) were 64.6 ⁇ 9.2 units/mL (14.2%), 1.6 ⁇ 0.8 units/mL (52.9%), and 44.4 ⁇ 5.5 units/mL (12.4%) for NDE, 7.5 ⁇ 1.3 units/mL (16.8%), 2.6 ⁇ 0.5 units/mL (20.3%), and 0.2 ⁇ 0.1 units/mL (63.33%) for ADE, and 12.6 ⁇ 1.3 units/mL (10.1%), 2.6 ⁇ 0.3 units/mL (11.45%), and 0.2 ⁇ 0.1 units/mL (34.5%) for ODE, respectively.
  • FIGS. 21A-21C set forth data showing dilutional linearity for an exemplary assay of the present invention. After screening of various control plasma samples, we found 3 plasma samples ( ⁇ , •, ⁇ ), showing a wide range of NDE (A), ADE (B), and ODE (C). Then, plasma dilution study was carried out to analyze the linearity and the slope among 3 plasma samples.
  • FIGS. 22A-22C set forth data showing enumeration of brain-derived exosomes in human subjects with Parkinson's Disease.
  • NDE (A), ADE (B), and ODE (C) were enumerated in 15 each of control, PD, MSA, and 7 PSP patients. Each dot represents a single individual, and p values were against controls.
  • FIGS. 23A-23C set forth data showing Receiver Operating Characteristic (ROC) of NDE and ODE for the differential diagnosis of Parkinson's Disease and controls.
  • ROC Receiver Operating Characteristic
  • A NDE
  • B ODE
  • AUC Area under curve
  • FIGS. 24A-24F set forth data showing the correlation between the levels of plasma BDE and disease severity in plasma samples from subjects with Parkinson's Disease.
  • NDE A, D
  • ADE B, E
  • ODE C, F
  • mRS mRS
  • Hoehn-Yahr D-F
  • Each dot represents a single individual, and statistical p-values were between each severity group and control.
  • FIGS. 25A-25D set forth data showing correlation of Brain-Derived Exosomes and Parkinson's Disease severity.
  • A ADE/NDE ratio and disease duration.
  • B ADE/NDE ratio and mRS score.
  • C ADE/NDE ratio and UPDRS part 3 (motor).
  • D ODE/NDE ratio and UPDRS part 3 (motor).
  • FIGS. 26A-26D set forth data showing correlation between the levels of plasma BDE and disease severity in MSA.
  • A ODE (A) of MSA patients was classified with the severity scores of mRS and shown with those of controls. Each dot represents a single individual.
  • B Same data as shown in FIG. 3C .
  • C-E ODE of each MSA patient was compared with disease duration in months (C), and scores of International Cooperative Ataxia Rating Scale (ICARS) (D) and Unified Parkinson's Disease Rating Scale (UPDRS) 3 (motor) (E). The r 2 and p values were shown in E.
  • ICARS International Cooperative Ataxia Rating Scale
  • UPDRS Unified Parkinson's Disease Rating Scale
  • FIGS. 27A-27F set forth data showing detection of pathological form of ⁇ -synuclein on the surface of neuron-, astrocyte-, and oligodendrocyte-derived exosomes.
  • FIGS. 28A-28B set forth data showing detection of dopamine receptor D2 (DRD2) on the surface of neuron-derived exosomes (NDE).
  • D2 dopamine receptor D2
  • NDE neuron-derived exosomes
  • FIGS. 29A-29C set forth data showing plasma dilution study results for DRD2.
  • FIGS. 30A-30G set forth data showing DRD2 detection in neuron-derived exosomes in plasma samples from humans with neurological disease.
  • FIG. 31 sets forth data showing ROC curve and area under the curve (AUC) for DRD2 levels between PD and control samples.
  • FIGS. 32A-32D set forth data showing detection of CD81, SNCA, SNCA oligomer F1, and TH on the surface of DAT+ vesicles.
  • FIG. 33 sets forth data showing detection of SNAP25, EAAT1, and OMG on the surface of vesicles in plasma.
  • FIG. 34 sets forth data showing particle size for captured DAT+CD81+ vesicles in plasma.
  • FIG. 35 sets forth data showing detection of various biomarkers on the surface of vesicles in plasma.
  • the present invention relates, in part, to the development of an efficient method for detecting and quantitating biomarkers on vesicles.
  • vesicles are selectively captured on a solid support, and membrane-bound and adsorbed biomarkers on the captured vesicles are detected without lysis or permeabilization of the vesicles.
  • the invention further relates to the use of such biomarkers identified on vesicles in diagnostic and prognostic methods for various diseases.
  • compositions for use in the methods described herein may include a solid support, capture agents which specifically bind to a surface marker (i.e., membrane protein or adsorbed protein) on vesicles to capture vesicles on the solid support, detection agents that specifically bind to biomarkers on vesicles, reagents for performing immunoassays, and other reagents for performing the methods described herein.
  • a surface marker i.e., membrane protein or adsorbed protein
  • kits for detecting biomarkers on vesicles may comprise a solid support, one or more capture agents which specifically bind and capture vesicles on the solid support, one or more detection agents which specifically bind a biomarker on vesicles, and optionally, immunoassay reagents and other reagents for performing the methods described herein, one or more containers for collecting and or holding the biological sample, and instructions for using the kits.
  • the present invention further relates to the discovery that exosomal biomarkers can be assayed to identify subjects having or likely to develop neurological disorders, including, for example, Alzheimer's disease (AD), multiple sclerosis (MS), and frontotemporal dementia (FTD).
  • AD Alzheimer's disease
  • MS multiple sclerosis
  • FTD frontotemporal dementia
  • the present invention is based, in part, on the discovery of unexpected decreases or increases in certain biomarkers in neuron-derived exosomes present in the circulation of subjects having neurological disease (e.g., Alzheimer's disease).
  • the present invention demonstrates that exosomal levels of these biomarkers may be assayed to diagnose a neurological disorder in a subject having a neurological disease.
  • the present invention further shows that measurement of certain biomarkers in neuron-derived exosomes from a subject may be used to predict the subsequent development of a neurological disease (e.g., identify a subject at risk of developing a neurological disorder).
  • a biological sample comprising vesicles may be obtained from a subject.
  • the biological sample obtained from the subject is typically blood, but can be any sample from bodily fluids, tissue or cells comprising the vesicles to be analyzed.
  • the biological sample may include, but is not limited to, whole blood, serum, plasma, urine, interstitial fluid, peritoneal fluid, cerebrospinal fluid, a cervical swab, tears, saliva, a buccal swab, skin, organs, and biopsies.
  • exosomes can be obtained from cultured cells by collection of secreted exosomes from the surrounding culture media.
  • the biological sample of the invention is obtained from blood.
  • about 1-10 mL of blood is drawn from a subject.
  • about 10-50 mL of blood is drawn from a subject.
  • Blood can be drawn from any suitable area of the body, including an arm, a leg, or blood accessible through a central venous catheter.
  • blood is collected following a treatment or activity.
  • blood can be collected following a medical exam.
  • the timing of collection can also be coordinated to increase the number and/or composition of vesicles (e.g., exosomes) present in the sample.
  • blood can be collected following exercise or a treatment that induces vascular dilation.
  • Blood may be combined with various components following collection to preserve or prepare samples for subsequent techniques.
  • blood is treated with an anticoagulant, a cell fixative, a protease inhibitor, a phosphatase inhibitor, a protein, a DNA, or an RNA preservative following collection.
  • blood is collected via venipuncture using vacuum collection tubes containing an anticoagulant such as EDTA or heparin.
  • an anticoagulant such as EDTA or heparin.
  • Blood can also be collected using a heparin-coated syringe and hypodermic needle.
  • Blood can also be combined with components that will be useful for cell culture.
  • blood is combined with cell culture media or supplemented cell culture media (e.g., cytokines).
  • Vesicles Exosomes, Microparticles, Microvesicles, Nanosomes, Extracellular Vesicles, and Ectosomes
  • Samples can be enriched for vesicles through positive selection, negative selection, or a combination of positive and negative selection.
  • vesicles are directly captured.
  • blood cells are captured and vesicles are collected from the remaining biological samples.
  • the vesicles enriched in the biological samples are exosomes, microparticles, microvesicles, nanosomes, extracellular vesicles, or ectosomes.
  • the vesicles enriched in the biological samples are neuron-derived exosomes, astrocyte-derived exosomes, oligodendrocyte-derived exosomes, or microglia-derived exosomes.
  • Samples can also be enriched for vesicles based on differences in the biochemical properties of vesicles.
  • samples can be enriched for vesicles based on antigen, nucleic acid, metabolic, gene expression, or epigenetic differences.
  • antigen differences antibody-conjugated magnetic or paramagnetic beads in magnetic field gradients or fluorescently labeled antibodies with flow cytometry are used.
  • nucleic acid differences flow cytometry is used.
  • dye uptake/exclusion measured by flow cytometry or another sorting technology is used.
  • cell culture with cytokines is used.
  • Samples can also be enriched for vesicles based on other biochemical properties known in the art. For example, samples can be enriched for vesicles based on pH or motility. Further, in some embodiments, more than one method is used to enrich for vesicles. In other embodiments, samples are enriched for vesicles using antibodies, ligands, or soluble receptors.
  • surface markers are used to positively enrich vesicles in the sample.
  • the vesicles are exosomes, microparticles, microvesicles, nanosomes, extracellular vesicles, or ectosomes.
  • NCAM, CD171, CD9, CD63, CD81, SNAP25, EAAT1, OMG, neuron-specific enolase, diverse neuron or astrocyte adhesive proteins, microglial CD18/11, or CD3 T cell membrane cell surface markers are used to enrich for exosomes.
  • cell surface markers that are not found on vesicles populations are used to negatively enrich vesicles by depleting cell populations.
  • Flow cytometry sorting may also be used to further enrich for exosomes using cell surface markers or intracellular or extracellular markers conjugated to fluorescent labels.
  • Intracellular and extracellular markers may include nuclear stains or antibodies against intracellular or extracellular proteins preferentially expressed in vesicles.
  • Cell surface markers may include antibodies against cell surface antigens that are preferentially expressed on exosomes (e.g., NCAM).
  • the cell surface marker is a neuron-derived exosome surface marker, including, for example, NCAM or CD171.
  • a monoclonal NCAM, CD9, CD63, CD81, neuron-specific enolase or CD171 antibody is used to enrich or isolate exosomes from the sample.
  • the NCAM, CD9, CD63, CD81, neuron-specific enolase or CD171 antibody is biotinylated.
  • biotinylated NCAM or CD171 antibody can form an antibody-exosome complex that can be subsequently isolated using streptavidin-agarose resin or beads.
  • the NCAM, CD9, CD63, CD81, neuron-specific enolase or CD171 antibody is a monoclonal anti-human NCAM, CD9, CD63, CD81, neuron-specific enolase or CD171 antibody.
  • the cell surface marker is a neuron-specific protein (e.g., synaptosome associated protein 25 (SNAP25), neurogranin (NRGN), tau, phosphorylated tau, ⁇ -42, and synaptophysin), an astrocyte-specific protein (e.g., glial fibrillary acidic protein (GFAP) and excitatory amino acid transporter 1 (EAAT1)), a microglia-specific protein (CD11b), an oligodendrocyte-specific protein (e.g., myelin basic protein (MBP), an oligodendrocyte myelin glycoprotein (OMG), a cytosolic protein (e.g., glyceraldehyde-3-phosphate dehydrogenase (GAPDH), alpha-synuclein (SNCA), cathepsin D (CTSD), AchE, LAMP1, REST, SYT, TH, SYP, SYNPO, PSD95, SV2A,
  • enriched vesicles from the biological sample are subsequently enriched for a specific type of vesicle.
  • the biological sample is enriched for exosomes and then the enriched exosomes are subsequently enriched for neural-derived exosomes.
  • the biological sample is enriched for individual neural cell sources of vesicles.
  • the neural cell sources of vesicles are microglia, neurons, or astrocytes.
  • vesicles are isolated or enriched from a biological sample by a method comprising: contacting a biological sample with an agent under conditions wherein a vesicle present in said biological sample binds to said agent to form a vesicle-agent complex; and isolating said vesicle from said vesicle-agent complex to obtain a sample containing said vesicle, wherein the purity of vesicles present in said sample is greater than the purity of vesicles present in said biological sample.
  • the agent is an antibody or a lectin. Lectins useful for forming a vesicle-lectin complex are described in U.S. Patent Application Publication No.
  • the vesicle is an exosome, a microparticle, a microvesicle, nanosomes, extracellular vesicles, or an ectosome.
  • the exosomes are neuron-derived exosomes, astrocyte-derived exosomes, oligodendrocyte-derived exosomes, or microglia-derived exosomes.
  • multiple isolating or enriching steps are performed. In certain aspects of the present embodiment, a first isolating step is performed to isolate exosomes from a blood sample and a second isolating step is performed to isolate neural-derived exosomes from other exosomes.
  • the methods further comprise releasing the vesicle from the vesicle-agent complex.
  • the vesicle is released by exposing the vesicle-agent complex to low pH between 3.5 and 1.5.
  • the vesicle is released using a competing peptide that competes for the binding of the selection antibody used in the methods of the present invention.
  • the released vesicle is neutralized by adding a high pH solution.
  • the released vesicle is lysed by incubating the released vesicles with a lysis solution.
  • the lysis solution contains inhibitors for proteases and phosphatases.
  • a subset of vesicles is separated from other vesicles in a biological sample using capture agents immobilized on a solid support.
  • capture agents bind selectively to a surface marker (e.g., membrane protein or adsorbed protein) on vesicles such that the capture agent can “capture” vesicles having the surface marker.
  • a surface marker e.g., membrane protein or adsorbed protein
  • capture is meant that the target vesicle can be separated from other vesicles in the sample by virtue of the binding of the capture agent to the surface marker on the vesicle.
  • the specificity of the capture agent determines the subset of vesicles from a biological sample that are captured on the solid support.
  • One or more capture agents can be used in combination in order to capture vesicles having different surface markers.
  • the solid support may comprise more than one type of capture agent associated therewith, for example, at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or more different capture agents that selectively bind to different biomarkers on the vesicles.
  • the vesicles targeted by a capture agent are exosomes, microparticles, microvesicles, nanosomes, extracellular vesicles, or ectosomes.
  • the capture agent selectively binds to neuron-derived exosomes, astrocyte-derived exosomes, oligodendrocyte-derived exosomes, or microglia-derived exosomes.
  • a capture agent can be chosen that selectively binds to an exosome surface marker (e.g., CD81) to capture exosomes generally, a neuron-specific protein (e.g., synaptosome associated protein 25 (SNAP25), neurogranin (NRGN), tau, phosphorylated tau, ⁇ -42, and synaptophysin) to capture neuron-derived exosomes, an astrocyte-specific protein (e.g., glial fibrillary acidic protein (GFAP) and excitatory amino acid transporter 1 (EAAT1) to capture astrocyte-derived exosomes, a microglia-specific protein (CD11b) to capture microglia-derived exosomes, an oligodendrocyte-specific protein (e.g.
  • the capture agent is associated with a solid support, either directly or indirectly.
  • Capture agents may be immobilized on the surface of a solid support, such as, but not limited to, a plate, slide, wafer, non-magnetic bead, magnetic bead, rod, particle, strand, disc, membrane, film, or the inner surface of a tube, channel, column, flow cell device, or microfluidic device.
  • a solid support may comprise various materials, including, but not limited to glass, quartz, silicon, metal, ceramic, plastic, nylon, polyacrylamide, agarose, resin, porous polymer monoliths, hydrogels, and composites thereof.
  • a substrate may be added to the surface of a solid support to facilitate attachment of a capture agent.
  • the vesicles can be screened for one or more membrane-bound and adsorbed biomarkers using detection agents without the need for lysis or permeabilization of the vesicles.
  • detection agents bind selectively to membrane-bound or adsorbed biomarkers on the vesicles.
  • the detection agent selectively binds to a neuron-specific protein (e.g., synaptosome associated protein 25 (SNAP25), neurogranin (NRGN), ⁇ -42, tau, phosphorylated tau, and synaptophysin), an astrocyte-specific protein (e.g., glial fibrillary acidic protein (GFAP) and excitatory amino acid transporter 1 (EAAT1)), a microglia-specific protein (CD11b), an oligodendrocyte-specific protein (e.g., myelin basic protein (MBP), an oligodendrocyte myelin glycoprotein (OMG), a cytosolic protein (e.g., glyceraldehyde-3-phosphate dehydrogenase (GAPDH), alpha-synuclein (SNCA), cathepsin D (CTSD), AchE, LAMP1, REST, SYT, TH, SYP, SYNPO, PSD95,
  • detection of biomarkers on vesicles captured on the solid support comprises using more than one type of detection agent, for example, at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or more different detection agents that selectively bind to different biomarkers on the vesicles.
  • Capture agents and detection agents may comprise, for example, antibodies, antibody fragments, antibody mimetics, or aptamers that specifically bind to a surface marker (e.g., membrane-bound or adsorbed protein) on a vesicle.
  • a surface marker e.g., membrane-bound or adsorbed protein
  • the phrase “specifically (or selectively) binds” refers to a binding reaction that is determinative of the presence of the surface marker on a vesicle in a heterogeneous population of proteins and other biologics.
  • the specified capture agents or detection agents bind to a particular surface marker on a vesicle at least two times the background and do not substantially bind in a significant amount to other proteins present in the sample.
  • the capture agent or detection agent comprises an antibody that specifically binds to a surface marker (e.g., membrane protein or adsorbed protein) on a vesicle.
  • a surface marker e.g., membrane protein or adsorbed protein
  • Any type of antibody may be used, including polyclonal and monoclonal antibodies, hybrid antibodies, altered antibodies, chimeric antibodies and, humanized antibodies, as well as: hybrid (chimeric) antibody molecules (see, for example, Winter et al. (1991) Nature 349:293-299; and U.S. Pat. No. 4,816,567); F(ab′) 2 and F(ab) fragments; F v molecules (noncovalent heterodimers, see, for example, Inbar et al.
  • the capture agent or detection agent comprises an aptamer that specifically binds to the target surface marker on a vesicle.
  • aptamer Any type of aptamer may be used, including a DNA, RNA, xeno-nucleic acid (XNA), or peptide aptamer that specifically binds to the target antibody isotype.
  • XNA xeno-nucleic acid
  • Such aptamers can be identified, for example, by screening a combinatorial library.
  • Nucleic acid aptamers e.g., DNA or RNA aptamers
  • that bind selectively to a target antibody isotype can be produced by carrying out repeated rounds of in vitro selection or systematic evolution of ligands by exponential enrichment (SELEX).
  • Peptide aptamers that bind to a target antibody isotype may be isolated from a combinatorial library and improved by directed mutation or repeated rounds of mutagenesis and selection.
  • Aptamers Tools for Nanotherapy and Molecular Imaging (R. N. Veedu ed., Pan Stanford, 2016), Nucleic Acid and Peptide Aptamers: Methods and Protocols (Methods in Molecular Biology, G. Mayer ed., Humana Press, 2009), Nucleic Acid Aptamers: Selection, Characterization, and Application (Methods in Molecular Biology, G.
  • the capture agent or detection agent comprises an antibody mimetic.
  • Any type of antibody mimetic may be used, including, but not limited to, affibody molecules (Nygren (2008) FEBS J. 275 (11):2668-2676), affilins (Ebersbach et al. (2007) J. Mol. Biol. 372 (1):172-185), affimers (Johnson et al. (2012) Anal. Chem. 84 (15):6553-6560), affitins (Krehenbrink et al. (2008) J. Mol. Biol.
  • Detection agents may further comprise a detectable label to facilitate detection and/or quantitation of biomarkers on vesicles.
  • Detectable labels include fluorescent, chemiluminescent, electrochemiluminescent, or bioluminescent tags, metals, dyes, radionuclides, and the like, attached to the specific binding agent (e.g., antibody, antibody fragment, antibody mimetic, or aptamer that specifically binds to a membrane-bound or adsorbed biomarker on vesicles).
  • the present invention provides methods for diagnosing or prognosing a neurological disorder in a subject, identifying a subject at risk of a neurological disorder, or prescribing a therapeutic regimen or predicting benefit from therapy in a subject having a neurological disorder. In some embodiments, the present invention provides methods for differential diagnosis of a neurological disorder in a subject.
  • the neurological disorder is selected from the group consisting of: Alzheimer's disease (AD), vascular disease dementia, frontotemporal dementia (FTD), corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), Lewy body dementia, tangle-predominant senile dementia, Pick's disease (PiD), argyrophilic grain disease, amyotrophic lateral sclerosis (ALS), other motor neuron diseases, Guam parkinsonism-dementia complex, FTDP-17, Lytico-Bodig disease, multiple sclerosis, traumatic brain injury (TBI), stroke, depression, bipolar disease, epilepsy, autism, schizophrenia, brain tumor, white matter disease, brain atrophy, mental retardation, cerebellar ataxia, concussion, subconcussive impacts, and Parkinson's disease.
  • AD Alzheimer's disease
  • FDD frontotemporal dementia
  • CBD corticobasal degeneration
  • PSP progressive supranuclear palsy
  • the present invention enables a medical practitioner to diagnose or prognose one or more neurological disorders in a subject. In other embodiments, the present invention enables a medical practitioner to rule out or eliminate one or more neurological diseases as a diagnostic possibility. In other embodiments, the methods of the present invention allow a medical practitioner to distinguish some forms of FTD from Alzheimer's disease. In yet other embodiments, the present invention enables a medical practitioner to identify a subject at risk of developing a neurological disorder. In other embodiments, the present invention enables a medical practitioner to predict whether a subject will later develop a neurological disorder. In further embodiments, the present invention enables a medical practitioner to prescribe a therapeutic regimen or predict benefit from therapy in a subject having a neurological disorder.
  • the present invention provides methods for diagnosing or prognosing cancer in a subject, identifying a subject at risk of developing cancer, or prescribing a therapeutic regimen or predicting benefit from therapy in a subject having cancer.
  • a cancer is characterized by the uncontrolled growth of abnormal cells anywhere in a body.
  • the abnormal cells may be termed cancer cells, malignant cells, or tumor cells. Cancer is not confined to humans; animals and other living organisms can get cancer.
  • the cancer may be malignant.
  • the cancer may be benign.
  • the cancer may be a recurrent and/or refractory cancer. Most cancers can be classified as a carcinoma, sarcoma, leukemia, lymphoma, myeloma, or a central nervous system cancer.
  • the cancer may be a sarcoma.
  • Sarcomas are cancers of the bone, cartilage, fat, muscle, blood vessels, or other connective or supportive tissue.
  • Sarcomas include, but are not limited to, bone cancer, fibrosarcoma, chondrosarcoma, Ewing's sarcoma, malignant hemangioendothelioma, malignant schwannoma, bilateral vestibular schwannoma, osteosarcoma, soft tissue sarcomas (e.g.
  • alveolar soft part sarcoma alveolar soft part sarcoma, angiosarcoma, cystosarcoma phylloides, dermatofibrosarcoma, desmoid tumor, epithelioid sarcoma, extraskeletal osteosarcoma, fibrosarcoma, hemangiopericytoma, hemangiosarcoma, Kaposi's sarcoma, leiomyosarcoma, liposarcoma, lymphangiosarcoma, lymphosarcoma, malignant fibrous histiocytoma, neurofibrosarcoma, rhabdomyosarcoma, and synovial sarcoma).
  • the cancer may be a carcinoma.
  • Carcinomas are cancers that begin in the epithelial cells, which are cells that cover the surface of the body, produce hormones, and make up glands.
  • carcinomas include breast cancer, pancreatic cancer, lung cancer, colon cancer, colorectal cancer, rectal cancer, kidney cancer, bladder cancer, stomach cancer, prostate cancer, liver cancer, ovarian cancer, brain cancer, vaginal cancer, vulvar cancer, uterine cancer, oral cancer, penic cancer, testicular cancer, esophageal cancer, skin cancer, cancer of the fallopian tubes, head and neck cancer, gastrointestinal stromal cancer, adenocarcinoma, cutaneous or intraocular melanoma, cancer of the anal region, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, cancer of the urethra, cancer of the renal pelvis, cancer of the ureter, cancer of the
  • the cancer is a skin cancer, such as a basal cell carcinoma, squamous, melanoma, nonmelanoma, or actinic (solar) keratosis.
  • the cancer is a prostate cancer.
  • the cancer may be a thyroid cancer, bladder cancer, or pancreatic cancer.
  • the cancer is a lung cancer.
  • Lung cancer can start in the airways that branch off the trachea to supply the lungs (bronchi) or the small air sacs of the lung (the alveoli).
  • Lung cancers include non-small cell lung carcinoma (NSCLC), small cell lung carcinoma, and mesotheliomia.
  • NSCLC non-small cell lung carcinoma
  • Examples of NSCLC include squamous cell carcinoma, adenocarcinoma, and large cell carcinoma.
  • the mesothelioma may be a cancerous tumor of the lining of the lung and chest cavity (pleura) or lining of the abdomen (peritoneum). The mesothelioma may be due to asbestos exposure.
  • the cancer may be a brain cancer, such as a glioblastoma.
  • the cancer may be a central nervous system (CNS) tumor.
  • CNS tumors may be classified as gliomas or nongliomas.
  • the glioma may be malignant glioma, high grade glioma, diffuse intrinsic pontine glioma. Examples of gliomas include astrocytomas, oligodendrogliomas (or mixtures of oligodendroglioma and astocytoma elements), and ependymomas.
  • Astrocytomas include, but are not limited to, low-grade astrocytomas, anaplastic astrocytomas, glioblastoma multiforme, pilocytic astrocytoma, pleomorphic xanthoastrocytoma, and subependymal giant cell astrocytoma.
  • Oligodendrogliomas include low-grade oligodendrogliomas (or oligoastrocytomas) and anaplastic oligodendriogliomas.
  • Nongliomas include meningiomas, pituitary adenomas, primary CNS lymphomas, and medulloblastomas. In some instances, the cancer is a meningioma.
  • the cancer may be a leukemia.
  • the leukemia may be an acute lymphocytic leukemia, acute myelocytic leukemia, chronic lymphocytic leukemia, or chronic myelocytic leukemia. Additional types of leukemias include hairy cell leukemia, chronic myelomonocytic leukemia, and juvenile myelomonocytic-leukemia.
  • the cancer is a lymphoma.
  • Lymphomas are cancers of the lymphocytes and may develop from either B or T lymphocytes.
  • the two major types of lymphoma are Hodgkin's lymphoma, previously known as Hodgkin's disease, and non-Hodgkin's lymphoma.
  • Hodgkin's lymphoma is marked by the presence of the Reed-Sternberg cell.
  • Non-Hodgkin's lymphomas are all lymphomas which are not Hodgkin's lymphoma.
  • Non-Hodgkin lymphomas may be indolent lymphomas and aggressive lymphomas.
  • Non-Hodgkin's lymphomas include, but are not limited to, diffuse large B cell lymphoma, follicular lymphoma, mucosa-associated lymphatic tissue lymphoma (MALT), small cell lymphocytic lymphoma, mantle cell lymphoma, Burkitt's lymphoma, mediastinal large B cell lymphoma, Waldenström macroglobulinemia, nodal marginal zone B cell lymphoma (NMZL), splenic marginal zone lymphoma (SMZL), extranodal marginal zone B cell lymphoma, intravascular large B cell lymphoma, primary effusion lymphoma, and lymphomatoid granulomatosis.
  • MALT mucosa-associated lymphatic tissue lymphoma
  • MALT mucosa-associated lymphatic tissue lymphoma
  • small cell lymphocytic lymphoma mantle cell lymphoma
  • Burkitt's lymphoma mediastinal large B cell
  • the present invention enables a medical practitioner to diagnose or prognose one or more cancers in a subject. In other embodiments, the present invention enables a medical practitioner to rule out or eliminate one or more cancers as a diagnostic possibility. In other embodiments, the methods of the present invention allow a medical practitioner to identify the origin of a cancer. In yet other embodiments, the present invention enables a medical practitioner to identify a subject at risk of developing cancer. In other embodiments, the present invention enables a medical practitioner to predict whether a subject will later develop cancer. In further embodiments, the present invention enables a medical practitioner to prescribe a therapeutic regimen or predict benefit from therapy in a subject having cancer. Exemplary biomarkers of the present invention that are useful in cancer diagnosis and prognosis include, but are not limited to, EpCAM, PD-L1, ErbB2, CK19.
  • the present invention provides methods for diagnosing or prognosing an immunological disorder in a subject, identifying a subject at risk of developing an immunological disorder, or prescribing a therapeutic regimen or predicting benefit from therapy in a subject having an immunological disorder.
  • Immunological disorders are diseases or conditions caused by a dysfunction of the immune system and include allergy, asthma, autoimmune diseases, autoinflammatory syndromes and immunological deficiency syndromes.
  • the present invention enables a medical practitioner to diagnose or prognose one or more immunological disorders in a subject. In other embodiments, the present invention enables a medical practitioner to rule out or eliminate one or more immunological disorders as a diagnostic possibility. In yet other embodiments, the present invention enables a medical practitioner to identify a subject at risk of developing an immunological disorder. In other embodiments, the present invention enables a medical practitioner to predict whether a subject will later develop an immunological disorder. In further embodiments, the present invention enables a medical practitioner to prescribe a therapeutic regimen or predict benefit from therapy in a subject having an immunological disorder. Exemplary biomarkers of the present invention that are useful in immunological disorder diagnosis and prognosis include, but are not limited to, TCR, CD16, CD28, CD32, CD79a, and TREM2.
  • the present invention provides methods for diagnosing or prognosing placental disease in a subject, identifying a subject at risk of developing a placental disease, or prescribing a therapeutic regimen or predicting benefit from therapy in a subject having a placental disease.
  • a placental disease is any disease, disorder, or pathology of the placenta.
  • the methods and biomarkers of the present invention may also be used for fetal assessment or diagnosis of fetal disorders, such as, for example, fetal alcohol syndrome or fetal genetic abnormalities.
  • Exemplary biomarkers of the present invention that are useful in placental disease or fetal assessment diagnosis and prognosis include, but are not limited to, PLAP, CSH1, and PSG1.
  • Biomarker levels on vesicles are assayed in a biological sample obtained from a subject having or at-risk of having a disease.
  • biomarker levels on vesicles are assayed in a biological sample obtained from a subject having or at-risk of having a neurological disorder (e.g., Alzheimer's disease).
  • biomarker levels on vesicles are assayed in a biological sample obtained from a subject having or at-risk of having cancer.
  • biomarker levels on vesicles are assayed in a biological sample obtained from a subject having or at-risk of having an immunological disorder.
  • biomarker levels on vesicles are assayed in a biological sample obtained from a subject having or at-risk of having a placental disorder.
  • one or more biomarkers are selected from the group consisting of a neuron-specific protein (e.g., synaptosome associated protein 25 (SNAP25), neurogranin (NRGN), tau, and synaptophysin), an astrocyte-specific protein (e.g., glial fibrillary acidic protein (GFAP) and excitatory amino acid transporter 1 (EAAT1)), a microglia-specific protein (CD11b), an oligodendrocyte-specific protein (e.g., myelin basic protein (MBP), an oligodendrocyte myelin glycoprotein (OMG)), and an extracellular vesicle-specific protein (dopamine transporter, DAT).
  • a neuron-specific protein e.g., synaptosome associated protein 25 (SNAP25), neurogranin (NRGN),
  • the biomarkers are CD171, phosphorylated tau T181, SNCA, and NRGN.
  • the biomarkers are acetylcholinesterase (AchE), Lysosomal Associated Membrane Protein 1 (LAMP1), CTSD, RE1 Silencing Transcription Factor (REST), synaptotagmin (SYT), monocyte chemotactic protein-1 (CCL2), IL34, glycogen synthase (GYS), (OR), death receptor 6 (DR6), heat shock protein (HSP), IL12beta, alpha-beta (A ⁇ ), and beta-secretase (BACE).
  • AchE acetylcholinesterase
  • LAMP1 Lysosomal Associated Membrane Protein 1
  • CTSD CTSD
  • REST RE1 Silencing Transcription Factor
  • SYT synaptotagmin
  • CCL2 monocyte chemotactic protein-1
  • IL34 glycogen synthase
  • one or more biomarkers are selected from the group consisting of cytosolic proteins, secretory proteins, membrane proteins and receptors and their pathological forms, including aggregates and mutated ones.
  • Biomarkers of the present invention include neurotransmitter receptors, such as, for example, dopamine receptors (D1 and D2), serotonin receptors (2A, 2C, and 3B), GABA receptors (1-6, 5. B1, B2), and glutamate receptors (1 and 2).
  • Other receptor biomarkers of the present invention include, insulin receptors, tumor necrosis factor receptors superfamily (TRAL, TNF receptor, death receptor 5 and 6), and neuropeptide receptors (orexin receptor, opioid receptor KOR).
  • Biomarkers of the present invention include membrane proteins, such as, for example, EpCAM, PD-L1, ErbB2, CK19, TCR, CD16, CD28, CD32, CD79a, TREM2, and NCAM.
  • membrane proteins such as, for example, EpCAM, PD-L1, ErbB2, CK19, TCR, CD16, CD28, CD32, CD79a, TREM2, and NCAM.
  • Other known neurological disorder biomarkers may be used in combination with the biomarkers of the present invention. Examples of such biomarkers are provided in US Patent Application Pub. No. 2015/0119278, the contents of which are hereby incorporated by reference.
  • immunoassay devices and methods are often used. These devices and methods can utilize labeled molecules in various sandwich, competitive, or non-competitive assay formats, to generate a signal that is related to the presence or amount of an analyte of interest. Additionally, certain methods and devices, such as biosensors and optical immunoassays, may be employed to determine the presence or amounts of analytes without the need for a labeled molecule.
  • the markers are analyzed using an immunoassay, although other methods are well known to those skilled in the art (for example, the measurement of marker RNA levels).
  • the presence or amount of a marker is generally determined using antibodies specific for each marker and detecting specific binding.
  • Any suitable immunoassay may be utilized, for example, an enzyme-linked immunosorbent assay (ELISA), immunofluorescent assay (IFA), immune-polymerase chain reaction assay, electro-chemiluminescence immunoassay (ECLIA), radioimmunoassay (RIA), competitive binding assay, planar waveguide technology, and the like. Specific immunological binding of the antibody to the marker can be detected directly or indirectly.
  • Direct labels include fluorescent or luminescent tags, metals, dyes, radionuclides, and the like, attached to the antibody.
  • Indirect labels include various enzymes well known in the art, such as alkaline phosphatase, horseradish peroxidase and the like.
  • immobilized antibodies specific for the surface markers on vesicles is also contemplated by the present invention.
  • the antibodies could be immobilized onto a variety of solid supports, such as magnetic or chromatographic matrix particles, the surface of an assay place (such as microtiter wells), pieces of a solid substrate material (such as plastic, nylon, paper), and the like.
  • An assay strip could be prepared by coating the antibody or a plurality of antibodies in an array on solid support. This strip could then be dipped into the test sample to capture vesicles through binding to surface markers, and then processed quickly through washes and detection steps with detection reagents, as described above, to generate a measurable signal, such as a colored spot.
  • the analysis of a plurality of markers may be carried out separately or simultaneously with one test sample.
  • Several markers on vesicles may be captured and/or detected using a combination of multiple capture agents and/or detection agents in one test for efficient processing of multiple of samples.
  • one skilled in the art would recognize the value of testing multiple samples (for example, at successive time points) from the same individual. Such testing of serial samples will allow the identification of changes in marker levels over time.
  • Increases or decreases in marker levels, as well as the absence of change in marker levels, would provide useful information about the disease status that includes, but is not limited to identifying the approximate time from onset of the event, the presence and amount of salvageable tissue, the appropriateness of drug therapies, the effectiveness of various therapies, identification of the severity of the event, identification of the disease severity, and identification of the patient's outcome, including risk of future events.
  • An assay consisting of a combination of the markers referenced in the instant invention may be constructed to provide relevant information related to differential diagnosis.
  • a panel may be constructed using 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or more or individual markers.
  • the analysis of a single marker or subsets of markers comprising a larger panel of markers could be carried out methods described within the instant invention to optimize clinical sensitivity or specificity in various clinical settings.
  • markers could be carried out in a variety of physical formats as well.
  • the use of microtiter plates or automation could be used to facilitate the processing of large numbers of test samples.
  • single sample formats could be developed to facilitate immediate treatment and diagnosis in a timely fashion, for example, in ambulatory transport or emergency room settings.
  • Particularly useful physical formats comprise surfaces having a plurality of discrete, addressable locations for the detection of a plurality of different analytes.
  • Such formats include protein microarrays, or “protein chips” and capillary devices.
  • Biomarkers of the present invention serve an important role in the early detection and monitoring of neurological disorders (e.g., Alzheimer's disease). Markers of such disorders are typically substances found in a bodily sample that can be measured. The measured amount can correlate to underlying disorder or disease pathophysiology, presence or absence of a neurological disorder, probability of a neurological disorder in the future. In patients receiving treatment for their condition the measured amount will also correlate with responsiveness to therapy.
  • a decrease or increase in the level of one or more biomarkers of the present invention is indicative of a neurological disorder. For example, an increase in phosphorylated tau T181 levels and/or a decrease in NRGN levels on exosomes having the CD171 membrane marker is indicative of Alzheimer's disease. Accordingly, the methods of the present invention are useful for the differential diagnosis of Alzheimer's disease.
  • a biomarker is measured by a method selected from the group consisting of immunohistochemistry, immunocytochemistry, immunofluorescence, immunoprecipitation, electro-chemiluminescence immunoassay, radioimmunoassay, immune-polymerase chain reaction, western blotting, and ELISA.
  • the methods of the present invention may be used in clinical assays to diagnose or prognose a neurological disorder in a subject, identify a subject at risk of a neurological disorder, and/or for prescribing a therapeutic regimen or predicting benefit from therapy in a subject having a neurological disorder.
  • Clinical assay performance can be assessed by determining the assay's sensitivity, specificity, area under the ROC curve (AUC), accuracy, positive predictive value (PPV), and negative predictive value (NPV).
  • AUC area under the ROC curve
  • PPV positive predictive value
  • NPV negative predictive value
  • the clinical performance of the assay may be based on sensitivity.
  • the sensitivity of an assay of the present invention may be at least about 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100%.
  • the clinical performance of the assay may be based on specificity.
  • the specificity of an assay of the present invention may be at least about 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100%.
  • the clinical performance of the assay may be based on area under the ROC curve (AUC).
  • the AUC of an assay of the present invention may be at least about 0.5, 0.55, 0.6, 0.65, 0.7, 0.75, 0.8, 0.85, 0.9, or 0.95.
  • the clinical performance of the assay may be based on accuracy.
  • the accuracy of an assay of the present invention may be at least about 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100%.
  • compositions useful in the methods of the present invention include compositions that specifically recognize a biomarker associated with a neurological disorder.
  • Such compositions may include capture agents and/or detection agents that recognize, for example, a neuron-specific protein biomarker, such as synaptosome associated protein 25 (SNAP25), ⁇ -42, neurogranin (NRGN), tau, phosphorylated tau, and synaptophysin, an astrocyte-specific protein biomarker, such as glial fibrillary acidic protein (GFAP) and excitatory amino acid transporter 1 (EAAT1), an oligodendrocyte-specific protein biomarker, such as myelin basic protein (MBP) and oligodendrocyte myelin glycoprotein (OMG), a microglia-specific protein (CD11b), a cytosolic protein (e.g., glyceraldehyde-3-phosphate dehydrogenase (GAPDH), alpha-synuclein (SNCA), catheps
  • the composition is selected from the group consisting of a peptide, a nucleic acid, an antibody, and a small molecule.
  • the present invention relates to compositions that specifically detect a biomarker associated with a neurological disorder.
  • the present invention is based upon the finding that GAPDH, CTSD, NRGN, MBP, GFAP, Tau, phosphorylated Tau, synaptophysin, ⁇ -42, CX3CL1, IL1b, IL34, CD81, CD63, CD171, SNAP25, EAAT1, SNCA, CD11b, OMG, AchE, LAMP1, REST, SYT, TH, SYP, SYNPO, PSD95, SV2A, CCL2, IL34, GYS, OR, DR6, HSP, IL12b, A ⁇ , and BACE can be used as biomarkers for AD and other neurological disorders.
  • compositions of the present invention specifically bind to and detect such biomarkers.
  • a composition may comprise a solid support comprising capture agents associated therewith that selectively bind to CD81, CD63, CD171, SNAP25, EAAT1, CD11b, or OMG.
  • a composition may comprise detection agents that selectively bind to GAPDH, CTSD, NRGN, MBP, GFAP, Tau, phosphorylated Tau, synaptophysin, ⁇ -42, SNCA, CX3CL1, IL1b, IL34, OMG, AchE, LAMP1, REST, SYT, TH, SYP, SYNPO, PSD95, SV2A, CCL2, IL34, GYS, OR, DR6, HSP, IL12b, A ⁇ , and/or BACE.
  • the composition comprises an antibody, where the antibody specifically binds to a biomarker or vesicles of the invention.
  • antibody as used herein and further discussed below is intended to include fragments thereof which are also specifically reactive with a biomarker or vesicle (e.g., exosome).
  • Antibodies can be fragmented using conventional techniques and the fragments screened for utility in the same manner as described above for whole antibodies. For example, F(ab) 2 fragments can be generated by treating antibody with pepsin. The resulting F(ab) 2 fragment can be treated to reduce disulfide bridges to produce Fab fragments.
  • Antigen-binding portions may also be produced by recombinant DNA techniques or by enzymatic or chemical cleavage of intact antibodies.
  • Antigen-binding portions include, inter alia, Fab, Fab′, F(ab′) 2 , Fv, dAb, and complementarity determining region (CDR) fragments, single-chain antibodies (scFv), single domain antibodies, bispecific antibodies, chimeric antibodies, humanized antibodies, diabodies and polypeptides that contain at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding to the polypeptide.
  • the antibody further comprises a label attached thereto and able to be detected (e.g., the label can be a radioisotope, fluorescent compound, enzyme or enzyme co-factor).
  • an antibody of the invention is a monoclonal antibody
  • the invention makes available methods for generating novel antibodies that specifically bind the biomarker or the exosome of the invention.
  • a method for generating a monoclonal antibody that specifically binds a biomarker or exosome may comprise administering to a mouse an amount of an immunogenic composition comprising the biomarker or exosome, or fragment thereof, effective to stimulate a detectable immune response, obtaining antibody-producing cells (e.g., cells from the spleen) from the mouse and fusing the antibody-producing cells with myeloma cells to obtain antibody-producing hybridomas, and testing the antibody-producing hybridomas to identify a hybridoma that produces a monocolonal antibody that binds specifically to the biomarker or exosome.
  • antibody-producing cells e.g., cells from the spleen
  • a hybridoma can be propagated in a cell culture, optionally in culture conditions where the hybridoma-derived cells produce the monoclonal antibody that binds specifically to the biomarker or exosome.
  • the monoclonal antibody may be purified from the cell culture.
  • the term “specifically reactive with” as used in reference to an antibody is intended to mean, as is generally understood in the art, that the antibody is sufficiently selective between the antigen of interest (e.g., a biomarker or exosome) and other antigens that are not of interest. In certain methods employing the antibody, such as therapeutic applications, a higher degree of specificity in binding may be desirable. Monoclonal antibodies generally have a greater tendency (as compared to polyclonal antibodies) to discriminate effectively between the desired antigens and cross-reacting polypeptides. One characteristic that influences the specificity of an antibody:antigen interaction is the affinity of the antibody for the antigen. Although the desired specificity may be reached with a range of different affinities, generally preferred antibodies will have an affinity (a dissociation constant) of about 10 ⁇ 6 , 10 ⁇ 7 , 10 ⁇ 8 , 10 ⁇ 9 or less.
  • Antibodies can be generated to bind specifically to an epitope of an exosome or a biomarker of the present invention, including, for example, neuron-derived exosomes, astrocyte-derived exosomes, oligodendrocyte-derived exosomes, and microglia-derived exosomes, or neuron-specific proteins selected from the group consisting of synaptosome associated protein 25 (SNAP25), neurogranin (NRGN), tau, phosphorylated tar, and synaptophysin, astrocyte-specific proteins selected from the group consisting of glial fibrillary acidic protein (GFAP) and excitatory amino acid transporter 1 (EAAT1), and oligodendrocyte-specific proteins selected from the group consisting of myelin basic protein (MBP) and oligodendrocyte myelin glycoprotein (OMG), a microglia-specific protein (CD11b), and chemokine (CX3CL1) or cytokine (IL1b, IL34,
  • the antibody generated is an anti-CD171 antibody, an anti-synaptosome associated protein 25 (SNAP25) antibody, an anti-neurogranin (NRGN) antibody, an anti-tau antibody, an anti-synaptophysin antibody, and anti-CD63 antibody, an anti-4-42 antibody, an anti-CD81 antibody, an anti-CTD antibody, an anti-GAPDH antibody, an anti-IL1b antibody, an anti-IL34 antibody, an anti-CX3CL1 antibody, an anti-glial fibrillary acidic protein (GFAP) antibody, an anti-excitatory amino acid transporter 1 (EAAT1) antibody, an anti-SNCA antibody, an anti-TH antibody, and anti-CD11b antibody, an anti-myelin basic protein (MBP) antibody, an anti-oligodendrocyte myelin glycoprotein (OMG) antibody, an anti-dopamine transporter (DAT) antibody, an anti-AchE antibody AchE, an anti-LAMP1 antibody LAMP
  • the techniques used to screen antibodies in order to identify a desirable antibody may influence the properties of the antibody obtained.
  • a variety of different techniques are available for testing interaction between antibodies and antigens to identify particularly desirable antibodies. Such techniques include ELISAs, surface plasmon resonance binding assays (e.g., the Biacore binding assay, Biacore AB, Uppsala, Sweden), sandwich assays (e.g., the paramagnetic bead system of IGEN International, Inc., Gaithersburg, Md.), western blots, immunoprecipitation assays, immunocytochemistry, and immunohistochemistry.
  • the present invention relates to compositions used for treating or preventing a neurological disorder.
  • the present invention is based upon the findings that the levels of CD81, GAPDH, CTSD, NRGN, MBP, GFAP, Tau, phosphorylated Tau (e.g., T181), synaptophysin, CD63, ⁇ -42, SNCA, CX3CL1, IL1b, IL34, AchE, LAMP1, REST, SYT, TH, SYP, SYNPO, PSD95, SV2A, CCL2, IL34, GYS, OR, DR6, HSP, IL12b, A ⁇ , and/or BACE are implicated in the pathology of a variety of neurological disorders, such as, for example, Alzheimer's disease.
  • biomarkers inside vesicles are analyzed in addition to the surface biomarkers.
  • the present invention relates to compositions for lysing vesicles (e.g., exosomes) in biological samples obtained from a subject.
  • Lytic agents useful in the methods of the present invention include: RIPA buffer; Tris-HCl (pH 6.8); glycerol; SDS; 2-mercaptoethanol; Triton-X 100; M-PER Reagent; T-PER solution; and CHAPS. Lytic agents may be incubated with biological samples to disrupt the membrane of the vesicles of the present invention and release vesicle cargo (e.g., exosomal proteins) for subsequent analysis.
  • vesicle cargo e.g., exosomal proteins
  • the present invention provides methods of treating a neurological disorder in a subject, comprising administering to the subject an effective amount of a composition, wherein the composition increases, decreases, or maintains the level of CD81, GAPDH, CTSD, NRGN, MBP, GFAP, Tau, phosphorylated Tau (e.g., T181), synaptophysin, CD63, ⁇ -42, SNCA, CX3CL1, IL1b, or IL34 in the subject.
  • a composition comprising administering to the subject an effective amount of a composition, wherein the composition increases, decreases, or maintains the level of CD81, GAPDH, CTSD, NRGN, MBP, GFAP, Tau, phosphorylated Tau (e.g., T181), synaptophysin, CD63, ⁇ -42, SNCA, CX3CL1, IL1b, or IL34 in the subject.
  • the composition prevents increases or decreases in CD81, GAPDH, CTSD, NRGN, MBP, GFAP, Tau, phosphorylated Tau (e.g., T181), synaptophysin, CD63, ⁇ -42, SNCA, CX3CL1, IL1b, IL34, AchE, LAMP1, REST, SYT, TH, SYP, SYNPO, PSD95, SV2A, CCL2, IL34, GYS, OR, DR6, HSP, IL12b, A ⁇ , or BACE levels.
  • CD81 e.g., GAPDH, CTSD, NRGN, MBP, GFAP
  • Tau phosphorylated Tau (e.g., T181)
  • synaptophysin CD63, ⁇ -42, SNCA, CX3CL1, IL1b, IL34, AchE, LAMP1, REST, SYT, TH, SYP, SYNPO, PSD95, SV2
  • the present invention provides methods of treating a neurological disorder in a subject, comprising administering to the subject an effective amount of a composition, wherein the composition normalizes the level of CD81, GAPDH, CTSD, NRGN, MBP, GFAP, Tau, phosphorylated Tau (e.g., T181), synaptophysin, CD63, ⁇ -42, SNCA, CX3CL1, IL1b, IL34, AchE, LAMP1, REST, SYT, TH, SYP, SYNPO, PSD95, SV2A, CCL2, IL34, GYS, OR, DR6, HSP, IL12b, A ⁇ , and/or BACE to a reference level.
  • a composition normalizes the level of CD81, GAPDH, CTSD, NRGN, MBP, GFAP, Tau, phosphorylated Tau (e.g., T181), synaptophysin, CD63, ⁇ -42, SNCA, CX3
  • the above-described assay reagents including a solid support with bound capture agents, detection agents, and optionally reagents for performing immunoassays, such as by ELISA, IFA, immune-polymerase chain reaction assay, ECLIA, or RIA, can be provided in kits, with suitable instructions and other necessary reagents, in order to conduct the assays for detecting biomarkers on vesicles, as described above.
  • the kit will normally contain in separate containers the solid support with bound capture agents, detection agents, control formulations (positive and/or negative), and other reagents that the assay format requires.
  • kit can also contain, depending on the particular assay used, other packaged reagents and materials (i.e., wash buffers, and the like). Assays, such as those described above, can be conducted using these kits.
  • the invention encompasses kits for detecting or monitoring a neurological disorder in a subject.
  • the kit will include the means for quantifying one or more biomarkers in a subject.
  • the kit will include means for collecting a biological sample, means for quantifying one or more biomarkers in the biological sample, and instructions for use of the kit contents.
  • the kit comprises a means for enriching or isolating exosomes in a biological sample.
  • the kit comprises a solid support with bound capture agents for isolating exosomes from a biological sample.
  • the kit comprises a means for quantifying the amount of a biomarker.
  • the means for quantifying the amount of a biomarker comprises reagents necessary to detect the amount of a biomarker.
  • the invention includes a kit for diagnosing or prognosing a neurological disorder in a subject, identifying a subject at risk of a neurological disorder, or prescribing a therapeutic regimen or predicting benefit from therapy in a subject having a neurological disorder
  • the kit comprising: a) a solid support comprising capture agents associated therewith, wherein at least one capture agent selectively binds to CD171, CD63, CD81, SNAP25, EAAT1, or OMG; and b) one or more detection agents, wherein the one or more detection agents selectively binds to CD81, GAPDH, CTSD, NRGN, MBP, GFAP, Tau, phosphorylated Tau (e.g., T181), CD63, ⁇ -42, CX3CL1, IL1b, IL34, AchE, LAMP1, REST, SYT, SYP, SYNPO, PSD95, SV2A, CCL2, IL34, GYS, OR, DR6, HSP, IL12
  • At least one capture agent or detection agent comprises an antibody, an antibody fragment, an antibody mimetic, or an aptamer that specifically binds to CD171, phosphorylated tau T181, or neurogranin.
  • the antibody is selected from the group consisting of a monoclonal antibody, a polyclonal antibody, a chimeric antibody, a nanobody, a recombinant fragment of an antibody, an Fab fragment, an Fab′ fragment, an F(ab′) 2 fragment, an F v fragment, and an scF v fragment.
  • the kit comprises an anti-neurogranin antibody, an anti-phosphorylated tau T181 antibody, and an anti-CD171 antibody.
  • Exosomes in biological fluids are a new resource of diagnostics in various diseases, because they carry physiological and pathological materials (proteins, metabolites, RNAs, small molecules, etc.) of the mother cells from which they originate and the microenvironment near the mother cells ( FIG. 1A ). Thus, quantification of such pathological materials in exosomes will be a foundation of the next generation of diagnostics.
  • exosome isolation and analysis are conducted on the same solid support without transferring the samples to other assay vessels, which avoids loss of material.
  • the method is performed using antibodies against at least two different markers on the surface of an exosome, which may include exosome membrane proteins and/or adsorbed markers.
  • an exosome marker such as a membrane marker (CD81, SNAP25, CD171, EAAT1, OMG, etc.) or adsorbed marker (Tau, NRGN, GFAP, MBP, etc.) is immobilized on an ELISA plate to allow capture of exosomes having that marker.
  • a second antibody against another exosome marker of interest which can also be a membrane marker (CD81, SNAP25, CD171, EAAT1, OMG, etc.) or adsorbed marker (Tau, NRGN, GFAP, MBP, etc.), is subsequently used to screen the exosomes captured by the first antibody.
  • a membrane marker CD81, SNAP25, CD171, EAAT1, OMG, etc.
  • adsorbed marker Tau, NRGN, GFAP, MBP, etc.
  • Cytosolic non-membrane proteins were detected on the surface of exosomes as follows. Neuron-specific anti-SNAP25 ( FIGS. 2A-2C ) or control mouse IgG ( FIGS. 2D-2F ) were immobilized onto white ELISA plates. Various volume (10, 2.5, and 0 ⁇ L) of plasma was suspended in PBS, 0.1% tween-20, or 0.1% triton-X100 PBS in a final volume of 40 ⁇ L, and applied to the ELISA plates.
  • GAPDH and CTSD are cytosolic proteins, we hypothesized that these would be detected only after permeabilization of exosomes by tween-20 or triton-X100, whereas membrane protein CD81 would show up in both cases. However, as shown in FIGS. 2A-2C , all 3 markers were detected in a plasma volume dependent manner, whereas no signal was detected on the mouse IgG-immobilized control ELISA plates. These results clearly showed that soluble cytosolic proteins such as GAPDH and CTSD may be detected on the surface of exosomes.
  • compositions of the present invention are useful for detecting biomarkers on vesicles captured on a solid support using a detection agent that selectively binds to the biomarkers, wherein the vesicles are not lysed or permeabilized.
  • cytosolic non-membrane proteins on the surface of exosomes was determined as follows. Antibodies against exosome surface marker CD81, neuron surface marker SNAP25, astrocyte surface marker (EAAT1), or oligodendrocyte surface marker (OMG) were immobilized onto white ELISA plates. Ten ⁇ L of plasma suspended in 40 ⁇ L of PBS were applied to the ELISA plates. After overnight incubation at 4° C., unbound materials were removed, then biotin-labeled antibodies against exosome surface marker CD81, general cytosolic marker GAPDH, cytosolic proteins in neuron (NRGN), oligodendrocyte (MBP), and astrocyte (GFAP) were applied to ELISA plates. No antibody control (tPBS) was also included. Then, conventional chemiluminescent ELISA procedure was carried out, and relative light unit (RLU) was determined ( FIGS. 3A-3D ).
  • RLU relative light unit
  • Marker proteins of the present invention were quantified as follows. Since immobilized antibody and biotin-labeled detection antibody bind to 2 different target molecules (such as CD81 and SNAP25), detectable targets standards should have both antigens in the single molecules. Thus, unlike conventional ELISAs, purified proteins or recombinant proteins are not applicable as a quantification standard. Thus, we first screened various plasma samples and found appropriate ones, which contained a large quantity of target exosomes. By assigning 100 units/mL to this plasma, dilution studies were carried out on SNAP25, EAAT1, and OMG plates ( FIGS. 4A-4D ).
  • Exosome adsorbed biomarkers were identified as follows. Control mouse IgG, mouse anti-human CD81, or mouse anti-human synaptosomal-associated protein 25 (SNAP25) were immobilized to ELISA plate, then pooled human plasma was applied to all of ELISA wells ( FIGS. 5A-5C ).
  • Various detection antibodies were used to carry out the ELISAs, which included not only antibodies against exosome membrane proteins (CD81, CD171, CD63, and SNAP25), but also, antibodies against various non-membrane proteins (glyceraldehyde 3-phosphate dehydrogenase (GAPDH), cathepsin D (CTSD), neurogranin (NRGN), myelin basic protein (MBP), glial fibrillary acidic protein (GFAP), tau, microtubule associated protein tau (tau) and phosphorylated tau T181, and amyloid b 1-42 peptide).
  • GPDH glycoprotein dehydrogenase
  • CSD cathepsin D
  • NRGN neurogranin
  • MBP myelin basic protein
  • GFAP glial fibrillary acidic protein
  • tau microtubule associated protein tau
  • tau phosphorylated tau T181, and amyloid b 1-42 peptide
  • ELISA signals were clearly positive for both the anti-CD81 and anti-SNAP25-immobilized plate, whereas very few signal was detected on control mouse IgG-immobilized plate, indicating that the system was CD81 or SNAP25 specific.
  • PBS non-specific signal without detection antibodies were very low, indicating that the assay was detection antibody specific.
  • the detection of membrane proteins confirmed that the assay captured exosomes ( FIGS. 5A and 5B ).
  • non-membrane proteins such as GAPDH, CTSD, NRGN, MBP, and GFAP were also detected ( FIG. 5C ).
  • the methods and compositions of the present invention were used to diagnose Alzheimer's disease in biological samples from humans as follows.
  • Anti-CD171 were immobilized on an ELISA plate. Twelve samples of EDTA plasma from subjects having Alzheimer's disease (AD) and age, gender-matched controls were applied to ELISA wells, and antibodies against CD81, CTSD. NRGN, p-tau T181 were used as detection antibodies ( FIGS. 6A-6E ).
  • the stability of plasma exosomal biomarker levels was determined as follows. Anti-CD81, anti-CD171, anti-SNAP25, anti-EAAT1, and anti-OMG were immobilized on separate ELISA plates. EDTA plasma was obtained from 7 control subjects every week for 2-3 weeks, and applied to the ELISA wells. For detection of exosome membrane targets ( FIGS.
  • an anti-CD81 probe was used on the CD81 plate (total exosome, TE); an anti-CD171 probe was used on the CD81 plate (CD171-based NDE, cNDE); an anti-SNAP25 probe was used on the CD81 plate (SNAP25-based NDE, sNDE); an anti-CD81 probe was used on the EAAT1 plate (ADE); and an anti-CD81 probe was used on the OMG plate (ODE).
  • GFAP probe on EAAT1 plate, MBP probe on OMG plate, NRGN probe on SNAP25 plate, tau probe on SNAP25 plate, and NRGN probe on CD171 plate respectively.
  • Plasma exosomal biomarkers were identified as follows. Anti-NRGN or control mouse IgG were immobilized on an ELISA plate. EDTA plasma or PBS alone were applied to the ELISA wells. Antibodies against NRGN, CD171, SNAP25, CD81, EAAT1, OMG were used for detection, and a PBS control was used for comparison.
  • Microglia targeting of exosome biomarkers was performed as follows. An anti-CD11b antibody or a control mouse IgG were immobilized on ELISA plates. EDTA plasma from 7 different donors (IR1-IR7) or PBS alone were applied to ELISA wells. Antibodies against GFAP ( FIG. 9B ), MBP ( FIG. 9C ), or NRGN ( FIG. 9D ) were used as detection antibodies.
  • microglia analysis can be done by assessing brain-derived proteins on the surface of CD11b + exosomes.
  • Four (GFAP) to five (MBP) donors showed positive signals higher than the 3 controls (PBS on IgG plate, plasma on IgG plate and PBS on CD1b plate), indicating the interaction of microglia with astrocytes and oligodendrocytes in these donors. None of the seven subjects, however, failed to show NRGN ( FIG. 9D ).
  • cytokines and chemokines were detected on the surface of brain-derived exosomes as follows.
  • Various concentrations of standard plasma FIGS. 10A, 10C, and 10E ) and fixed dilution of seven control plasma samples (1 ⁇ 8 dilution for IL1b and IL34 and 1 ⁇ 4 dilution for CX3CL1) and PBS control was applied to ELISA plates containing immobilized antibodies (anti-SNAP25, anti-EAAT1, anti-OMG, and control mouse IgG) to capture NDE, ADE, and ODE, respectively.
  • Control mouse IgG was used to evaluate non-specific bindings.
  • IL1b, IL34 and CX3CL1 levels on SNAP25, EAAT1, and OMG ELISA plates showed volume dependent increases in IL1b, IL34 and CX3CL1 levels on SNAP25, EAAT1, and OMG ELISA plates compared to control IgG-immobilized plates.
  • IL1b and IL34 on anti-OMG-immobilized plates were saturated at 2.5% plasma (see FIGS. 10A and 10C ).
  • Plasma samples were negative for IL1b, IL34, and CX3CL1, similar to those of negative control PBS alone.
  • IR2 was IL1b positive on the SNAP25 ELISA plate
  • IR3 was CX3CL1 positive on EAAT1 and OMG ELISA plates.
  • IRS showed positive IL1b levels and IL34 on SNAP25 plates, and detectable levels of CX3CL1 on EAAT1 and OMG ELISA plates.
  • IR7 showed positive IL1b on all ELISA plates, IL34 on SNAP25 ELISA plate, and detectable levels of CX3CL1 on both EAAT1 and OMG ELISA plates.
  • results showed that methods of the present invention are useful for detecting cytokines (IL1b and IL34) and chemokines (CX3CL1) on the surface of exosomes (SNAP25+ NDE, EAAT1+ ADE, and OMG+ ODE). These results also showed that cytokine and chemokine profiles are unique to each subset of brain-derived exosomes (NDE, ADE, and ODE). These results showed that the methods and compositions of the present invention are useful for identifying, detecting, and measuring biomarkers on vesicles, wherein the vesicles are not lysed or permeabilized. These results further suggested that the methods and compositions of the present invention would be useful for diagnosing neurological disease in a subject. These results further suggested that the methods and compositions of the present invention would be useful for diagnosing a disease or disorder.
  • ELISA assays require quantification standards to determine levels of target molecules in a test sample.
  • the exemplary assays of the present invention utilize two antibodies against different target molecules. Accordingly, purified or recombinant proteins are not applicable as a standard for such ELISA assays.
  • various plasma samples were screened, and one particular plasma sample was selected, for having high values of exosomes including NDE, ADE, and ODE positive exosomes. This plasma sample was subsequently used as a quantification standard. As shown in FIG. 11 , this plasma sample performed well as a standard for multiple ELISAs of the present invention. A standard curve was generated for each target by using four parameter logistic regression analysis.
  • the methods and compositions of the present invention were used for differential diagnosis of Alzheimer's disease and mild cognitive impairment in biological samples from humans as follows.
  • Anti-SNAP25 antibodies were immobilized on an ELISA plate.
  • FIG. 12 duplicate variation for all 16 biomarkers was small.
  • FIG. 13 shows the standard curves for all 16 biomarkers. Quantification of target surface protein biomarkers is shown in FIG. 14 . As shown in FIG. 14 , AchE, LAMP1, and CCL2 levels were significantly higher in MCI compared to control sample levels. These results showed that the methods and compositions of the present invention could be used to diagnose MCI and for the differential diagnosis of Alzheimer's disease and MCI.
  • the target biomarker levels in the plasma samples assayed above were normalized to SYT levels. Since neural-derived exosomes were captured by SNAP25-immobilized plates, the SYT normalization results in a second normalization of neuron markers. As shown in FIG. 16 , AchE/SYT, LAMP1/SYT, REST/SYT, CCL2/SYT, HSP/SYT, and IL12b/SYT levels were significantly different between AD, MCI, AD+MCI, and control samples. These results showed that the methods and compositions of the present invention could be used to diagnose AD, MCI, and for differential diagnosis of AD and MCI. These results further suggested that the methods and compositions of the present invention would be useful for diagnosing a disease or disorder.
  • Example 14 Enumeration of Brain-Derived Exosomes in Human Subjects with Parkinsonism-Plus Syndromes
  • NDEs neuron-derived, astrocyte-derived, and oligodendrocyte-derived exosomes
  • PD Parkinson's Disease
  • MSA multiple system atrophy
  • PSP progressive supranuclear palsy
  • Plasma samples were taken through venous puncture, and a total of 8 mL of blood was collected in EDTA-containing tubes. After collection, plasma was separated by centrifugation for 15 min at 2,000 g and distributed in polypropylene vials, then stored at ⁇ 80° C. until analysis.
  • ROC receiver operating characteristics
  • ELISA specificity was determined as follows. ELISA plates were immobilized with either target antibodies (anti-SNAP25, anti-EAAT1, and anti-OMG) or control IgG, then serial dilutions of plasma was applied. As shown in FIGS. 18A-18C , ELISA readings (RLU) increased in proportion to the volume of plasma on target antibodies-immobilized ELISA plates (filled circles; •), not control IgG-immobilized plates (open triangles; A). We also tested ELISA without biotin antibodies to assess non-specific binding of SA-HRP, however, no signal was detected. Since biotin antibodies were mixed with 10 ⁇ volume of control mouse IgG, non-specific IgG binding was eliminated in the assay.
  • Limit of detection (LOD) of NDE, ADE, and ODE was 0.519, 0, and 0.062 units/mL, respectively.
  • plasma levels of NDE were significantly higher in patients with PD than those with MSA ( FIGS.
  • FIGS. 23A-23C shows the ROC curves for the classification of patients with PD and controls based on the levels of plasma NDE, ADE, and ODE.
  • the AUC of the ROC curve for plasma NDE, ADE and ODE were 0.89, 0.83, and 0.88, respectively, indicating that plasma levels of NDE, ADE, and ODE will be applicable to diagnostic tests for PD with substantial accuracy.
  • Example 15 Detection of Pathological Form of ⁇ -Synuclein on the Surface of Neuron-, Astrocyte-, and Oligodendrocyte-Derived Exosomes
  • D2 dopamine receptor D2
  • Anti-SNAP25 neuron marker
  • anti-DRD2 anti-DRD2
  • control mouse IgG were immobilized on an ELISA plate.
  • human EDTA plasma or buffer control phosphate buffered saline, PBS
  • PBS buffer control
  • anti-SNAP25 and control mouse IgG were immobilized on an ELISA plate, human EDTA plasma or PBS was applied to the ELISA plate, followed by a reaction with biotinylated antibodies against SNAP25, CD81 (exosome common marker), DRD2, as well as PBS control. As shown in FIG. 28B , plasma was positive in anti-SNAP25, CD81, and DRD2 probes on anti-SNAP25 antibody ELISA plate.
  • a plasma dilution study was performed as follows. Anti-SNAP25 was immobilized on an ELISA plate and serial dilutions of human EDTA plasma samples (3 different donors) were applied to the ELISA plate, followed by the reaction with biotinylated anti-DRD2 and anti-CD81. As shown in FIGS. 29A and 29B , plasma dilution slope was almost identical among 3 donors. Therefore, the ratio of DRD2/CD81 was consistent (see FIG. 29C ).
  • DRD2 was detected in neuron-derived exosomes in plasma samples from humans with neurological disease.
  • Standard plasma was assigned to 100 units/mL, then using the dilution curve, ELISA reading (DRD2 probes on SNAP25 plate, and CD81 probes on SNAP25 plate (NDE)) of each sample was converted to units/mL (U/mL).
  • Plasma samples from 15 each of control, Parkinson's disease (PD), multiple system atrophy (MSA), and 7 patients with progressive supranuclear palsy (PSP) were analyzed. PD and MSA patients were further categorized by the severity of disease (mRS score). As shown in FIGS.
  • the levels of DRD2 in PD were significantly lower than control, although the levels of NDE were significantly higher in PD than control.
  • the ratio (DRD2/NDE) showed more significant differences between PD samples and control samples.
  • the decrease in DRD2 was shown in the mild cases of PD (mRS score 1+2), indicating that DRD2 may be used as a marker for the early screening of PD patients as well as the monitoring of PD progression.
  • ROC curve and area under the curve (AUC) for DRD2 levels between PD and control samples are shown in FIG. 31 .
  • ELISA enzyme linked immunosorbent assay
  • FIG. 32A antibodies against monomer
  • FIG. 32B antibodies against monomer
  • FIG. 32C oligomer ⁇ -synuclein
  • TH tyrosine hydroxylase
  • the plasma samples on DAT + ELISA wells all showed dose-dependent signal changes, whereas those of control IgG ELISA wells stayed very low, 1/10 to 1/100 lower than those of DAT + ELISA wells. More importantly, the slopes of three plasma samples were very similar, indicating that the dilution curve of one particular plasma can be used as a universal quantification standard.
  • Example 18 Detection of SNAP25-Positive, EAAT1-Positive, and OMG-Positive Extracellular Vesicles
  • SNAP25-positive, EAAT1-positive, and OMG-positive extracellular vesicles were captured on ELISA plates and analyzed as follows. Plasma samples obtained from control donors were applied to ELISA plates where antibodies against SNAP25, EAAT1, and OMG were previously immobilized. After captured extracellular vesicles (EV) were washed extensively, EVs were eluted by incubation with a pH 12.5 elution buffer for 5 minutes, then immediately neutralized. These samples were applied to Nanoparticle Tracking Analysis (NanoSight).
  • EV was detected in all samples with the size range from 100-400 nm. Moreover, EV size of SNAP25 was smaller than those of EAAT1 and OMG.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Urology & Nephrology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Cell Biology (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Analytical Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
US15/989,120 2017-05-24 2018-05-24 Detection of biomarkers on vesicles for the diagnosis and prognosis of diseases and disorders Abandoned US20180340945A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/989,120 US20180340945A1 (en) 2017-05-24 2018-05-24 Detection of biomarkers on vesicles for the diagnosis and prognosis of diseases and disorders

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201762510726P 2017-05-24 2017-05-24
US201762547024P 2017-08-17 2017-08-17
US15/989,120 US20180340945A1 (en) 2017-05-24 2018-05-24 Detection of biomarkers on vesicles for the diagnosis and prognosis of diseases and disorders

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/945,058 Continuation US20240192228A1 (en) 2022-09-14 Detection of biomarkers on vesicles for the diagnosis and prognosis of diseases and disorders

Publications (1)

Publication Number Publication Date
US20180340945A1 true US20180340945A1 (en) 2018-11-29

Family

ID=64397085

Family Applications (1)

Application Number Title Priority Date Filing Date
US15/989,120 Abandoned US20180340945A1 (en) 2017-05-24 2018-05-24 Detection of biomarkers on vesicles for the diagnosis and prognosis of diseases and disorders

Country Status (6)

Country Link
US (1) US20180340945A1 (de)
EP (1) EP3630947A4 (de)
JP (2) JP7399712B2 (de)
CA (1) CA3065999A1 (de)
IL (1) IL270864A (de)
WO (1) WO2018218090A1 (de)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109738630A (zh) * 2019-01-14 2019-05-10 复旦大学附属华山医院 一种高灵敏的磁珠分选联合胶体金标记的外泌体免疫电镜方法
US20200191778A1 (en) * 2018-12-18 2020-06-18 The University Of Memphis Research Foundation Compositions and methods for the detection and molecular profiling of membrane bound vesicles
CN113614531A (zh) * 2019-01-31 2021-11-05 新加坡国立大学 检测神经变性疾病的方法
US11186636B2 (en) 2017-04-21 2021-11-30 Amgen Inc. Anti-human TREM2 antibodies and uses thereof
WO2022058881A1 (en) * 2020-09-15 2022-03-24 Neurodex, Inc. Diagnostic assays employing neuron-derived exosomes
CN114280016A (zh) * 2021-12-07 2022-04-05 广州兆瑞医学生物科技有限公司 一种外泌体检测方法
US11407991B2 (en) 2011-05-11 2022-08-09 Exosome Diagnostics, Inc. Nucleic acid extraction from heterogeneous biological materials
US20220395795A1 (en) * 2020-12-25 2022-12-15 Nantong University Method for extracting nerve tissue-derived exosomes
EP3993776A4 (de) * 2019-07-02 2023-08-30 Ohio State Innovation Foundation Therapien für neurodegenerative erkrankungen unter verwendung der haut-hirn achse
US11852635B2 (en) * 2016-11-16 2023-12-26 Nano Somix, Inc Quantification of subpopulations of exosomes and diagnosis of neurogenerative disorders
US11899024B2 (en) * 2017-07-12 2024-02-13 Exosome Diagnostics, Inc. Treatment and diagnosis of parkinson's disease using isolated and enriched populations of biofluid-derived extracellular vesicles

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021107155A1 (ja) * 2019-11-29 2021-06-03 富士フイルム和光純薬株式会社 パーキンソン病の診断を補助する方法、バイオマーカー、試薬キット及び装置
WO2021186478A1 (en) * 2020-03-19 2021-09-23 Urvogelbio Private Limited Biomarker panels, systems, and methods for risk stratification of a subject for alzheimer's disease
US11828210B2 (en) 2020-08-20 2023-11-28 Denso International America, Inc. Diagnostic systems and methods of vehicles using olfaction
US11881093B2 (en) 2020-08-20 2024-01-23 Denso International America, Inc. Systems and methods for identifying smoking in vehicles
US11636870B2 (en) 2020-08-20 2023-04-25 Denso International America, Inc. Smoking cessation systems and methods
US11760170B2 (en) 2020-08-20 2023-09-19 Denso International America, Inc. Olfaction sensor preservation systems and methods
US11813926B2 (en) 2020-08-20 2023-11-14 Denso International America, Inc. Binding agent and olfaction sensor
US11932080B2 (en) 2020-08-20 2024-03-19 Denso International America, Inc. Diagnostic and recirculation control systems and methods
US11760169B2 (en) 2020-08-20 2023-09-19 Denso International America, Inc. Particulate control systems and methods for olfaction sensors
CN117916600A (zh) * 2021-09-08 2024-04-19 东曹株式会社 阿尔茨海默病的检测方法和检测试剂
WO2023112482A1 (ja) * 2021-12-14 2023-06-22 シスメックス株式会社 細胞外小胞の測定方法、神経変性に関する情報の取得方法、細胞外小胞の単離方法及び試薬キット

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150119278A1 (en) * 2013-10-24 2015-04-30 Nanosomix, Inc. Biomarkers and diagnostic methods for alzheimer's disease and other neurodegenerative disorders
US20160334402A1 (en) * 2015-05-13 2016-11-17 Miltenyl Biotec GmbH Method for analyzing markers on the surface of vesicles

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2459228B (en) 2007-07-25 2012-06-20 Univ Louisville Res Found Exosome-associated microrna as a diagnostic marker
WO2010065765A2 (en) * 2008-12-04 2010-06-10 Aethlon Medical, Inc. Affinity capture of circulating biomarkers
WO2012170037A1 (en) * 2011-06-10 2012-12-13 Hitachi Chemical Co., Ltd. Vesicle capturing devices and methods for using same
EP3366784B1 (de) * 2014-02-28 2019-11-13 Exosome Sciences Inc. Hirnspezifische exosom-basierte diagnostika und extrakorporale therapien
EP3161482B1 (de) 2014-06-27 2020-08-26 XY Evergreen Technology Company Verfahren zur anreicherung von exosomen aus dem zns
AU2015330855A1 (en) * 2014-10-09 2017-04-27 Celularity Inc. Placenta-derived adherent cell exosomes and uses thereof
WO2016121821A1 (ja) 2015-01-27 2016-08-04 Jsr株式会社 分離方法、検出方法、シグナル測定方法、疾患の判定方法、薬効評価方法、キット、液状組成物及び検体希釈液
US10203342B2 (en) * 2015-06-11 2019-02-12 Nanosomix, Inc. Biomarkers and differential diagnosis of alzheimer's disease and other neurodegenerative disorders

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150119278A1 (en) * 2013-10-24 2015-04-30 Nanosomix, Inc. Biomarkers and diagnostic methods for alzheimer's disease and other neurodegenerative disorders
US20160334402A1 (en) * 2015-05-13 2016-11-17 Miltenyl Biotec GmbH Method for analyzing markers on the surface of vesicles

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11407991B2 (en) 2011-05-11 2022-08-09 Exosome Diagnostics, Inc. Nucleic acid extraction from heterogeneous biological materials
US11852635B2 (en) * 2016-11-16 2023-12-26 Nano Somix, Inc Quantification of subpopulations of exosomes and diagnosis of neurogenerative disorders
US11186636B2 (en) 2017-04-21 2021-11-30 Amgen Inc. Anti-human TREM2 antibodies and uses thereof
US11899024B2 (en) * 2017-07-12 2024-02-13 Exosome Diagnostics, Inc. Treatment and diagnosis of parkinson's disease using isolated and enriched populations of biofluid-derived extracellular vesicles
US20200191778A1 (en) * 2018-12-18 2020-06-18 The University Of Memphis Research Foundation Compositions and methods for the detection and molecular profiling of membrane bound vesicles
US11988663B2 (en) * 2018-12-18 2024-05-21 The University Of Memphis Research Foundation Compositions and methods for the detection and molecular profiling of membrane bound vesicles
CN109738630A (zh) * 2019-01-14 2019-05-10 复旦大学附属华山医院 一种高灵敏的磁珠分选联合胶体金标记的外泌体免疫电镜方法
CN109738630B (zh) * 2019-01-14 2023-08-22 复旦大学附属华山医院 一种高灵敏的磁珠分选联合胶体金标记的外泌体免疫电镜方法
CN113614531A (zh) * 2019-01-31 2021-11-05 新加坡国立大学 检测神经变性疾病的方法
EP3993776A4 (de) * 2019-07-02 2023-08-30 Ohio State Innovation Foundation Therapien für neurodegenerative erkrankungen unter verwendung der haut-hirn achse
WO2022058881A1 (en) * 2020-09-15 2022-03-24 Neurodex, Inc. Diagnostic assays employing neuron-derived exosomes
US20220395795A1 (en) * 2020-12-25 2022-12-15 Nantong University Method for extracting nerve tissue-derived exosomes
US11731098B2 (en) * 2020-12-25 2023-08-22 Nantong University Method for extracting nerve tissue-derived exosomes
CN114280016A (zh) * 2021-12-07 2022-04-05 广州兆瑞医学生物科技有限公司 一种外泌体检测方法

Also Published As

Publication number Publication date
EP3630947A4 (de) 2021-04-07
JP2023153781A (ja) 2023-10-18
IL270864A (en) 2020-01-30
EP3630947A1 (de) 2020-04-08
JP7399712B2 (ja) 2023-12-18
WO2018218090A1 (en) 2018-11-29
CA3065999A1 (en) 2018-11-29
JP2020523559A (ja) 2020-08-06

Similar Documents

Publication Publication Date Title
US20180340945A1 (en) Detection of biomarkers on vesicles for the diagnosis and prognosis of diseases and disorders
US20230168252A1 (en) Dectection of exosomes and exosomal biomarkers for the diagnosis and prognosis of diseases and disorders
US11619637B2 (en) Biomarkers and diagnostic methods for Alzheimer's disease and other neurodegenerative disorders
US11852635B2 (en) Quantification of subpopulations of exosomes and diagnosis of neurogenerative disorders
US10203342B2 (en) Biomarkers and differential diagnosis of alzheimer's disease and other neurodegenerative disorders
JP7177702B2 (ja) アルツハイマー病および他の神経変性障害のシナプスタンパク質バイオマーカーおよび鑑別診断
US20200341011A1 (en) Astrocyte exosome complement-based assay for neuroinflammation in alzheimer's disease and uses thereof
US20180080945A1 (en) Biomarkers and diagnostic methods for alzheimer's disease and other neurodegenerative disorders
US20240192228A1 (en) Detection of biomarkers on vesicles for the diagnosis and prognosis of diseases and disorders

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

AS Assignment

Owner name: NANOSOMIX, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MITSUHASHI, MASATO;REEL/FRAME:054868/0064

Effective date: 20180824

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

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