US20190240194A1 - Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases - Google Patents

Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases Download PDF

Info

Publication number
US20190240194A1
US20190240194A1 US16/328,956 US201716328956A US2019240194A1 US 20190240194 A1 US20190240194 A1 US 20190240194A1 US 201716328956 A US201716328956 A US 201716328956A US 2019240194 A1 US2019240194 A1 US 2019240194A1
Authority
US
United States
Prior art keywords
day
cromolyn
compound
cells
microglial
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
US16/328,956
Other languages
English (en)
Inventor
David R. Elmaleh
Rudolph E. Tanzi
Timothy M. Shoup
Ana Gricluc
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.)
General Hospital Corp
Original Assignee
General Hospital Corp
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 General Hospital Corp filed Critical General Hospital Corp
Priority to US16/328,956 priority Critical patent/US20190240194A1/en
Assigned to THE GENERAL HOSPITAL CORPORATION reassignment THE GENERAL HOSPITAL CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SHOUP, TIMOTHY M., GRICLUC, Ana, TANZI, RUDOLPH E., ELMALEH, DAVID R.
Publication of US20190240194A1 publication Critical patent/US20190240194A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/428Thiazoles condensed with carbocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/007Pulmonary tract; Aromatherapy
    • A61K9/0073Sprays or powders for inhalation; Aerolised or nebulised preparations generated by other means than thermal energy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/22Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/22Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4
    • C07D311/24Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 4 with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2

Definitions

  • NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice,” Nature, 2013, 493(7434):674-8; Theeriault, et al., “The dynamics of monocytes and microglia in Alzheimer's disease,” Alzheimers Res Ther., 2015, 7:41; Nau et al., “Strategies to increase the activity of microglia as efficient protectors of the brain against infections,” Front Cell Neurosci., 2014, 8:138.)
  • AD Alzheimer's Disease
  • ALS Amyotrophic Lateral Sclerosis
  • a ⁇ amyloid- ⁇
  • microglial cells surrounding A ⁇ plaques are not as efficacious in degrading A ⁇ as newly infiltrated macrophages or monocytes (See, Thériault, et al., 2015; Varnum, et al., “The classification of microglial activation phenotypes on neurodegeneration and regeneration in Alzheimer's disease brain,” Arch. Immunol. Ther. Exp. (Warsz), 2012, 60(4):251-66)
  • microglia are indeed capable of internalizing fibrillar and soluble A ⁇ , but are unable to process these peptides.
  • See Chung, et al. “Uptake, degradation, and release of fibrillar and soluble forms of Alzheimer's amyloid beta-peptide by microglial cells,” J Biol. Chem., 1999, 274:32301-8).
  • microglia undergo a switch from an M2- to an M1-skewed activation phenotype during aging.
  • Mphi1 and Mphi2 can be re-polarized by Th2 or Th1 cytokines, respectively, and respond to exogenous danger signals
  • microglia are activated by extracellularly deposited A ⁇ peptide (Lotz, et al., “Amyloid beta peptide 1-40 enhances the action of Toll-like receptor-2 and -4 agonists but antagonizes Toll-like receptor-9-induced inflammation in primary mouse microglial cell cultures,” J. Neurochem., 2005, 94:289-298; Reed-Geaghan, et al., “CD14 and toll-like receptors 2 and 4 are required for fibrillar A ⁇ -stimulated microglial activation,” J. Neurosci., 2009, 29:11982-11992).
  • M1 activated microglia can produce reactive oxygen species and result in increased production of pro-inflammatory cytokines such as TNF ⁇ and interleukin (IL)-1 ⁇ .
  • the M1-type response of microglial cells has been shown to lower amyloid load but exacerbate neurofibrillary tangle pathology.
  • Shaftel et al. (Shaftel, et al., “Sustained hippocampal IL-1 ⁇ overexpression mediates chronic neuroinflammation and ameliorates Alzheimer plaque pathology,” J. Clin. Invest., 2007, 117(6):1595-604) have shown that IL-1 ⁇ expression may underlie a beneficial neuroinflammatory response in AD, and that IL-1 ⁇ overexpression in the hippocampus of APP/PS1 transgenic mice results in decreased amyloid burden.
  • the authors suggest that IL-1 ⁇ -mediated activation of microglia is the mechanism for the reductions in amyloid deposition. Further, Montgomery et al.
  • Macrophage M2 activation is associated with mediators that are known to contribute to the anti-inflammatory actions and reorganization of extracellular matrix (Zhu, et al., “Acidic mammalian chitinase in asthmatic Th2 inflammation and IL-13 pathway activation”, Science, 2004, 304(5677):1678-82; Walker, et al., 2015; Wilcock, et al., 2012).
  • Microglia with M2a phenotypes have increased phagocytosis and produce growth factors such as insulin-like growth factor-1 and anti-inflammatory cytokines such as IL-10.
  • M2a Stimulation of macrophages by IL-4 and/or IL-13 results in an M2a state, sometimes called a wound-healing macrophage (Edwards, et al., “Biochemical and functional characterization of three activated macrophage populations,” J. Leukoc Biol., 2006, 80(6):1298-307) and it is generally characterized by low production of pro-inflammatory cytokines (IL-1, TNF and IL-6).
  • IL-1, TNF and IL-6 pro-inflammatory cytokines
  • the M2a responses are primarily observed in allergic responses, extracellular matrix deposition, and remodeling.
  • M2b macrophages are unique in that they express high levels of pro-inflammatory cytokines, characteristic of M1 activation, but also express high levels of the anti-inflammatory cytokine IL-10. (See, Moser D M., “The many faces of macrophage activation,” J. Leukoc Biol., 2003, 73(2):209-12).
  • M2c macrophage state is stimulated by IL-10 and is sometimes referred to as a regulatory macrophage.
  • M2c macrophages have anti-inflammatory activity that plays a role in the phagocytosis of cellular debris without the classical pro-inflammatory response (See, Moser D M., 2003). These cells express TGF ⁇ and high IL-10 as well as matrix proteins. (See, Mantovani, et al., “The chemokine system in diverse forms of macrophage activation and polarization,” Trends Immunol., 2004, 25:677-686; Wilcock, et al., 2012). Plunkett et al.
  • microglia activated by extracellularly deposited A ⁇ protect neurons by triggering anti-inflammatory/neurotrophic M2 activation and by clearing A ⁇ via phagocytosis. This is a potential avenue for new therapeutic targets.
  • Yamamoto, et al. “Interferon-gamma and tumor necrosis factor-alpha regulate amyloid-beta plaque deposition and beta-secretase expression in Swedish mutant APP transgenic mice,” Am. J. Pathol., 2007, 170:680-692; Yamamoto, et al., “Cytokine-mediated inhibition of fibrillar amyloid-beta peptide degradation by human mononuclear phagocytes,” J. Immunol., 2008, 181:3877-3886).
  • Mantovani et al. (Mantovani, et al., 2004) studied the effect of IL-4 as an important modulator of M2a microglial activation. It has been shown that gene delivery of IL-4 into APP+PS1 mice partially suppressed glial accumulation in the hippocampus, directly enhanced neurogenesis, restored impaired spatial learning, and also reduced A ⁇ deposition (Kiyota, et al., 2010).
  • Yamamoto et al. (Yamamoto, et al., 2007, 2008) examined macrophage-mediated A ⁇ degradation using pro- and anti-inflammatory cytokines in primary cultured human monocyte-derived macrophages (MDM) and microglia. These studies showed that anti-inflammatory and regulatory cytokines lead to an increase in M2a or M2c activation and enhanced A ⁇ clearance. Kiyota et al. (Kiyota et al., 2011) have shown sustained expression of IL-4 reduced astro/microgliosis, amyloid- ⁇ peptide (A ⁇ ) oligomerization and deposition, and enhanced neurogenesis.
  • MDM monocyte-derived macrophages
  • the invention encompasses methods of treating a neuron inflammation condition comprising administering a therapeutically effective amount to a patient in need thereof of at least one compound having the following formula:
  • the method uses the following compounds:
  • the neuron inflammation condition is at least one of ALS, AD, ischemic stroke, or prion disease.
  • the compounds may be administered intraperitoneally (IP) and/or intravenously (IV).
  • IP intraperitoneally
  • IV intravenously
  • the compounds may be administered at a dose between about 1 mg and about 1000 mg per day.
  • the method of administration may be transdermally or by inhalation.
  • the method is a method of treating ALS further comprising co-administering CD4+; siRNA; miRNA that ameliorate ALS; glial morphology modifier; SOD1 control; Riluzole; or another M1; M2 conversion active drug that controls neuroinflammation.
  • the invention relates to any of the methods described herein, provided the compound is not cromolyn disodium. In certain embodiments, the invention relates to any of the methods described herein, provided the compound is not cromolyn disodium, F-cromolyn disodium, ET-cromolyn, or F-ET-cromolyn when the neuron inflammation condition is AD.
  • the invention relates to any one of the following compounds:
  • FIG. 1B illustrates representative images of localization of amyloid deposits (6E10) and microglia (Iba1) in mice treated with Cromolyn Sodium (3.15 mg/kg) or PBS daily for seven days.
  • FIG. 1C illustrates the effect of Cromolyn Sodium on microglial A ⁇ uptake in vitro, where after the incubation, the concentrations of A ⁇ x-40 ( FIG. 1C left) A ⁇ x-42 ( FIG. 1C , right) in media were measured using A ⁇ ELISA.
  • FIG. 2 illustrates the plaques and the microglial cells surrounding those deposits in Tg-2576 mice of the study of Example 2.
  • the figure shows representative pictures of amyloid deposits and Iba-1 positive microglia.
  • FIG. 3 illustrates the results of BV2 microglial cells treated with cromolyn, and with cromolyn and ibuprofen exhibit increased A ⁇ 42 uptake levels relative to BV2 microglia treated with the vehicle.
  • FIG. 4 illustrates the results of an A ⁇ aggregation inhibition assay using various compounds described herein.
  • FIG. 5 graphically illustrates that Cromolyn significantly affects the levels of brain TBS soluble A ⁇ and the ratios of A ⁇ (42:40).
  • FIG. 6A shows na ⁇ ve BV2 microglial cells treated with DMSO (control) for 16 h. Afterwards, cells were incubated with fluorescently-labeled A ⁇ 42 and DMSO or cromolyn sodium for 2 hours. After incubation, cells were labeled with a plasma membrane dye (PM) and imaged.
  • PM plasma membrane dye
  • FIG. 6B shows na ⁇ ve BV2 microglial cells treated with DMSO (control) for 16 h. Afterwards, cells were incubated with fluorescently-labeled A ⁇ 42 and DMSO or cromolyn sodium for 2 hours.
  • FIG. 6C showns na ⁇ ve BV2 microglial cells treated with cromolyn sodium (500 ⁇ M) for 16 hours. Afterwards, cells were incubated with fluorescently-labeled A ⁇ 42 and DMSO or cromolyn sodium for 2 hours. After incubation, cells were labeled with a plasma membrane dye (PM) and imaged.
  • PM plasma membrane dye
  • FIG. 6D showns na ⁇ ve BV2 microglial cells treated with cromolyn sodium (500 ⁇ M) for 16 hours. Afterwards, cells were incubated with fluorescently-labeled A ⁇ 42 and DMSO or cromolyn sodium for 2 hours.
  • FIG. 7A graphically illustrates that cromolyn sodium promotes microglial A ⁇ 42 uptake.
  • BV2 microglial cells were treated with DMSO or different concentrations of cromolyn sodium for 16 hours. Then, cells were incubated with soluble untagged A ⁇ 42 and DMSO or cromolyn sodium for 2 hours, and collected for ELISA analysis. Both na ⁇ ve BV2 and BV2-CD33 WT microglial cells treated with cromolyn sodium exhibited increased A ⁇ 42 uptake levels in comparison to cells treated with the vehicle (DMSO).
  • FIG. 7B graphically illustrates that cromolyn sodium promotes microglial A ⁇ 42 uptake.
  • BV2 cells stably expressing CD33 (BV2-CD33 WT ) were treated with DMSO or different concentrations of cromolyn sodium for 16 hours. Then, cells were incubated with soluble untagged A ⁇ 42 and DMSO or cromolyn sodium for 2 hours, and collected for ELISA analysis. Both na ⁇ ve BV2 and BV2-CD33 WT microglial cells treated with cromolyn sodium exhibited increased A ⁇ 42 uptake levels in comparison to cells treated with the vehicle (DMSO).
  • DMSO vehicle
  • FIG. 8 graphically illustrates that compound C8 displays toxicity when tested at 100 ⁇ M or higher concentration in LDH assay.
  • Na ⁇ ve BV2 microglial cells were treated with DMSO or cromolyn derivatives for 3 hours at different concentrations.
  • C1, C2, C5, C6, C7 and C8 were tested at 10, 50, 100 and 150 ⁇ M, while C3 and C4 were assessed at 5, 25, 50 and 75 ⁇ M due to solubility limit in DMSO.
  • cells were incubated with soluble untagged A ⁇ 42 peptide and DMSO or cromolyn derivatives for 2 hours.
  • LDH lactate dehydrogenase
  • BV2 microglial cells treated with the cromolyn derivative C8 exhibited increased toxicity at 100 and 150 ⁇ M in comparison to cells treated with the vehicle (DMSO).
  • FIG. 9 graphically illustrates that compound C4 promotes A ⁇ 42 uptake in na ⁇ ve BV2 microglial cells.
  • BV2 cells were treated with DMSO (vehicle) or cromolyn derivatives at different concentrations ranging from 5 to 150 ⁇ M for 3 hours. Then, cells were incubated with soluble untagged A ⁇ 42 and DMSO or cromolyn derivatives for additional 2 hours and collected for ELISA analysis.
  • BV2 microglial cells treated with the cromolyn derivative C4 at 75 ⁇ M exhibited significantly increased A ⁇ 42 uptake levels in comparison to cells treated with the vehicle.
  • FIG. 10 graphically illustrates that compound C4 promotes A ⁇ 42 uptake in microglial BV2-CD33 WT cells.
  • Microglial cells stably expressing CD33 WT were treated with DMSO as control or cromolyn derivatives (C1, C3-8) at different concentrations for 3 hours. Afterwards, cells were incubated with DMSO or cromolyn derivatives in the presence of the A ⁇ 42 peptide for additional 2 hours. Cell lysates were analyzed for intracellular levels of A ⁇ 42 using an A ⁇ 42-specific ELISA kit. Treatment with the cromolyn derivative C4 at 75 ⁇ M led to increased uptake of A ⁇ 42 in BV2-CD33 WT cells in comparison to DMSO treatment and displayed a dose-dependent effect at 50 ⁇ M.
  • FIG. 11 graphically illustrates that compound C4 promotes A ⁇ 42 uptake in BV2-CD33 WT cells.
  • BV2-CD33 WT cells were treated with DMSO (vehicle) or cromolyn derivatives (C1, C2, and C4-7) at different concentrations for 3 hours. Afterwards, cells were treated with DMSO or cromolyn derivatives and soluble A ⁇ 42 peptide for 2 hours. Cell lysates were analyzed using A ⁇ 42-specific ELISA kit and intracellular A ⁇ 42 levels were quantified. The cromolyn derivative C4 effectively induced A ⁇ 42 uptake at 50 and 75 ⁇ M in BV2-CD33 WT cells in comparison to cells treated with DMSO.
  • DMSO vehicle
  • cromolyn derivatives C1, C2, and C4-7
  • the invention encompasses anti-inflammatory compounds to reduce the toxic effect of pro-inflammatory cytokines by converting microglia from a pro-inflammatory M1 state to an M2 state in which the toxic effects are reduced and their phagocytic activity toward amyloidosis, tauopathies and other cytotoxic events is enhanced.
  • the invention also encompasses the use of the compounds to affect therapy early in the disease process.
  • the compounds described herein are the only effective, non-cytokine (e.g. IL-10) compounds exhibiting M1-to-M2 activity.
  • the invention encompasses the compounds and the methods of treating neuron inflammation conditions by administration of a therapeutic effective amount of at least one of the compounds.
  • compounds of the invention include those having the following formula and their analogs and isomers:
  • X may include, but is not limited to, halides, and OCO(C 1 -C 8 alkyls).
  • Alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, and pentyl.
  • Halides include fluoro, chloro, bromo, and iodo.
  • Y may include, but is not limited to, —CH 2 OH, —CH 2 OAc, or —CH 2 OMe.
  • the compounds of the invention include those compounds attached at the 5 position.
  • compounds also include 5-[3-(2-carboxy-4-oxochromen-5-yl)oxy-2-hydroxypropoxy]-4-oxochromene-2-carboxylic acid derivatives and isomeric forms.
  • the invention encompasses methods of treating a variety of neuron inflammation conditions.
  • Neuron inflammation conditions include, but are not limited to, diseases such as ALS, autism spectrum disorder (ASD), ischemic stroke, and prion disease.
  • the compounds may be used to treat ALS including, but not limited to, slowing down or halting the progression of the disease.
  • the compounds may be administered in combination with other anti-inflammatory agents to control the spread of the progressive and fatal effect of ALS.
  • the invention encompasses a combination treatment for ALS of M1, M2 conversion active drugs that control neuroinflammation, such as the drugs in the above formulas, with other immune targeting therapies such as CD4+, siRNA, miRNA that ameliorates ALS, glial morphology modifiers, SOD1 controls, or Riluzole, the only approved drug for ALS.
  • M1, M2 conversion active drugs that control neuroinflammation such as the drugs in the above formulas
  • other immune targeting therapies such as CD4+, siRNA, miRNA that ameliorates ALS, glial morphology modifiers, SOD1 controls, or Riluzole, the only approved drug for ALS.
  • the compounds will slow down or halt neuron damage for neurons located in the brain stem and/or the spinal cord, neurons, or motor neurons that affect voluntary body muscles.
  • the compounds may be administered using known methods for the administration of drugs, for example, IP, IV, transdermally, by inhalation.
  • the invention relates to methods of treating or slowing down the aggressive progression of a neurological disease, such as AD, Ischemic Stroke, ALS, or Prion, and the compound is administered by infusion or intraperitoneal administration.
  • the invention also provides pharmaceutical compositions comprising one or more compounds described herein in association with a pharmaceutically acceptable carrier.
  • these compositions are in unit dosage forms such as tablets, pills, capsules, powders, granules, sterile parenteral solutions or suspensions, metered aerosol or liquid sprays, drops, ampoules, auto-injector devices or suppositories; for oral, parenteral, intranasal, sublingual or rectal administration, or for administration by inhalation or insufflation.
  • the compounds may be incorporated into transdermal patches designed to deliver the appropriate amount of the drug in a continuous fashion.
  • the principal active ingredient is mixed with a pharmaceutically acceptable carrier, e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g. water, to form a solid preformulation composition containing a homogeneous mixture.
  • a pharmaceutically acceptable carrier e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g. water
  • a pharmaceutically acceptable carrier e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical dilu
  • a dry powder composition is micronized for inhalation to the lungs. See for example, U.S. Patent Application publication 2016/0263257, expressly incorporated herein by reference in its entirety, and in particular regarding the dry powder cromolyn formulations described therein.
  • the dry powder composition further comprises at least one excipient.
  • the at least one excipient comprises Lactose monohydrate and/or Magnesium stearate.
  • the compounds may be administered in doses that treat the particular indication.
  • the dose is specifically tailored to lead to blood, brain, and CSF concentrations that allow the drugs to act as M1-to-M2 modifiers.
  • Such doses may include from about 1 mg to about 1000 mg per day.
  • the dosage of the active agents will generally be dependent upon a number of factors, including the pharmacodynamic characteristics of the compound, mode and route of administration of the compound, the health of the patient being treated, the extent of treatment desired, the nature and kind of concurrent therapy, if any, and the frequency of treatment and the nature of the effect desired.
  • dosage ranges of the compound often range from about 0.001 to about 250 mg/kg body weight per day.
  • a dosage may range from about 0.1 to about 25 mg/kg body weight.
  • some variability in this general dosage range may be required depending on the age and weight of the subject being treated, the intended route of administration, the particular agent being administered, and the like.
  • the determination of dosage ranges and optimal dosages for a particular mammal is also well within the ability of one of ordinary skill in the art having the benefit of the instant disclosure.
  • Dosages for compounds may be as low as 5 ng/d.
  • Dosage ranges for active agents may be from 5 ng/d to 100mg/day. In certain embodiments, dosage ranges for active agents may be from about 5 ng/day to about 10 ng/day, about 15 ng/day, about 20 ng/day, about 25 ng/day, about 30 ng/day, about 35 ng/day, about 40 ng/day, about 45 ng/day, about 50 ng/day, about 60 ng/day, about 70 ng/day, about 80 ng/day, about 90 ng/day, about 100 ng/day, about 200 ng/day, about 300 ng/day, about 400 ng/day, about 500 ng/day, about 600 ng/day, about 700 ng/day, about 800 ng/day, or about 900 ng/day.
  • dosage ranges for compounds may be from about 1 ⁇ g/day to about 2 ⁇ g/day, about 3m/day, about 4 ⁇ g/day, about 5 ⁇ g/day, about 10 ⁇ g/day, about 15 ⁇ g/day, about 20 ⁇ g/day, about 30 ⁇ g/day, about 40 ⁇ g/day, about 50 ⁇ g/day, about 60 ⁇ g/day, about 70 ⁇ g/day, about 80 ⁇ g/day, about 90 ⁇ g/day, about 100 ⁇ g/day, about 200 ⁇ g/day, about 300 ⁇ g/day, about 400 ⁇ g/day about 500 ⁇ g/day, about 600 ⁇ g/day, about 700 ⁇ g/day, about 800 ⁇ g/day, or about 900 ⁇ g/day.
  • dosage ranges for active agents may be from about 1mg/day to about 2 mg/day, about 3 mg/day, about 4 mg/day, about 5 mg/day, about 10 mg/day, about 15 mg/day, about 20 mg/day, about 30 mg/day, about 40 mg/day, about 50 mg/day, about 60 mg/day, about 70 mg/day, about 80 mg/day, about 90 mg/day, about 100 mg/day, about 200 mg/day, about 300 mg/day, about 400 mg/day, about 500 mg/day, about 600 mg/day, about 700 mg/day, about 800 mg/day, or about 900 mg/day.
  • the compounds are administered in pM or nM concentrations. In certain embodiments, the compounds are administered in about 1 pM, about 2 pM, about 3 pM, about 4 pM, about 5 pM, about 6 pM, about 7 pM, about 8 pM, about 9 pM, about 10 pM, about 20 pM, about 30 pM, about 40 pM, about 50 pM, about 60 pM, about 70 pM, about 80 pM, about 90 pM, about 100 pM, about 200 pM, about 300 pM, about 400 pM, about 500 pM, about 600 pM, about 700 pM, about 800 pM, about 900 pM, about 1 nM, about 2 nM, about 3 nM, about 4 nM, about 5 nM, about 6 nM, about 7 nM, about 8 nM, about 9 nM, about 10 nM, about 20 pM,
  • the dosage form is a solid dosage form, and the size of the compound in the dosage form is important.
  • the compound is less than about 3 ⁇ m, less than about 2 ⁇ m, or less than about 1 ⁇ m in diameter.
  • the active agent is about 0.1 ⁇ m to about 3.0 ⁇ m in diameter. In certain embodiments, the active agent is from about 0.5 ⁇ m to about 1.5 ⁇ m in diameter.
  • the active agent is about 0.2 ⁇ m, about 0.3 ⁇ m, about 0.4 ⁇ m, about 0.5 ⁇ m, about 0.6 ⁇ m, about 0.7 ⁇ m, about 0.8 ⁇ m, about 0.9 ⁇ m, about 1.0 ⁇ m, about 1.1 ⁇ m, about 1.2 ⁇ m, about 1.3 ⁇ m, about 1.4 ⁇ m, or about 1.5 ⁇ m in diameter.
  • a formulation intended for oral administration to humans may contain from about 0.1 mg to about 5 g of the active agent (or compound) compounded with an appropriate and convenient carrier material varying from about 5% to about 95% of the total composition.
  • Unit dosages will generally contain between about 0.5 mg to about 1500 mg of the active agent.
  • the dosage may be about: 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 21 mg, 22 mg, 23 mg, 24 mg 25 mg, 26 mg, 27 mg, 28 mg, 29 mg, 30 mg, 31 mg, 32 mg, 33 mg, 34 mg 35 mg, 36 mg, 37 mg, 38 mg, 39 mg, 40 mg, 41 mg, 42 mg, 43 mg, 44 mg, 45 mg, 46 mg, 47 mg, 48 mg, 49 mg, 50 mg, 55 mg, 60 mg, 65, mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 800 mg, or 100 mg, etc., up to about 1500 mg of the compound.
  • the invention relates to combination of two active agents.
  • the ratio of the first active agent to the second active agent is about: 200:1, 190:1, 180:1, 170:1, 160:1, 150:1, 140:1, 130:1, 120:1, 110:1, 100:1, 90:1, 80:1, 70:1, 60:1, 50:1, 40:1, 30:1, 20:1, 15:1, 10:1, 9:1, 8:1, 7:1, 6:1, or 5:1. It further may be preferable to have a more equal distribution of pharmaceutical agents.
  • the ratio of the first active agent to the second active agent is about: 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, or 1:4. It may also be advantageous for the pharmaceutical combination to have a relatively large amount of the second component compared to the first component. In certain instances, the ratio of the second active agent to the first active agent is about 200:1, 190:1, 180:1, 170:1, 160:1, 150:1, 140:1, 130:1, 120:1, 110:1, 100:1, 90:1, 80:1, 70:1, 60:1, 50:1, 40:1, 30:1, 20:1, 15:1, 10:1, 9:1, 8:1, 7:1, 6:1, or 5:1.
  • a composition comprising any of the above identified combinations of the first therapeutic agent and second therapeutic agent may be administered in divided doses about 1, 2, 3, 4, 5, 6, or more times per day or in a form that will provide a rate of release effective to attain the desired results.
  • the dosage form may contain both the first and second active agents.
  • the dosage form may be administered one time per day if it contains both the first and second active agents.
  • a formulation intended for oral administration to humans may contain from about 0.1 mg to about 5 g of the first therapeutic agent and about 0.1 to about 5 g of the second therapeutic agent, both of which are compounded with an appropriate and convenient about of carrier material varying from about 5% to about 95% of the total composition.
  • Unit dosages will generally contain between about 0.5 mg to about 1500 mg of the first therapeutic agent and 0.5 mg to 1500 mg of the second therapeutic agent.
  • the dosage may be about: 25 mg, 50 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 800 mg, or 100 mg, etc., up to about 1500 mg of the first therapeutic agent.
  • the dosage may be about: 25 mg, 50 mg, 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 800 mg, or 100 mg, etc., up to about 1500 mg of the second therapeutic agent.
  • the inventions relates to a method of treating a Alzheimer's disease comprising administering by inhalation a micronized, dry powder comprising about 1 mg to 100 mg of Cromolyn Disodium per day to a patient in need thereof.
  • FIG. 1B illustrates representative images of localization of amyloid deposits (6E10) and microglia (Iba1) in mice treated with Cromolyn Sodium (3.15mg/kg) or PBS daily for seven days.
  • FIG. 1C illustrates the effect of Cromolyn Sodium on microglial A ⁇ uptake in vitro. Microglial cells were cultured and incubated with 50 nM of synthetic A ⁇ 40 or A ⁇ 42 and 0, 10 nM, 10 ⁇ M or 1 mM of Cromolyn Sodium for 16 hours. After the incubation, the concentrations of A ⁇ x-40 ( FIG.
  • FIG. 2 illustrates representative plaques of all the plaques and the microglial cells surrounding those deposits in Tg-2576 mice of the study.
  • An image analysis looking at the percentage of Iba-1 positive processes colocalizing with the amyloid staining versus the total amount of Iba-1 signal surrounding the plaque demonstrated that there was more Iba-1/Amyloid colocalization when the mice were treated with Cromolyn Sodium as opposed to any other groups. This result correlates with our results in Example 1 and our in vitro data.
  • BV2 microglial cell cultures were treated with cromolyn and/or ibuprofen (10 ⁇ M, 100 ⁇ M, 1 mM) for 16 hours. Afterwards, cells were incubated with soluble A ⁇ 42 and the compounds for 3 hours. After incubation, cells were collected for ELISA analysis.
  • FIG. 3 graphically illustrates the results of BV2 microglial cells treated with cromolyn, and with cromolyn and ibuprofen exhibit increased A ⁇ 42 uptake levels relative to BV2 microglia treated with the vehicle.
  • Acetic anhydride (0.5 g, 4.6 mmol) was slowly added to a mixture of 5,5′-[(2-hydroxy-1,3-propanediyl)bis(oxy)]bis[4-oxo-4H-1-benzopyran-2-ethanol (0.5 g, 1.14 mmol) in pyridine (20 mL) cooled to 0-5° C. The mixture was stirred for 3 hr at 0-5° C. and then allowed to warm to room temperature. TLC indicted the reaction was complete. Methylene chloride was added and the mixture was washed with 10% HCl until the aqueous phase was acidic. The methylene chloride layer was dried over anhydrous sodium sulfate and solvent was evaporated.
  • Synthetic Aa ⁇ 42 in final 5 uM was incubated with 10, 100, 1,000 nM of test compounds for 1 hour.
  • the aggregation was initiated with heparin at 0.5 mg/ml in final concentration.
  • the assay buffer consisted of 125 mM NaCl, 2.5 mM KCl, 1 mM MgCl 2 , 1.25 mM Na 2 H 2 PO 4 , 2 mM CaCl 2 , 25 mM Glucose, and NaHCO 3 to adjust pH to 7.4.
  • the assay buffer was used as a control.
  • the aggregation was measured by intensity of Thioflavin T binding, which was detected by fluorescent excitation/emission at 450 nm/480 nm (Spectra Max M3 plate reader, Molecular Devices) in a kinetic mode. Aggregation was recorded as the kinetics was calculated as Vmax by the plate reader's software. The assay was performed in triplicate and expressed as standard mean ⁇ SD. Blue dotted line indicate the assay buffer control.
  • FIG. 4 illustrates the results of the assay.
  • FIG. 5 graphically illustrates the results of a one-way of the differences in the A ⁇ levels and the ratios of A ⁇ (42:40).
  • BV2-CD33 WT The effect of compounds in BV2 cells stably expressing full-length human CD33 was assessed to explore whether they reverse CD33-mediated inhibition of A ⁇ uptake (Griciuc et al., 2013 Neuron 78, 631-643).
  • the compound numbers, molecular weight and concentration of the stock solutions are summarized in Table 1.
  • Cromolyn derivatives, C3 and C4 displayed lower solubility in DMSO in comparison to C1, C2, C5, C6, C7 and C8. Therefore, a 25 mM stock solutions for all the compounds except for C3 and C4 were prepared.
  • the stock solutions for C3 and C4 were prepared at 5 mM and 7.5 mM, respectively.
  • C1 is the parent compound—cromolyn disodium.
  • na ⁇ ve BV2 cells were treated with DMSO (control) or cromolyn at 500 ⁇ M for 16 hours. Afterwards, cells were washed with PBS and treated with DMSO or cromolyn in the presence of the fluorescently-tagged A ⁇ 42 peptide (400 nM, red) for 2 hours. At the end of the treatment, the cells were washed and labeled them with a plasma membrane dye (green). Using confocal microscopy and the fluorescence signal in the red channel, the levels of intracellular A ⁇ 42 peptide were quantified. All the quantifications were performed by a blind observer with the ImageJ software. Remarkably, cromolyn sodium led to increased uptake of A ⁇ 42 in na ⁇ ve BV2 microglial cells ( FIG. 6A - FIG. 6D ).
  • cromolyn sodium modulates A ⁇ 42 uptake in na ⁇ ve BV2 microglial cells was determined by using the ELISA assay. Additionally, whether cromolyn sodium leads to increased A ⁇ 42 uptake levels in BV2 cells stably expressing full-length human CD33 (BV2-CD33 WT ) was determined. To this purpose, both na ⁇ ve BV2 and BV2-CD33 WT cell lines were treated with DMSO (control) or cromolyn at different concentrations for 16 hours. Then, the cells were washed with PBS and treated with DMSO or cromolyn and soluble untagged A ⁇ 42 peptide (400 nM) for 2 hours. The collected cell lysates were analyzed for A ⁇ 42 uptake levels using the A ⁇ 42-specific ELISA kit from Wako. The ELISA results were normalized to the protein concentration levels that were previously quantified using the BCA assay.
  • na ⁇ ve BV2 or BV2-CD33 WT cells were plated in proliferating media.
  • cells were treated with DMSO (control) or the compounds at different concentrations in proliferating media for 3 hours.
  • C1, C2, C5, C6, C7 and C8 were tested at 10, 50, 100 and 150 ⁇ M, while C3 and C4 were assessed at 5, 25, 50 and 75 ⁇ M due to solubility limit in DMSO.
  • cells were washed with PBS and treated with DMSO or compounds in the presence of the untagged A ⁇ 42 peptide (400 nM) in DMEM media for 2 hours.
  • na ⁇ ve BV2 microglial cells were incubated with DMSO (vehicle) or cromolyn derivatives at different concentrations for 3 hours. The cells were then washed and incubated with DMSO or compounds and soluble untagged A ⁇ 42 for additional 2 hours. Afterwards, the cell media was collected and measured LDH released by the damaged cells to identify the compounds that induce cytolysis. The LDH assay showed that the cromolyn derivative C8 is the only compound showing toxicity when tested at 100 and 150 ⁇ M ( FIG. 8 ). Therefore, 100 and 150 ⁇ M concentrations for C8 were excluded from the A ⁇ 42 uptake assays.
  • na ⁇ ve BV2 microglial cells were treated with DMSO (control) or cromolyn derivative compounds at different concentrations for 3 hours. Afterwards, the cells were washed and treated with DMSO or compounds in the presence of untagged A ⁇ 42 peptide for 2 hours. At the end of the treatment, the cell lysates were collected. The analysis for intracellular A ⁇ 42 levels is performed using an A ⁇ 42-specific ELISA kit. The parent compound C1 (cromolyn sodium) led to a modest increase of A ⁇ 42 uptake at 100 and 150 ⁇ M in BV2 cells.
  • BV2-CD33 WT cells were treated with DMSO (control) or cromolyn derivatives at different concentrations ranging between 5 and 150 ⁇ M.
  • the cromolyn derivatives C1 and C3-8 were tested.
  • the compound C2 was tested with other cromolyn derivatives in the second set of experiments.
  • Treatment with the compound C4 at 75 ⁇ M resulted in a two-fold increase in A ⁇ 42 uptake in comparison to DMSO treatment and displayed a dose-dependent effect at 50 ⁇ M ( FIG. 10 ).
  • the IC 50 for C4 was 54.7 ⁇ M in BV2-CD33 WT cells.
  • the compound C6 exhibits a dose-dependent effect in mediating inhibition of A ⁇ 42 uptake in BV2-CD33 WT cells when compared to DMSO treatment.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Psychology (AREA)
  • Otolaryngology (AREA)
  • Dermatology (AREA)
  • Pulmonology (AREA)
  • Vascular Medicine (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
US16/328,956 2016-08-31 2017-08-31 Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases Abandoned US20190240194A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/328,956 US20190240194A1 (en) 2016-08-31 2017-08-31 Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201662382192P 2016-08-31 2016-08-31
US16/328,956 US20190240194A1 (en) 2016-08-31 2017-08-31 Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases
PCT/US2017/049702 WO2018045217A1 (en) 2016-08-31 2017-08-31 Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/049702 A-371-Of-International WO2018045217A1 (en) 2016-08-31 2017-08-31 Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/323,514 Continuation US11679095B2 (en) 2016-08-31 2021-05-18 Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases

Publications (1)

Publication Number Publication Date
US20190240194A1 true US20190240194A1 (en) 2019-08-08

Family

ID=61309436

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/328,956 Abandoned US20190240194A1 (en) 2016-08-31 2017-08-31 Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases
US17/323,514 Active 2037-09-15 US11679095B2 (en) 2016-08-31 2021-05-18 Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases
US18/210,992 Pending US20240082207A1 (en) 2016-08-31 2023-06-16 Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases

Family Applications After (2)

Application Number Title Priority Date Filing Date
US17/323,514 Active 2037-09-15 US11679095B2 (en) 2016-08-31 2021-05-18 Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases
US18/210,992 Pending US20240082207A1 (en) 2016-08-31 2023-06-16 Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases

Country Status (8)

Country Link
US (3) US20190240194A1 (zh)
EP (1) EP3506894B1 (zh)
JP (2) JP2019524865A (zh)
KR (1) KR20190044647A (zh)
CN (2) CN116889562A (zh)
AU (1) AU2017321782B2 (zh)
CA (1) CA3033079A1 (zh)
WO (1) WO2018045217A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022178278A1 (en) * 2021-02-19 2022-08-25 The Trustees Of Columbia University In The City Of New York Compositions and methods for modeling human microglia

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9925282B2 (en) 2009-01-29 2018-03-27 The General Hospital Corporation Cromolyn derivatives and related methods of imaging and treatment
CN109846862A (zh) 2012-10-25 2019-06-07 通用医疗公司 治疗阿尔茨海默病及相关疾病的组合疗法
US10525005B2 (en) 2013-05-23 2020-01-07 The General Hospital Corporation Cromolyn compositions and methods thereof
CN106102737B (zh) 2013-10-22 2019-06-14 综合医院公司 色甘酸衍生物以及成像和治疗的相关方法
WO2017091644A1 (en) * 2015-11-23 2017-06-01 Aztherapies, Inc. Compositions and methods for treating ischemic stroke
WO2018045217A1 (en) 2016-08-31 2018-03-08 The General Hospital Corporation Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases
KR20200024854A (ko) * 2017-07-08 2020-03-09 더 제너럴 하스피탈 코포레이션 알츠하이머 질환의 치료를 위한 치료 약물 또는 제제를 확인하기 위한 스크리닝 플랫폼
US10561612B2 (en) 2017-07-20 2020-02-18 The General Hospital Corporation Powdered formulations of cromolyn sodium and ibuprofen
JP2021521122A (ja) * 2018-04-09 2021-08-26 ザ ジェネラル ホスピタル コーポレイション 筋萎縮性側索硬化症および関連障害の治療のための併用療法
WO2020010049A1 (en) * 2018-07-02 2020-01-09 The General Hospital Corporation POWDERED FORMULATIONS OF CROMOLYN SODIUM AND α-LACTOSE
US20220218652A1 (en) * 2018-09-05 2022-07-14 The General Hospital Corporation Methods of treating cytokine release syndrome
JP2022515293A (ja) * 2018-12-10 2022-02-17 ザ ジェネラル ホスピタル コーポレイション クロモリンエステルおよびそれらの使用
CN112375058A (zh) * 2020-11-11 2021-02-19 杭州卢普生物科技有限公司 色甘酸二乙酯和色甘酸钠的制备方法
WO2022146914A1 (en) * 2020-12-28 2022-07-07 The General Hospital Corporation Cromolyn derivatives and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060142241A1 (en) * 2004-11-01 2006-06-29 Yoo Seo H Methods and compositions for reducing neurodegeneration in amyotrophic lateral sclerosis
US10092564B2 (en) * 2016-03-25 2018-10-09 Ab Science Use of masitinib for treatment of an amyotrophic lateral sclerosis patient subpopulation

Family Cites Families (126)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3686412A (en) 1965-03-25 1972-08-22 Colin Fitzmaurice Compositions containing bis-chromonyl compounds for inhibiting antigen-antibody reactions
GB1144905A (en) 1965-03-25 1969-03-12 Fisons Pharmaceuticals Ltd Substituted bis-(2-carboxy-chromonyl-oxy) derivatives and preparation and pharmaceutical compositions thereof
GB1242211A (en) 1967-08-08 1971-08-11 Fisons Pharmaceuticals Ltd Pharmaceutical composition
US3957965A (en) 1967-08-08 1976-05-18 Fisons Limited Sodium chromoglycate inhalation medicament
GB1257162A (zh) 1968-02-16 1971-12-15
US4405598A (en) 1976-01-30 1983-09-20 Fisons, Limited Composition for treating asthma
DE3104294A1 (de) 1981-02-07 1982-08-19 Hoechst Ag, 6000 Frankfurt Substituierte diazaspirodecane, ihre herstellung, ihre verwendung als stabilisatoren fuer organische polymere, sowie die so stabilisierten polymeren
US4429545A (en) 1981-08-03 1984-02-07 Ocean & Atmospheric Science, Inc. Solar heating system
BE897058A (fr) 1982-06-25 1983-12-16 Sandoz Sa Utilisation d'un derive du spiro-succinimide dans le traitement de la demence du type alzheimer
US4996296A (en) 1983-07-27 1991-02-26 Yeda Research & Development Co., Ltd. Cromolyn binding protein in highly purifed form, and methods for the isolation thereof
US4919915A (en) 1987-03-03 1990-04-24 Paul Averback Method for detecting the ability to prevent red-to-green congophilic birefringence
US5231170A (en) 1986-08-27 1993-07-27 Paul Averback Antibodies to dense microspheres
JPH05501505A (ja) 1989-04-28 1993-03-25 ライカー ラボラトリーズ,インコーポレイティド 乾燥粉末吸入装置
US5376386A (en) 1990-01-24 1994-12-27 British Technology Group Limited Aerosol carriers
DE4405387A1 (de) 1994-02-19 1995-08-24 Hoechst Ag Verfahren zur Herstellung von Polyalkyl-1-oxa-diazaspirodecan-Verbindungen
US6168776B1 (en) 1994-07-19 2001-01-02 University Of Pittsburgh Alkyl, alkenyl and alkynyl Chrysamine G derivatives for the antemortem diagnosis of Alzheimer's disease and in vivo imaging and prevention of amyloid deposition
TR199600329A2 (tr) 1995-05-05 1997-03-21 Hoffmann La Roche Seker alkollerin yeni sülfürik asit esterleri.
US6026809A (en) 1996-01-25 2000-02-22 Microdose Technologies, Inc. Inhalation device
US6126919A (en) 1997-02-07 2000-10-03 3M Innovative Properties Company Biocompatible compounds for pharmaceutical drug delivery systems
DE69813853T3 (de) 1997-09-29 2011-05-12 Novartis Ag Perforierte mikroteilchen und deren verwendung
US6309623B1 (en) 1997-09-29 2001-10-30 Inhale Therapeutic Systems, Inc. Stabilized preparations for use in metered dose inhalers
US6565885B1 (en) 1997-09-29 2003-05-20 Inhale Therapeutic Systems, Inc. Methods of spray drying pharmaceutical compositions
US5904937A (en) 1997-10-03 1999-05-18 Fmc Corporation Taste masked pharmaceutical compositions
TWI239847B (en) 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
NZ508663A (en) 1998-06-12 2003-02-28 Microdose Technologies Inc A dry powder inhaler with a vibrator to deaggregate the powder and feedback control for controlling dosing
US6197963B1 (en) 1998-08-13 2001-03-06 The Trustees Of The University Of Pennsylvania Non-peptide peptidomimetics
DK1131059T3 (da) 1998-11-13 2003-06-30 Jago Res Ag Tørpulver til inhalation
EA004936B1 (ru) 1998-12-24 2004-10-28 Янссен Фармацевтика Н.В. Препаративная форма галантамина с регулируемым выделением
JP2001151673A (ja) 1999-09-06 2001-06-05 Nikken Chem Co Ltd 吸入用粉末製剤の製造方法
US20020107173A1 (en) 1999-11-04 2002-08-08 Lawrence Friedhoff Method of treating amyloid beta precursor disorders
US20020009491A1 (en) 2000-02-14 2002-01-24 Rothbard Jonathan B. Compositions and methods for enhancing drug delivery across biological membranes and tissues
HUP0300067A3 (en) 2000-02-21 2010-03-29 Lundbeck & Co As H Novel method for down-regulation of amyloid
AU5702201A (en) 2000-04-13 2001-10-30 Mayo Foundation Abeta<sub>42</sub> lowering agents
US20080021085A1 (en) 2000-04-13 2008-01-24 Mayo Foundation For Medical Education And Research Method of reducing abeta42 and treating diseases
PE20011227A1 (es) 2000-04-17 2002-01-07 Chiesi Farma Spa Formulaciones farmaceuticas para inhaladores de polvo seco en la forma de aglomerados duros
AR035642A1 (es) 2000-05-26 2004-06-23 Pharmacia Corp Uso de una composicion de celecoxib para el alivio rapido del dolor
US20020016359A1 (en) 2000-06-29 2002-02-07 Hellberg Mark R. Compositions and methods of treating neurodegenerative diseases
US8519005B2 (en) 2000-07-27 2013-08-27 Thomas N. Thomas Compositions and methods to prevent toxicity of antiinflammatory agents and enhance their efficacy
FR2815030A1 (fr) 2000-10-05 2002-04-12 Lipha Derives nitroso de la diphenylamine, compositions pharmaceutiques les contenant et leur utilisation pour la preparation de medicaments
US6511960B2 (en) 2001-01-05 2003-01-28 Alphamed Pharmaceuticals Corp Cromolyn for eye and ear infections
ES2274973T3 (es) 2001-04-23 2007-06-01 The Trustees Of The University Of Pennsylvania Inhibidores de la agregacion de placas amiloides y agentes de obtencion de imagenes diagnosticas.
AU2002352974A1 (en) 2001-11-29 2003-06-10 Emisphere Technologies, Inc. Formulations for oral administration of cromolyn sodium
ATE453423T1 (de) 2002-05-10 2010-01-15 Oriel Therapeutics Inc Trockenpulverinhalator mit einer durch piezoelektrisches polymer angetriebenen abgabevorrichtung, und zugehörige blisterverpackung mit einem piezoelektrischen polymermaterial
EP1601380A1 (en) 2003-02-13 2005-12-07 Licentia OY Use of a mast cell activation or degranulation blocking agent in the manufacture of a medicament for the treatment of cerebral ischemia
US8163734B2 (en) * 2003-02-13 2012-04-24 Perttu J. Lindsberg Use of a mast cell activation or degranulation blocking agent in the manufacture of a medicament for the treatment of a patient subjected to thrombolyses
US20040223918A1 (en) 2003-05-07 2004-11-11 Chrysalis Technologies Incorporated Aerosolization of cromolyn sodium using a capillary aerosol generator
GB0321607D0 (en) 2003-09-15 2003-10-15 Vectura Ltd Manufacture of pharmaceutical compositions
JP2007509941A (ja) 2003-10-28 2007-04-19 グラクソ グループ リミテッド ラクトース無水物を用いる吸入医薬製剤およびその投与方法
GB0326632D0 (en) 2003-11-14 2003-12-17 Jagotec Ag Dry powder formulations
DE10355559A1 (de) 2003-11-21 2005-06-23 Orthogen Ag Transskin
US8088935B2 (en) 2003-12-23 2012-01-03 Ironwood Pharmaceuticals, Inc. Compounds and methods for the treatment of asthma
US20070093457A1 (en) 2004-02-11 2007-04-26 Nadir Arber Compositions for treatment of cancer and inflammation with curcumin and at least one nsaid
US20050182044A1 (en) 2004-02-17 2005-08-18 Bruinsma Gosse B. Combinatorial therapy with an acetylcholinesterase inhibitor and (3aR)-1,3a,8-trimethyl-1,2,3,3a,8,8a-hexahydropyrrolo[2,3,-b]indol-5-yl phenylcarbamate
US20070293538A1 (en) 2004-04-13 2007-12-20 Myriad Genetics, Incorporated Pharmaceutical Composition And Methods For Treating Neurodegenerative Disorders
MXPA06012240A (es) 2004-04-23 2007-01-31 Cydex Inc Formulacion para inhalador de polvo seco que contiene eter sulfoalquilico-ciclodextrina.
BRPI0514725A (pt) 2004-08-30 2008-06-24 Seo Hong Yoo efeito neuroprotetor de udca solubilizado no modelo isquêmico focal
GB0426146D0 (en) 2004-11-29 2004-12-29 Bioxell Spa Therapeutic peptides and method
AU2006238812A1 (en) 2005-04-22 2006-11-02 Genentech, Inc. Method for treating dementia or Alzheimer's disease with a CD20 antibody
CA2609980C (en) 2005-05-27 2015-10-13 Queen's University At Kingston Treatment of protein folding disorders
EP1973523A2 (en) 2005-12-15 2008-10-01 Acusphere, Inc. Processes for making particle-based pharmaceutical formulations for pulmonary or nasal administration
US8263645B2 (en) 2006-02-03 2012-09-11 Pari Pharma Gmbh Disodium cromoglycate compositions and methods for administering same
AR059356A1 (es) 2006-02-14 2008-03-26 Astrazeneca Ab Nuevos radioligandos
WO2007102059A1 (en) 2006-03-06 2007-09-13 Pfizer Japan Inc. Sulfonyl benzimidazole derivatives
MX2008011553A (es) 2006-03-09 2008-12-09 Waratah Pharmaceuticals Inc Formulacion de polialcohol de ciclohexano para el tratamiento de transtornos de agregacion de proteinas.
WO2009017467A1 (en) 2007-07-27 2009-02-05 Elan Pharma International Limited Treatment of amyloidogenic diseases
EP2023919A4 (en) 2006-05-08 2010-12-22 Molecular Neuroimaging Llc COMPOUNDS AND AMYLOID PROBES FOR THERAPY AND IMAGING USES
US8445437B2 (en) 2006-07-27 2013-05-21 The Brigham And Women's Hospital, Inc. Treatment and prevention of cardiovascular disease using mast cell stabilizers
US20100173960A1 (en) 2006-09-21 2010-07-08 Antonio Cruz The Combination of a Cyclohexanehexol and a NSAID for the Treatment of Neurodegenerative Diseases
JP2010510254A (ja) 2006-11-24 2010-04-02 ワラタ ファーマシューティカルズ, インコーポレイテッド アルツハイマー病および関連の神経変性疾患のための併用処置
MX2009011127A (es) 2007-04-18 2010-03-10 Janssen Alzheimer Immunotherap Metodo de prevencion y tratamiento de angiopatia amiloide cerebral.
JP5675340B2 (ja) 2007-04-18 2015-02-25 プロビオドルグ エージー 新規阻害剤
US7858803B2 (en) 2007-04-27 2010-12-28 The General Hospital Corporation Imaging tracers for early detection and treatment of amyloid plaques caused by Alzheimer's disease and related disorders
US20090110679A1 (en) 2007-07-13 2009-04-30 Luk-Chiu Li Methods and compositions for pulmonary administration of a TNFa inhibitor
GB0714134D0 (en) 2007-07-19 2007-08-29 Norton Healthcare Ltd Dry-powder medicament
JO3076B1 (ar) 2007-10-17 2017-03-15 Janssen Alzheimer Immunotherap نظم العلاج المناعي المعتمد على حالة apoe
AU2009225407B2 (en) 2008-03-21 2013-09-05 The General Hospital Corporation Compounds and compositions for the detection and treatment of Alzheimer's disease and related disorders
JP5933258B2 (ja) 2008-04-29 2016-06-08 ファーネクストPharnext ゾニサミドおよびアカンプロセートを用いるアルツハイマー病および関連障害の処置のための併用組成物
CN102292328A (zh) 2009-01-22 2011-12-21 拉夸里亚创药株式会社 具有cb2受体激动活性的n-取代饱和杂环砜化合物
US8381454B1 (en) 2009-01-23 2013-02-26 Markus R. Robinson Segmented, elongated, expandable, 4-season, double-walled, low-cost, rigid extruded plastic panel structures
CA2751125C (en) * 2009-01-29 2017-06-20 The General Hospital Corporation Cromolyn derivatives and related methods of imaging and treatment
US9925282B2 (en) 2009-01-29 2018-03-27 The General Hospital Corporation Cromolyn derivatives and related methods of imaging and treatment
EP3235509A1 (en) 2009-03-05 2017-10-25 AbbVie Inc. Il-17 binding proteins
JP2012520311A (ja) 2009-03-11 2012-09-06 プレキシコン インコーポレーテッド Rafキナーゼの阻害のためのピロロ[2,3−b]ピリジン誘導体
CN102395359A (zh) 2009-03-16 2012-03-28 Ipintl有限责任公司 治疗阿尔茨海默病和骨质疏松症以及减缓衰老
EP2248517B1 (en) 2009-05-08 2014-03-26 PARI Pharma GmbH Concentrated mast cell stabilizing pharmaceutical formulations
WO2011017030A2 (en) * 2009-08-06 2011-02-10 Neuraltus Pharmaceuticals, Inc. Treatment of macrophage-related disorders
FI3792254T3 (fi) 2009-09-24 2023-09-22 Wista Lab Ltd Kiteisiä metyylitioniniumkloridihydraatteja
EP2322163A1 (en) 2009-11-03 2011-05-18 Pharnext New therapeutics approaches for treating alzheimer disease
US20110262442A1 (en) 2009-11-06 2011-10-27 Adenios, Inc. Compositions for treating cns disorders
NZ600123A (en) 2009-11-30 2015-04-24 Aptalis Pharmatech Inc Compressible-coated pharmaceutical compositions and tablets and methods of manufacture
EP2377860A1 (en) 2010-04-16 2011-10-19 AC Immune S.A. Novel compounds for the treatment of diseases associated with amyloid or amyloid-like proteins
WO2011136754A1 (en) 2010-04-26 2011-11-03 Mahmut Bilgic A medicament developed for the treatment of respiratory diseases
WO2012021856A1 (en) * 2010-08-12 2012-02-16 Revalesio Corporation Compositions and methods for treatment of taupathy
PL2621494T3 (pl) 2010-09-30 2019-04-30 Chiesi Farm Spa Zastosowanie stearynianu magnezu w preparatach w postaci suchego proszku do inhalacji
US20120121656A1 (en) 2010-11-15 2012-05-17 Revalesio Corporation Methods and compositions for protecting against neurotoxicity of a neurotoxic agent, and improving motor coordination associated with a neurodegenerative condition or disease
CN105853439A (zh) 2011-02-11 2016-08-17 维斯塔实验室有限公司 吩噻嗪二胺鎓盐和其用途
BR112014024287A2 (pt) 2012-03-27 2018-05-08 Duke University composições e métodos para a prevenção e tratamento do derrame vascular induzido por mastócitos
WO2013177594A2 (en) 2012-05-25 2013-11-28 Xlear, Inc. Xylitol-based anti-mucosal compositions and related methods and compositions
CA2883130A1 (en) * 2012-08-31 2014-03-06 The General Hospital Corporation Biotin complexes for treatment and diagnosis of alzheimer's disease
CN104981242A (zh) 2012-10-23 2015-10-14 帝人制药株式会社 肿瘤溶解综合征的治疗药和预防药
US10058530B2 (en) 2012-10-25 2018-08-28 The General Hospital Corporation Combination therapies for the treatment of Alzheimer's disease and related disorders
CN109846862A (zh) * 2012-10-25 2019-06-07 通用医疗公司 治疗阿尔茨海默病及相关疾病的组合疗法
EP2999337A4 (en) * 2013-05-23 2017-03-15 Aztherapies, Inc Methods for delivering cromolyn
US10525005B2 (en) 2013-05-23 2020-01-07 The General Hospital Corporation Cromolyn compositions and methods thereof
CN106102737B (zh) 2013-10-22 2019-06-14 综合医院公司 色甘酸衍生物以及成像和治疗的相关方法
WO2015120392A1 (en) * 2014-02-10 2015-08-13 Patara Pharma, LLC Mast cell stabilizers for lung disease treatment
US9962363B2 (en) 2014-02-10 2018-05-08 Patara Pharma, LLC Mast cell stabilizers treatment for systemic disorders
KR20170137030A (ko) 2014-11-21 2017-12-12 바이오하벤 파마슈티컬 홀딩 컴퍼니 엘티디. 릴루졸의 설하 투여
WO2016196401A1 (en) 2015-05-29 2016-12-08 The Texas A&M University System Antimicrobial and anti-inflammatory compositions
EP3331522A1 (en) 2015-08-07 2018-06-13 Patara Pharma LLC Methods for the treatment of mast cell related disorders with mast cell stabilizers
WO2017027402A1 (en) 2015-08-07 2017-02-16 Patara Pharma, LLC Methods for the treatment of systemic disorders treatable with mast cell stabilizers, including mast cell related disorders
WO2017072335A1 (en) * 2015-10-28 2017-05-04 Ab Science Use of masitinib and other mast cell inhibitors for treatment of parkinson's disease
JP2018534336A (ja) * 2015-11-19 2018-11-22 エーゼットセラピーズ, インコーポレイテッド アルツハイマー病および関連障害を処置するための方法
WO2017091644A1 (en) * 2015-11-23 2017-06-01 Aztherapies, Inc. Compositions and methods for treating ischemic stroke
WO2018045217A1 (en) 2016-08-31 2018-03-08 The General Hospital Corporation Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases
EP3509596A4 (en) 2016-09-08 2020-05-06 Emergo Therapeutics, Inc. MAST CELL STABILIZERS FOR TREATING HYPERCYTOKINEMIA AND VIRUS INFECTIONS
US10561612B2 (en) 2017-07-20 2020-02-18 The General Hospital Corporation Powdered formulations of cromolyn sodium and ibuprofen
CN108164409B (zh) 2018-01-24 2021-06-18 温州医科大学 一种2-亚苄基-1-茚酮类似物及应用
JP2021521122A (ja) 2018-04-09 2021-08-26 ザ ジェネラル ホスピタル コーポレイション 筋萎縮性側索硬化症および関連障害の治療のための併用療法
CN108403708B (zh) 2018-05-22 2019-06-14 滨州医学院 秦皮苷在制备预防或治疗急性呼吸窘迫综合征药物中的应用
WO2020010049A1 (en) 2018-07-02 2020-01-09 The General Hospital Corporation POWDERED FORMULATIONS OF CROMOLYN SODIUM AND α-LACTOSE
US20220218652A1 (en) 2018-09-05 2022-07-14 The General Hospital Corporation Methods of treating cytokine release syndrome
JP2022515293A (ja) 2018-12-10 2022-02-17 ザ ジェネラル ホスピタル コーポレイション クロモリンエステルおよびそれらの使用
JP2023520580A (ja) 2020-04-06 2023-05-17 ザ ジェネラル ホスピタル コーポレイション コロナウイルス誘発炎症状態の処置方法
EP4161524A1 (en) 2020-06-04 2023-04-12 The General Hospital Corporation Methods of treating a coronavirus infection
WO2022146914A1 (en) 2020-12-28 2022-07-07 The General Hospital Corporation Cromolyn derivatives and uses thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060142241A1 (en) * 2004-11-01 2006-06-29 Yoo Seo H Methods and compositions for reducing neurodegeneration in amyotrophic lateral sclerosis
US10092564B2 (en) * 2016-03-25 2018-10-09 Ab Science Use of masitinib for treatment of an amyotrophic lateral sclerosis patient subpopulation

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022178278A1 (en) * 2021-02-19 2022-08-25 The Trustees Of Columbia University In The City Of New York Compositions and methods for modeling human microglia

Also Published As

Publication number Publication date
US20220079914A1 (en) 2022-03-17
JP2022171666A (ja) 2022-11-11
US20240082207A1 (en) 2024-03-14
CN116889562A (zh) 2023-10-17
WO2018045217A8 (en) 2019-03-14
CN109922800B (zh) 2023-06-13
EP3506894A4 (en) 2020-07-29
EP3506894B1 (en) 2023-08-23
EP3506894A1 (en) 2019-07-10
CN109922800A (zh) 2019-06-21
JP2019524865A (ja) 2019-09-05
AU2017321782B2 (en) 2022-03-10
AU2017321782A1 (en) 2019-03-07
CA3033079A1 (en) 2018-03-08
US11679095B2 (en) 2023-06-20
EP3506894C0 (en) 2023-08-23
WO2018045217A1 (en) 2018-03-08
KR20190044647A (ko) 2019-04-30

Similar Documents

Publication Publication Date Title
US11679095B2 (en) Macrophages/microglia in neuro-inflammation associated with neurodegenerative diseases
US11414379B2 (en) SPAK kinase inhibitors as neuroprotective agents
US9475750B2 (en) Phenylketone carboxylate compounds and pharmaceutical uses thereof
JP4788999B2 (ja) 分枝鎖カルボン酸化合物およびその用途
JP2017521494A (ja) プロテオパチーの処置のためのベンジリデングアニジン誘導体の新規な治療的使用
AU2020358970A1 (en) Chinone-, hydrochinome- and naphthochinone-analogues of vatiquione for treatment of mitochondrial disorder diseases
JP2021520413A (ja) 眼科的状態のための療法
JP4354180B2 (ja) 軽度認知障害治療剤
JP7295145B2 (ja) 神経変性疾患を治療するための医薬及びその使用
KR101052345B1 (ko) 뉴로트로핀 관련 질환 치료용 약학 조성물
JP2022529742A (ja) 神経修復方法
JP2021535093A (ja) 緑内障の治療のためのカルパイン−2選択的阻害剤化合物
WO2016191323A1 (en) Lithium co-crystals for treatment of neuropsychiatric disorders
KR20210123321A (ko) 류코트리엔 합성 억제제
JP2021520412A (ja) タンパク質ミスフォールディング疾患のための療法

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE GENERAL HOSPITAL CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ELMALEH, DAVID R.;TANZI, RUDOLPH E.;SHOUP, TIMOTHY M.;AND OTHERS;SIGNING DATES FROM 20171208 TO 20180122;REEL/FRAME:048615/0304

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: 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