US20160331746A1 - Sortilin-Binding Small Molecules for Increasing Glucose Uptake - Google Patents

Sortilin-Binding Small Molecules for Increasing Glucose Uptake Download PDF

Info

Publication number
US20160331746A1
US20160331746A1 US15/151,068 US201615151068A US2016331746A1 US 20160331746 A1 US20160331746 A1 US 20160331746A1 US 201615151068 A US201615151068 A US 201615151068A US 2016331746 A1 US2016331746 A1 US 2016331746A1
Authority
US
United States
Prior art keywords
sortilin
ligand
acid
composition
dioxo
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
US15/151,068
Inventor
Niketa A. Patel
Robert Pleasants Sparks
Wayne Charles Guida
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.)
University of South Florida
US Department of Veterans Affairs VA
Original Assignee
University of South Florida
US Department of Veterans Affairs VA
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 University of South Florida, US Department of Veterans Affairs VA filed Critical University of South Florida
Priority to US15/151,068 priority Critical patent/US20160331746A1/en
Assigned to UNIVERSITY OF SOUTH FLORIDA reassignment UNIVERSITY OF SOUTH FLORIDA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SPARKS, ROBERT PLEASANTS, GUIDA, WAYNE CHARLES
Assigned to UNIVERSITY OF SOUTH FLORIDA, THE UNITED STATES GOVERNMENT AS REPRESENTED BY THE DEPARTMENT OF VETERANS AFFAIRS reassignment UNIVERSITY OF SOUTH FLORIDA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PATEL, NIKETA A.
Publication of US20160331746A1 publication Critical patent/US20160331746A1/en
Priority to US16/022,803 priority patent/US10849896B2/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
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/498Pyrazines or piperazines ortho- and peri-condensed with carbocyclic ring systems, e.g. quinoxaline, phenazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/197Carboxylic acids, e.g. valproic acid having an amino group the amino and the carboxyl groups being attached to the same acyclic carbon chain, e.g. gamma-aminobutyric acid [GABA], beta-alanine, epsilon-aminocaproic acid, pantothenic acid
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • C40B30/02
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16CCOMPUTATIONAL CHEMISTRY; CHEMOINFORMATICS; COMPUTATIONAL MATERIALS SCIENCE
    • G16C20/00Chemoinformatics, i.e. ICT specially adapted for the handling of physicochemical or structural data of chemical particles, elements, compounds or mixtures
    • G16C20/60In silico combinatorial chemistry
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16CCOMPUTATIONAL CHEMISTRY; CHEMOINFORMATICS; COMPUTATIONAL MATERIALS SCIENCE
    • G16C20/00Chemoinformatics, i.e. ICT specially adapted for the handling of physicochemical or structural data of chemical particles, elements, compounds or mixtures
    • G16C20/60In silico combinatorial chemistry
    • G16C20/64Screening of libraries
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/4035Isoindoles, e.g. phthalimide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B35/00ICT specially adapted for in silico combinatorial libraries of nucleic acids, proteins or peptides

Definitions

  • Sortilin which is generated from sortilin WT transcript, is highly conserved across species. It has an endoplasmic reticulum (ER) import signal, a vacuolar protein sorting 10 (VSP10) luminal domain, a transmembrane domain and a cytosolic domain. Sortilin ligands bind to the VSP10 domain while the cytosolic domain contains the active sorting signal that directs sortilin and its ligands to lysosomes. Protease cleavage within the extracellular stalk of sortilin releases the sortilin ectodomain by an event called shedding.
  • ER endoplasmic reticulum
  • VSP10 vacuolar protein sorting 10
  • Sortilin is a major component of the Glut4 containing vesicles in adipocytes and is located in the low density microsomes along with Glut4 in the adipocytes. It plays an essential role in the development of the insulin responsive transport system in cultured adipocytes and muscle cells.
  • Glut4 is often times mis-localized in diabetic patients, and more than 80% of diabetic patients are obese or overweight. It has been shown that decreasing sortilin inhibits insulin-induced glucose transport and biogenesis of insulin-responsive compartment. Morbidly obese mice have decreased sortilin which contributes to defects in glucose transport.
  • FIG. 1 shows a bar graph of the qPCR quantification of sortilin WT and 17b from non-diabetic (lean) and diabetic (obese) patients.
  • FIG. 2A shows a photograph of a silver stained Western blot analysis plate with antibodies as indicated
  • FIG. 2B shows a photograph of a silver stained PCR plate for sortilin WT and 17b.
  • FIG. 3 shows the structure of the scaffold 1 exemplary compound, 2-methyl-3,5-dioxo-4-azatricyclo[5.2.1.0(2,6)]dec-8-en-4-yl)acetic acid, and a Schrödinger Glide graphical representation of its binding to sortilin, according to an embodiment of the invention.
  • FIG. 4 shows the structure of the scaffold 1 exemplary compound, methyl 2-(1,3-dioxo-1,3,3a,4,7,7a-hexahydro-2H-4,7-methanoisoindol-2-yl)propanoate, and a Schrödinger Glide graphical representation of its binding to sortilin, according to an embodiment of the invention.
  • FIG. 5 shows the structure of the scaffold 1 exemplary compound, 2-(1,3-dioxo-1,3,3a,4,7,7a-hexahydro-2H-4,7-methanoisoindol-2-yl)-4-(methylthio)butanoic acid, and a Schrödinger Glide graphical representation of its binding to sortilin, according to an embodiment of the invention.
  • FIG. 6 shows the structure of the scaffold 1 exemplary compound, 2-(3,5-dioxo-4-azatricyclo[5.2.1.0(2,6)]dec-8-en-4-yl)-4-methylpentanoic acid, and a Schrödinger Glide graphical representation of its binding to sortilin, according to an embodiment of the invention.
  • FIG. 7 shows the structure of the scaffold 2 exemplary compound, 4-[(3,4-dichlorophenyl)amino]-3-(3-methylbenzyl)-4-oxobutanoic acid, and a Schrödinger Glide graphical representation of its binding to sortilin, according to an embodiment of the invention.
  • FIG. 8 shows the structure of the scaffold 2 exemplary compound, 3-benzyl-4-[(3-chloro-2-methylphenyl)amino]-4-oxobutanoic acid, and a Schrödinger Glide graphical representation of its binding to sortilin, according to an embodiment of the invention.
  • FIG. 9 shows the structure of the scaffold 3 exemplary compound, (3-oxo-1,2,3,4-tetrahydro-2-quinoxalinyl)acetic acid, and a Schrödinger Glide graphical representation of its binding to sortilin, according to an embodiment of the invention.
  • FIG. 10 shows various species of Scaffolds 1-3, according to embodiments of the invention.
  • FIG. 11 shows photographs of FIG. 11A silver stained plates for Western blot analysis with antibodies and FIG. 11B silver stained plates for PCR for sortilin WT and 17b where C is Control, 1 is 2-(3,5-dioxo-4-azatricyclo[5.2.1.0(2,6)]dec-8-en-4-yl)-4-methylpentanoic acid, and 2 is 4-[(3,4-dichlorophenyl)amino]-3-(3-methylbenzyl)-4-oxobutanoic acid.
  • FIG. 12 shows a bar graph of glucose uptake when 2-(3,5-dioxo-4-azatricyclo[5.2.1.0(2,6)]dec-8-en-4-yl)-4-methylpentanoic acid (2) or 4-[(3,4-dichlorophenyl)amino]-3-(3-methylbenzyl)-4-oxobutanoic acid (3), according to embodiments of the invention were added to 3T3L1 cells for 24 h measured four times, where p ⁇ 0.001 and highly significant by a two-tailed Student's t-test.
  • Embodiments of the invention are directed to compounds with the ability to dock at a site within sortilin to promote the increase of glucose uptake.
  • the compounds are identified by their ability to suitably bind to a region of the sortilin protein with high affinity and thereby mediate the interaction of sortilin with various protein molecules or other natural substrates.
  • Compounds of three scaffolds, according to embodiments of the invention bind to sortilin both in vivo and in silico.
  • Sortilin is alternatively spliced, to generate two splice variants in humans.
  • Exon 17b is a pseudoexon, which is absent in sortilin WT. Inclusion of this exon via alternative splicing generates a transcript with an extra coding exon between VSP10 and transmembrane domains.
  • downregulation of TDP-43 as seen in frontotemporal lobal degeneration causes an increase in splice variant Sortilin 17b. It appears that splice variant sortilin 17b can bind to the sortilin ligand but is unable to efficiently mediate endocytosis. The expression of splice variants and their significance in adipocytes is not known.
  • sortilin The biological significance of sortilin and its role in glucose uptake is apparent by evaluating obese and lean patient samples. Primers to decipher the presence of alternatively spliced sortilin variants were designed, and experiments were carried out to analyze identified compounds for their ability to increase glucose uptake and their effect on sortilin expression on RNA and protein levels.
  • FIG. 1 shows the results of qPCR, which indicate that in lean nondiabetic individuals, sortilin WT is the predominantly expressed variant.
  • the quantity of sortilin WT is decreased 8-fold in obese diabetic humans and the splice variant sortilin 17b is increased 2-fold.
  • sortilin in mouse 3T3L1 adipocytes serves as an in vitro model for studying human adipogenesis and mechanisms underlying obesity, as further human and mouse sortilin have >90% homology.
  • the 3T3L1 murine preadipocyte cell line is widely used, as it authentically reproduces adipogenesis including expression of adipogenic genes and morphological changes.
  • Studies of the expression of sortilin splice variants during adipogenesis indicate that both variants (WT and 17b) are expressed during adipogenesis.
  • the level of sortilin WT increases as adipocytes mature, from day 6 onward, while sortilin 17b levels decline in mature adipocytes.
  • FIG. 2A shows western blot analysis, where sortilin antibody does not distinguish between variants; PPAR ⁇ and adiponectin are markers of adipogenesis.
  • FIG. 2B shows PCR using primers that simultaneously amplify sortilin WT and sortilin 17b. Clearly sortilin WT increases by day 6 while sortilin 17b decreases upon adipocyte maturation.
  • a docking scheme was constructed and used to conduct virtual screens of sortilin for small molecules that bind to sortilin in a correct orientation. Identification of small molecules suitable for testing as potential ligands for sortilin, in silico screening of potential compounds was carried out using Schrödinger software. The screening proceeded by constructing a suitable structure of sortilin from the protein data bank with structure 3F6K using the Schrödinger Protein Prep Wizard. Ligands were assembled from the NCI Diversity set IV and from Chembridge's commercially procurable databases. Potential ligands and, subsequently, suitable analogues were found via the Chembridge compound libraries. The virtual ligands were procured and prepared using Schrödinger Ligprep.
  • Resulting compounds are scrutinized and suitable compounds are selected for testing.
  • Software is employed to generate analogues of these ligands and prepared virtual structures for docking.
  • Target ligands are subsequently procured from commercial sources, when possible, and tested for various activities in adipocytes, including adipocyte differentiation and glucose uptake effects by the presence of these molecules.
  • Small molecule compounds for the stimulation of increased glucose uptake are of three identified molecular scaffolds.
  • the three molecular scaffolds were identified for testing due to suitable log p to permeate the cell membrane and reach sortilin within the cell.
  • Species of the first scaffold are norbornene anhydride amino acid adducts (including the monocarbonyl reduced equivalent norbornene amide carboxylates) and esters thereof, as shown below, and represented by the compounds: 2-methyl-3,5-dioxo-4-azatricyclo[5.2.1.0(2,6)]dec-8-en-4-yl)acetic acid; methyl 2-(1,3-dioxo-1,3,3a,4,7,7a-hexahydro-2H-4,7-methanoisoindol-2-yl)propanoate; 2-(1,3-dioxo-1,3,3a,4,7,7a-hexahydro-2H-4,7-methanoisoindol-2-yl)-4-(methylthio)butanoic acid; and 2-(3,5-dioxo-4-azatricyclo[5.2.1.0(2,6)]dec-8-en-4-yl)-4-methylpentanoic acid
  • R is a carboxylic acid or ester thereof and where any of the carbons of the norbornene ring can be substituted with a C 1 to C6 alkyl group or the carboxylic acid can be condensed as a C 1 to C 5 ester.
  • the second scaffolds are N-phenyl-amide-acids of benzyl substituted glutaric acid as shown below, and represented by the two compounds: 4-[(3,4-dichlorophenyl)amino]-3-(3-methylbenzyl)-4-oxobutanoic acid; and 3-benzyl-4-[(3-chloro-2-methylphenyl)amino]-4-oxobutanoic acid.
  • X is independently C 1 to C 5 alkyl, acyl, amino, sulfono, chloro, bromo, iodo, or flouro and R can be C 1 to C 5 alkyl, or the acid can be replaced with a tetrazol.
  • the methylene units can independently be lengthened to ethylene or propylene units.
  • the third scaffold is 2 substituted 3-oxo-1,2,3,4-tetrahydro-2-quinoxalines, shown below and represented by the compound (3-oxo-1,2,3,4-tetrahydro-2-quinoxalinyl)acetic acid.
  • x is C 1 to C 5 alkyl, acyl, amino, sulfono, chloro, bromo, iodo, or flouro and where the carboxylic acid may be separated from the ring by one to three carbons which may be substituted with a C 1 to C 5 alkyl.
  • the acid may be converted to a C 1 to C 5 ester, or replaced with a tetrazole.
  • FIGS. 3-9 The structures of potential ligands from the three scaffolds and the graphical binding to the active site of a sortilin are shown in FIGS. 3-9 .
  • compositions and dosage formulations involve one or more of these species from any of the three scaffolds combined with other vehicles for administration, such as, solvents, buffering agents, transporters, salts, binders, fillers, disintegrants, lubricants, encapsulates, emulsifiers, suspending agents, penetration enhancers, flavoring agents, preservatives, coloring agents, propellants, and/or one or more additional therapeutic agents.
  • vehicles for administration such as, solvents, buffering agents, transporters, salts, binders, fillers, disintegrants, lubricants, encapsulates, emulsifiers, suspending agents, penetration enhancers, flavoring agents, preservatives, coloring agents, propellants, and/or one or more additional therapeutic agents.
  • these components are combined with the sortilin ligand, for a desired mode of delivery, or for an improvement of therapeutic effectiveness of these ligands, by improving absorption, distribution, metabolism, and excretion (ADME) characteristics and/or improving the timing of therapeutics
  • composition can be administered intravenously, orally, rectally, sublingually, sublabially, epidurally, intracerebrally, intracerebroventrically, topically, nasally, intervitrally, subcutaneously, transdermally, by inhalation, or in any other manner of administration.
  • dosage forms include, but are not limited to: tablets; caplets; capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms; pastes; powders; dressings; creams; plasters; solutions; patches; aerosols; gels; aqueous or non-aqueous liquid suspensions; oil-in-water or water-in-oil liquid emulsions; solutions; and elixirs.
  • Solvents for liquid solutions or suspensions include, but are not limited to, one or more of water, glycols, oils, ethanol or other alcohols.
  • Excipients for solid forms include, but are not limited to, one or more of starches, sugars, and micro-crystalline cellulose.
  • Pharmaceutically acceptable additives include: suspending agents, such as, but not limited to, one or more of sorbitol syrup, cellulose derivatives and hydrogenated edible fats.
  • Emulsifying agents such as, but not limited to, one or more of lecithin, acacia, almond oil, oily esters, ethyl alcohol, fractionated vegetable oils.
  • Preservatives include, but not limited to, one or more of methyl-p-hydroxybenzoate, propyl-p-hydroxybenzoate, and sorbic acid.
  • Binders include, but are not limited to, one or more of corn starch, potato starch, or other starches, gelatin, acacia, sodium alginate, alginic acid, powdered tragacanth, guar gum, cellulose, ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose, polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, and microcrystalline cellulose.
  • Fillers include, but are not limited to, one or more of talc, calcium carbonate, microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, and pre-gelatinized starch.
  • Disintegrants include, but are not limited to, one or more of agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato starch, tapioca starch, pre-gelatinized starch, and clays.
  • Lubricants include, but are not limited to, one or more calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, stearic acid, sodium lauryl sulfate, talc, hydrogenated peanut oil, hydrogenated cottonseed oil, hydrogenated sunflower oil, hydrogenated sesame oil, olive oil, hydrogenated corn oil, hydrogenated soybean oil, zinc stearate, ethyl oleate, ethyl laureate, agar, syloid silica gel, coagulated aerosol of synthetic silica, and pyrogenic silicon dioxide.
  • Penetration enhancers include, but are not limited to, one or more of acetone, ethanol, dimethyl sulfoxide, dimethyl acetamide, dimethyl formamide, polyethylene glycol, polyvinylpyrrolidone, urea, Tween 80, and Span 60.
  • the pH of a pharmaceutical compositions, dosage forms, or the tissue to which the pharmaceutical composition or dosage form is applied, can be adjusted to improve delivery of one or more active ingredients.
  • Propellants include, but are not limited to, dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, tetrafluoroethane, hexafluoropropane, and carbon dioxide.

Abstract

Various scaffolds of small molecules capable of binding to the active site of sortilin are identified by in silico methods. These scaffolds include norbornene anhydride amino acid adducts, phenyl-amide-acids of benzyl substituted glutaric acids, and 2-substituted 3-oxo-1,2,3,4-tetrahydro-2-quinoxalines. These sortilin ligands increase the uptake of glucose in 3T3L1 cells and can be employed in compositions to increase uptake of glucose for the treatment of diabetic patents.

Description

    CROSS-REFERENCE TO A RELATED APPLICATION
  • This application claims the benefit of U.S. Provisional Application Ser. No. 62/160,220, filed May 12, 2015, the disclosure of which is hereby incorporated by reference in its entirety, including all figures, tables and drawings.
  • This invention was made with government support under VAMR 821-MR-EN-20606 awarded by Veteran Affairs. The government has certain rights in the invention.
  • BACKGROUND OF INVENTION
  • Sortilin, which is generated from sortilin WT transcript, is highly conserved across species. It has an endoplasmic reticulum (ER) import signal, a vacuolar protein sorting 10 (VSP10) luminal domain, a transmembrane domain and a cytosolic domain. Sortilin ligands bind to the VSP10 domain while the cytosolic domain contains the active sorting signal that directs sortilin and its ligands to lysosomes. Protease cleavage within the extracellular stalk of sortilin releases the sortilin ectodomain by an event called shedding. Sortilin is a major component of the Glut4 containing vesicles in adipocytes and is located in the low density microsomes along with Glut4 in the adipocytes. It plays an essential role in the development of the insulin responsive transport system in cultured adipocytes and muscle cells.
  • Regulation of blood glucose levels in mammals is achieved by translocation of fat and muscle specific transporter-Glut4 to the plasma membrane. Insulin resistance, inability for glucose transport by insulin, is a key hallmark of type 2 diabetes mellitus. Glut4 is often times mis-localized in diabetic patients, and more than 80% of diabetic patients are obese or overweight. It has been shown that decreasing sortilin inhibits insulin-induced glucose transport and biogenesis of insulin-responsive compartment. Morbidly obese mice have decreased sortilin which contributes to defects in glucose transport.
  • Discovering that sortilin levels are decreased in diabetic patients, the identification and demonstration of compounds that can stabilize sortilin and increase glucose transport in adipocytes allows the discovery of effective therapeutics for correcting defects in glucose transport and addressing obesity in diabetic patents.
  • BRIEF DESCRIPTION OF DRAWINGS
  • FIG. 1 shows a bar graph of the qPCR quantification of sortilin WT and 17b from non-diabetic (lean) and diabetic (obese) patients.
  • FIG. 2A shows a photograph of a silver stained Western blot analysis plate with antibodies as indicated, and FIG. 2B shows a photograph of a silver stained PCR plate for sortilin WT and 17b.
  • FIG. 3 shows the structure of the scaffold 1 exemplary compound, 2-methyl-3,5-dioxo-4-azatricyclo[5.2.1.0(2,6)]dec-8-en-4-yl)acetic acid, and a Schrödinger Glide graphical representation of its binding to sortilin, according to an embodiment of the invention.
  • FIG. 4 shows the structure of the scaffold 1 exemplary compound, methyl 2-(1,3-dioxo-1,3,3a,4,7,7a-hexahydro-2H-4,7-methanoisoindol-2-yl)propanoate, and a Schrödinger Glide graphical representation of its binding to sortilin, according to an embodiment of the invention.
  • FIG. 5 shows the structure of the scaffold 1 exemplary compound, 2-(1,3-dioxo-1,3,3a,4,7,7a-hexahydro-2H-4,7-methanoisoindol-2-yl)-4-(methylthio)butanoic acid, and a Schrödinger Glide graphical representation of its binding to sortilin, according to an embodiment of the invention.
  • FIG. 6 shows the structure of the scaffold 1 exemplary compound, 2-(3,5-dioxo-4-azatricyclo[5.2.1.0(2,6)]dec-8-en-4-yl)-4-methylpentanoic acid, and a Schrödinger Glide graphical representation of its binding to sortilin, according to an embodiment of the invention.
  • FIG. 7 shows the structure of the scaffold 2 exemplary compound, 4-[(3,4-dichlorophenyl)amino]-3-(3-methylbenzyl)-4-oxobutanoic acid, and a Schrödinger Glide graphical representation of its binding to sortilin, according to an embodiment of the invention.
  • FIG. 8 shows the structure of the scaffold 2 exemplary compound, 3-benzyl-4-[(3-chloro-2-methylphenyl)amino]-4-oxobutanoic acid, and a Schrödinger Glide graphical representation of its binding to sortilin, according to an embodiment of the invention.
  • FIG. 9 shows the structure of the scaffold 3 exemplary compound, (3-oxo-1,2,3,4-tetrahydro-2-quinoxalinyl)acetic acid, and a Schrödinger Glide graphical representation of its binding to sortilin, according to an embodiment of the invention.
  • FIG. 10 shows various species of Scaffolds 1-3, according to embodiments of the invention.
  • FIG. 11 shows photographs of FIG. 11A silver stained plates for Western blot analysis with antibodies and FIG. 11B silver stained plates for PCR for sortilin WT and 17b where C is Control, 1 is 2-(3,5-dioxo-4-azatricyclo[5.2.1.0(2,6)]dec-8-en-4-yl)-4-methylpentanoic acid, and 2 is 4-[(3,4-dichlorophenyl)amino]-3-(3-methylbenzyl)-4-oxobutanoic acid.
  • FIG. 12 shows a bar graph of glucose uptake when 2-(3,5-dioxo-4-azatricyclo[5.2.1.0(2,6)]dec-8-en-4-yl)-4-methylpentanoic acid (2) or 4-[(3,4-dichlorophenyl)amino]-3-(3-methylbenzyl)-4-oxobutanoic acid (3), according to embodiments of the invention were added to 3T3L1 cells for 24 h measured four times, where p<0.001 and highly significant by a two-tailed Student's t-test.
  • DETAILED DISCLOSURE
  • Embodiments of the invention are directed to compounds with the ability to dock at a site within sortilin to promote the increase of glucose uptake. The compounds are identified by their ability to suitably bind to a region of the sortilin protein with high affinity and thereby mediate the interaction of sortilin with various protein molecules or other natural substrates. Compounds of three scaffolds, according to embodiments of the invention, bind to sortilin both in vivo and in silico.
  • Sortilin is alternatively spliced, to generate two splice variants in humans. Exon 17b, is a pseudoexon, which is absent in sortilin WT. Inclusion of this exon via alternative splicing generates a transcript with an extra coding exon between VSP10 and transmembrane domains. In neurons, downregulation of TDP-43 as seen in frontotemporal lobal degeneration, causes an increase in splice variant Sortilin 17b. It appears that splice variant sortilin 17b can bind to the sortilin ligand but is unable to efficiently mediate endocytosis. The expression of splice variants and their significance in adipocytes is not known.
  • The biological significance of sortilin and its role in glucose uptake is apparent by evaluating obese and lean patient samples. Primers to decipher the presence of alternatively spliced sortilin variants were designed, and experiments were carried out to analyze identified compounds for their ability to increase glucose uptake and their effect on sortilin expression on RNA and protein levels.
  • As obese diabetic humans have decreased sortilin expression, adipose tissue from four obese diabetic individuals and two non-diabetic individuals were obtained. RNA was isolated and real time qPCR was performed for sortilin WT and sortilin 17b. FIG. 1 shows the results of qPCR, which indicate that in lean nondiabetic individuals, sortilin WT is the predominantly expressed variant. The quantity of sortilin WT is decreased 8-fold in obese diabetic humans and the splice variant sortilin 17b is increased 2-fold.
  • Expression of sortilin in mouse 3T3L1 adipocytes serves as an in vitro model for studying human adipogenesis and mechanisms underlying obesity, as further human and mouse sortilin have >90% homology. The 3T3L1 murine preadipocyte cell line is widely used, as it authentically reproduces adipogenesis including expression of adipogenic genes and morphological changes. Studies of the expression of sortilin splice variants during adipogenesis indicate that both variants (WT and 17b) are expressed during adipogenesis. The level of sortilin WT increases as adipocytes mature, from day 6 onward, while sortilin 17b levels decline in mature adipocytes. FIG. 2A shows western blot analysis, where sortilin antibody does not distinguish between variants; PPARγ and adiponectin are markers of adipogenesis. FIG. 2B shows PCR using primers that simultaneously amplify sortilin WT and sortilin 17b. Clearly sortilin WT increases by day 6 while sortilin 17b decreases upon adipocyte maturation.
  • A docking scheme was constructed and used to conduct virtual screens of sortilin for small molecules that bind to sortilin in a correct orientation. Identification of small molecules suitable for testing as potential ligands for sortilin, in silico screening of potential compounds was carried out using Schrödinger software. The screening proceeded by constructing a suitable structure of sortilin from the protein data bank with structure 3F6K using the Schrödinger Protein Prep Wizard. Ligands were assembled from the NCI Diversity set IV and from Chembridge's commercially procurable databases. Potential ligands and, subsequently, suitable analogues were found via the Chembridge compound libraries. The virtual ligands were procured and prepared using Schrödinger Ligprep. Resulting compounds are scrutinized and suitable compounds are selected for testing. Software is employed to generate analogues of these ligands and prepared virtual structures for docking. Target ligands are subsequently procured from commercial sources, when possible, and tested for various activities in adipocytes, including adipocyte differentiation and glucose uptake effects by the presence of these molecules.
  • Specifically, virtual compounds were docked into the active site and scoring was biased to electrostatic interactions, particularly hydrogen bonding, with sortilin at amino acid residues: Serine 272, Arginine 292, Phenylalanine 273, Serine 283 and Tyrosine 318. Docking was performed using Schrödinger Glide on settings HTVS, SP, and XP. Further docking was refined using the Schrödinger Quantum Polarized Ligand Docking (QPLD). After this enrichment and refinement of results for selection of ligands, compounds were tested in vivo. Preliminary targets were selected from the Chembridge library or other databases of compounds, based on various ADME properties and in regard to an overall docking score from the various virtual screenings performed.
  • Small molecule compounds for the stimulation of increased glucose uptake, according to embodiments of the invention, are of three identified molecular scaffolds. The three molecular scaffolds were identified for testing due to suitable log p to permeate the cell membrane and reach sortilin within the cell. Species of the first scaffold are norbornene anhydride amino acid adducts (including the monocarbonyl reduced equivalent norbornene amide carboxylates) and esters thereof, as shown below, and represented by the compounds: 2-methyl-3,5-dioxo-4-azatricyclo[5.2.1.0(2,6)]dec-8-en-4-yl)acetic acid; methyl 2-(1,3-dioxo-1,3,3a,4,7,7a-hexahydro-2H-4,7-methanoisoindol-2-yl)propanoate; 2-(1,3-dioxo-1,3,3a,4,7,7a-hexahydro-2H-4,7-methanoisoindol-2-yl)-4-(methylthio)butanoic acid; and 2-(3,5-dioxo-4-azatricyclo[5.2.1.0(2,6)]dec-8-en-4-yl)-4-methylpentanoic acid.
  • Figure US20160331746A1-20161117-C00001
  • where R is a carboxylic acid or ester thereof and where any of the carbons of the norbornene ring can be substituted with a C1 to C6 alkyl group or the carboxylic acid can be condensed as a C1 to C5 ester.
  • The second scaffolds are N-phenyl-amide-acids of benzyl substituted glutaric acid as shown below, and represented by the two compounds: 4-[(3,4-dichlorophenyl)amino]-3-(3-methylbenzyl)-4-oxobutanoic acid; and 3-benzyl-4-[(3-chloro-2-methylphenyl)amino]-4-oxobutanoic acid.
  • Figure US20160331746A1-20161117-C00002
  • where X is independently C1 to C5 alkyl, acyl, amino, sulfono, chloro, bromo, iodo, or flouro and R can be C1 to C5 alkyl, or the acid can be replaced with a tetrazol. The methylene units can independently be lengthened to ethylene or propylene units.
  • The third scaffold is 2 substituted 3-oxo-1,2,3,4-tetrahydro-2-quinoxalines, shown below and represented by the compound (3-oxo-1,2,3,4-tetrahydro-2-quinoxalinyl)acetic acid.
  • Figure US20160331746A1-20161117-C00003
  • where x is C1 to C5 alkyl, acyl, amino, sulfono, chloro, bromo, iodo, or flouro and where the carboxylic acid may be separated from the ring by one to three carbons which may be substituted with a C1 to C5 alkyl. The acid may be converted to a C1 to C5 ester, or replaced with a tetrazole.
  • The structures of potential ligands from the three scaffolds and the graphical binding to the active site of a sortilin are shown in FIGS. 3-9. Other species of the three scaffolds, according to embodiments of the invention, are given in Table 1, below, where the title can be matched with the chemical structure of FIG. 10.
  • TABLE 1
    Species of the Three Scaffolds, where structures are given in FIG. 10
    RMS
    Entry Derivative- Glide Dockin Glide Glide Glide
    Title ID OPLS-2005 lignum g score gscore lipo hbond
    ligand_26 1928 0.039 26 −9.994 −9.994 −2.382 −0.72
    ligand_1682 1929 0.034 296 −9.803 −9.803 −2.044 −0.729
    ligand_61963 1930 0.028 299 −9.781 −9.781 −2.832 −1.04
    ligand_28 1931 0.019 28 −9.71 −9.71 −2.027 −0.86
    ligand_13 1932 0.006 13 −9.586 −9.586 −2.785 −1.025
    ligand_32 1933 0.023 32 −9.573 −9.573 −2.026 −0.742
    ligand_29 1934 0.002 29 −9.569 −9.569 −2.019 −0.743
    ligand_9 1935 0.015 9 −9.516 −9.516 −2.552 −1.034
    ligand_41 1936 0.007 41 −9.46 −9.46 −2.083 −0.707
    ligand_38 1937 0 38 −9.352 −9.352 −2.093 −0.78
    ligand_78 1938 0.038 78 −9.329 −9.329 −2.008 −0.982
    ligand_19 1939 0.002 19 −9.32 −9.32 −2.431 −1.025
    ligand_79 1940 0.001 79 −9.307 −9.307 −1.991 −1.009
    ligand_46 1941 0.005 46 −9.275 −9.275 −2.031 −0.755
    ligand_218 1942 0.005 218 −9.166 −9.166 −2.023 −0.717
    ligand_33 1943 0.023 33 −9.134 −9.134 −1.918 −0.745
    ligand_10 1944 0.049 10 −9.002 −9.002 −2.485 −0.954
    ligand_1682 1945 0.012 293 −8.916 −8.916 −1.698 −0.753
    ligand_61963 1946 0.047 300 −8.877 −8.877 −2.738 −0.709
    ligand_20 1947 0.038 20 −8.81 −8.81 −2.552 −0.94
    ligand_36 1948 0.001 36 −8.678 −8.678 −1.719 −0.755
    ligand_155 1949 0.003 155 −8.49 −8.49 −0.721 −1.082
    ligand_154 1950 0.013 154 −8.484 −8.484 −0.719 −1.086
    ligand_24 1951 0.049 24 −8.477 −8.477 −2.646 −0.823
    ligand_5892 1952 0.014 298 −8.447 −8.447 −0.739 −0.999
    ligand_166 1953 0.023 166 −8.44 −8.44 −1.055 −0.733
    ligand_50 1954 0.029 50 −8.434 −8.434 −1.742 −0.754
    ligand_167 1955 0.01 167 −8.42 −8.42 −1.055 −0.731
    ligand_40 1956 0.017 40 −8.408 −8.408 −1.76 −0.751
    ligand_43 1957 0.008 43 −8.405 −8.405 −1.758 −0.751
    ligand_77 1958 0.001 77 −8.38 −8.38 −1.054 −0.752
    ligand_34 1959 0.026 34 −8.35 −8.35 −1.399 −0.72
    ligand_25 1960 0.006 25 −8.314 −8.314 −1.371 −0.728
    ligand_5892 1961 0.016 297 −8.241 −8.241 −1.665 −1.15
    ligand_14 1962 0.018 14 −8.207 −8.207 −2.31 −0.671
    ligand_27 1963 0.007 27 −8.201 −8.201 −1.237 −0.731
    ligand_159 1964 0.026 159 −8.159 −8.159 −0.795 −0.728
    ligand_158 1965 0.018 158 −8.155 −8.155 −0.801 −0.727
    ligand_1682 1966 0.047 295 −8.146 −8.146 −1.019 −0.733
    ligand_153 1967 0.024 153 −8.135 −8.135 −0.885 −0.728
    ligand_1682 1968 0.037 294 −8.133 −8.133 −1.026 −0.743
    ligand_156 1969 0.012 156 −8.097 −8.097 −0.649 −1.015
    ligand_164 1970 0.018 164 −8.057 −8.057 −0.777 −0.871
    ligand_44 1971 0.003 44 −8.027 −8.027 −1.561 −0.689
    ligand_168 1972 0.033 168 −8.024 −8.024 −0.79 −0.72
    ligand_165 1973 0.045 165 −8.019 −8.019 −0.847 −0.722
    ligand_163 1974 0.024 163 −8.002 −8.002 −0.779 −0.866
    ligand_1 1975 0.001 1 −7.289 −7.289 −1.72 −0.868
    ligand_102 1976 0.015 102 −7.222 −7.222 −2.047 −0.574
    ligand_93 1977 0.039 93 −7.187 −7.187 −2.063 −0.604
    ligand_96 1978 0.029 96 −7.186 −7.186 −2.063 −0.607
    ligand_54 1979 0.023 54 −7.151 −7.151 −2.091 −0.725
    ligand_94 1980 0.017 94 −7.128 −7.128 −1.936 −0.664
    ligand_67 1981 0.017 67 −7.093 −7.093 −2.093 −0.728
    ligand_104 1982 0.002 104 −7.07 −7.07 −2.085 −0.603
    ligand_66 1983 0.035 66 −7.063 −7.063 −2.088 −0.723
    ligand_101 1984 0.003 101 −7.054 −7.054 −2.083 −0.603
    ligand_111 1985 0.003 111 −7.053 −7.053 −2.112 −0.6
    ligand_112 1986 0.005 112 −7.047 −7.047 −2.129 −0.604
    ligand_141 1987 0.013 141 −7.041 −7.041 −1.963 −0.696
    ligand_144 1988 0.028 144 −7.034 −7.034 −1.963 −0.694
    ligand_110 1989 0.017 110 −7.033 −7.033 −2.103 −0.603
    ligand_99 1990 0.047 99 −7.014 −7.014 −2.118 −0.591
  • In embodiments of the invention, pharmaceutical compositions and dosage formulations involve one or more of these species from any of the three scaffolds combined with other vehicles for administration, such as, solvents, buffering agents, transporters, salts, binders, fillers, disintegrants, lubricants, encapsulates, emulsifiers, suspending agents, penetration enhancers, flavoring agents, preservatives, coloring agents, propellants, and/or one or more additional therapeutic agents. These components are combined with the sortilin ligand, for a desired mode of delivery, or for an improvement of therapeutic effectiveness of these ligands, by improving absorption, distribution, metabolism, and excretion (ADME) characteristics and/or improving the timing of therapeutics, such as time release modifications. The composition can be administered intravenously, orally, rectally, sublingually, sublabially, epidurally, intracerebrally, intracerebroventrically, topically, nasally, intervitrally, subcutaneously, transdermally, by inhalation, or in any other manner of administration.
  • Examples of dosage forms include, but are not limited to: tablets; caplets; capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms; pastes; powders; dressings; creams; plasters; solutions; patches; aerosols; gels; aqueous or non-aqueous liquid suspensions; oil-in-water or water-in-oil liquid emulsions; solutions; and elixirs. Solvents for liquid solutions or suspensions include, but are not limited to, one or more of water, glycols, oils, ethanol or other alcohols. Excipients for solid forms include, but are not limited to, one or more of starches, sugars, and micro-crystalline cellulose. Pharmaceutically acceptable additives include: suspending agents, such as, but not limited to, one or more of sorbitol syrup, cellulose derivatives and hydrogenated edible fats. Emulsifying agents, such as, but not limited to, one or more of lecithin, acacia, almond oil, oily esters, ethyl alcohol, fractionated vegetable oils. Preservatives include, but not limited to, one or more of methyl-p-hydroxybenzoate, propyl-p-hydroxybenzoate, and sorbic acid. Binders include, but are not limited to, one or more of corn starch, potato starch, or other starches, gelatin, acacia, sodium alginate, alginic acid, powdered tragacanth, guar gum, cellulose, ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose, polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, and microcrystalline cellulose. Fillers include, but are not limited to, one or more of talc, calcium carbonate, microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, and pre-gelatinized starch. Disintegrants include, but are not limited to, one or more of agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato starch, tapioca starch, pre-gelatinized starch, and clays. Lubricants include, but are not limited to, one or more calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, stearic acid, sodium lauryl sulfate, talc, hydrogenated peanut oil, hydrogenated cottonseed oil, hydrogenated sunflower oil, hydrogenated sesame oil, olive oil, hydrogenated corn oil, hydrogenated soybean oil, zinc stearate, ethyl oleate, ethyl laureate, agar, syloid silica gel, coagulated aerosol of synthetic silica, and pyrogenic silicon dioxide. Penetration enhancers include, but are not limited to, one or more of acetone, ethanol, dimethyl sulfoxide, dimethyl acetamide, dimethyl formamide, polyethylene glycol, polyvinylpyrrolidone, urea, Tween 80, and Span 60. The pH of a pharmaceutical compositions, dosage forms, or the tissue to which the pharmaceutical composition or dosage form is applied, can be adjusted to improve delivery of one or more active ingredients. Propellants include, but are not limited to, dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, tetrafluoroethane, hexafluoropropane, and carbon dioxide.
  • Methods and Materials
  • 2-(3,5-Dioxo-4-azatricyclo[5.2.1.0˜2,6˜]dec-8-en-4-yl)-4-methylpentanoic acid and 4-[(3,4-dichlorophenyl)amino]-3-(3-methylbenzyl)-4-oxobutanoic acid were used to treat 3T3L1 cells at 500 nM concentration on day 3 of differentiation and maintained in culture up to day 8 when the cells mature into adipocytes with well-developed lipid bodies. Whole cell lysate was isolated and western blot analysis was performed using anti-sortilin antibody. Results are given in FIG. 11A, which indicate increase in sortilin expression in the presence of these compounds. To determine the compounds effect sortilin gene expression, 3T3L1 cells were treated as above and total RNA was isolated. RT-PCR was performed using sortilin primers. FIG. 11A shows that the mRNA levels of sortilin were not affected by the compounds.
  • To examine glucose uptake, 3T3L1 cells were treated on day 3 of differentiation with either 2-(3,5-dioxo-4-azatricyclo[5.2.1.0(2,6)]dec-8-en-4-yl)-4-methylpentanoic acid or 4-[(3,4-dichlorophenyl)amino]-3-(3-methylbenzyl)-4-oxobutanoic acid at 500 nM concentration and maintained in culture up to day 8 when the cells mature into adipocytes with well-developed lipid bodies. After 100 nM insulin was added to cells and maintained for 30 minutes, [3-H]-deoxy-2-D-Gluocose uptake was measured. As indicated in FIG. 12, an increase in glucose uptake was observed in the presence or either compound, and the uptake was further increased by addition of insulin. These results indicate that the small molecule compounds according to embodiments of the invention function to stabilize sortilin and promote glucose uptake.
  • It should be understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application.

Claims (8)

We claim:
1. A composition for the stabilization of sortilin and increase of glucose uptake, comprising at least one small molecule selected from the scaffolds consisting of norbornene anhydride amino acid adducts, phenyl-amide-acids of benzyl substituted glutaric acids, and 2-substituted 3-oxo-1,2,3,4-tetrahydro-2-quinoxalines, and a vehicle for administration to a patient.
2. The composition of claim 1, wherein the norbornene anhydride amino acid adduct is selected from 2-methyl-3,5-dioxo-4-azatricyclo[5.2.1.0(2,6)]dec-8-en-4-yl)acetic acid, methyl 2-(1,3-dioxo-1,3,3a,4,7,7a-hexahydro-2H-4,7-methanoisoindol-2-yl)propanoate, 2-(1,3-dioxo-1,3,3a,4,7,7a-hexahydro-2H-4,7-methanoisoindol-2-yl)-4-(methylthio)butanoic acid, and 2-(3,5-dioxo-4-azatricyclo[5.2.1.0(2,6)]dec-8-en-4-yl)-4-methylpentanoic acid.
3. The composition of claim 1, wherein the phenyl-amide-acids of benzyl substituted glutaric acid is selected from 4-[(3,4-dichlorophenyl)amino]-3-(3-methylbenzyl)-4-oxobutanoic acid and 3-benzyl-4-[(3-chloro-2-methylphenyl)amino]-4-oxobutanoic acid.
4. The composition of claim 1, wherein the 2-substituted 3-oxo-1,2,3,4-tetrahydro-2-quinoxaline is (3-oxo-1,2,3,4-tetrahydro-2-quinoxalinyl)acetic acid.
5. The composition of claim 1, wherein the vehicle for administration comprises one or more solvents, buffering agents, transporters, salts, binders, fillers, disintegrants, lubricants, encapsulates, emulsifiers, suspending agents, penetration enhancers, flavoring agents, preservatives, propellants, and/or coloring agents.
6. A method of treating a diabetic patent, comprising administration of a composition for the increase of glucose uptake according to claim 1.
7. The method of claim 6, wherein administration occurs intravenously, orally, rectally, sublingually, sublabially, epidurally, intracerebrally, intracerebroventrically, topically, nasally, intervitrally, subcutaneously, transdermally, or by inhalation.
8. A method for identification of a small molecule for stabilizing sortilin and increasing the glucose uptake according to claim 1, comprising in silico screening of the docking of a small molecule having a log p value indicative of good permeability through a cell membrane to the active site of sortilin, wherein the docking is assessed with a scoring of electrostatic interactions at amino acid residues: Serine 272, Arginine 292, Phenylalanine 273, Serine 283 and Tyrosine 318.
US15/151,068 2015-05-12 2016-05-10 Sortilin-Binding Small Molecules for Increasing Glucose Uptake Abandoned US20160331746A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US15/151,068 US20160331746A1 (en) 2015-05-12 2016-05-10 Sortilin-Binding Small Molecules for Increasing Glucose Uptake
US16/022,803 US10849896B2 (en) 2015-05-12 2018-06-29 Sortilin-binding small molecules for increasing glucose uptake

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562160220P 2015-05-12 2015-05-12
US15/151,068 US20160331746A1 (en) 2015-05-12 2016-05-10 Sortilin-Binding Small Molecules for Increasing Glucose Uptake

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/022,803 Division US10849896B2 (en) 2015-05-12 2018-06-29 Sortilin-binding small molecules for increasing glucose uptake

Publications (1)

Publication Number Publication Date
US20160331746A1 true US20160331746A1 (en) 2016-11-17

Family

ID=57275866

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/151,068 Abandoned US20160331746A1 (en) 2015-05-12 2016-05-10 Sortilin-Binding Small Molecules for Increasing Glucose Uptake
US16/022,803 Active US10849896B2 (en) 2015-05-12 2018-06-29 Sortilin-binding small molecules for increasing glucose uptake

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/022,803 Active US10849896B2 (en) 2015-05-12 2018-06-29 Sortilin-binding small molecules for increasing glucose uptake

Country Status (1)

Country Link
US (2) US20160331746A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180078500A1 (en) * 2015-03-19 2018-03-22 Allergan, Inc. Fixed dose combination of brimonidine and timolol
EP3666281A1 (en) 2018-12-14 2020-06-17 Insusense ApS Compositions comprising sortilin-1
EP4032891A1 (en) 2021-01-20 2022-07-27 Insusense ApS Pyridine derivatives as modulators of sortilin activity
EP4079748A1 (en) 2021-04-23 2022-10-26 Insusense ApS Modulators of sortilin activity
EP4089102A1 (en) 2021-05-14 2022-11-16 Insusense ApS Modulators of sortilin activity
EP4144352A1 (en) 2021-09-03 2023-03-08 Insusense ApS 2-amino-5,5-dimethylhexanoic acid derivatives as sortilin modulators for use in the treatment of disease of the central nervous system
EP4234014A1 (en) 2022-02-28 2023-08-30 Insusense ApS Amino acid based carbamates and/or ureas for the treatment of sortilin dependent diseases
WO2024047227A1 (en) 2022-09-02 2024-03-07 INSUSENSE ApS Sortilin modulators

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2899227T3 (en) 2015-04-07 2022-03-10 Alector Llc Anti-sortilin antibodies and methods of using them
ES2940311T3 (en) 2018-07-13 2023-05-05 Alector Llc Anti-sortilin antibodies and methods of use thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6369057B1 (en) * 1991-04-15 2002-04-09 Aventis Pharma Deutschland Gmbh Quinoxalines, processes for their preparation and their use
ATE446951T1 (en) 2003-06-10 2009-11-15 Biondi Ricardo Miguel USE OF A COMPOUND ACCORDING TO FORMULA I FOR PRODUCING A PHARMACEUTICAL PREPARATION

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180078500A1 (en) * 2015-03-19 2018-03-22 Allergan, Inc. Fixed dose combination of brimonidine and timolol
EP3666281A1 (en) 2018-12-14 2020-06-17 Insusense ApS Compositions comprising sortilin-1
WO2020120749A1 (en) 2018-12-14 2020-06-18 INSUSENSE ApS Compositions comprising sortilin-1
EP4032891A1 (en) 2021-01-20 2022-07-27 Insusense ApS Pyridine derivatives as modulators of sortilin activity
WO2022157271A1 (en) 2021-01-20 2022-07-28 INSUSENSE ApS Pyridine derivatives as modulators of sortilin activity
EP4079748A1 (en) 2021-04-23 2022-10-26 Insusense ApS Modulators of sortilin activity
WO2022223805A1 (en) 2021-04-23 2022-10-27 INSUSENSE ApS Modulators of sortilin activity
EP4089102A1 (en) 2021-05-14 2022-11-16 Insusense ApS Modulators of sortilin activity
WO2022238565A2 (en) 2021-05-14 2022-11-17 INSUSENSE ApS Compounds
WO2022238565A3 (en) * 2021-05-14 2023-02-23 INSUSENSE ApS Modulators of sortilin activity
EP4144352A1 (en) 2021-09-03 2023-03-08 Insusense ApS 2-amino-5,5-dimethylhexanoic acid derivatives as sortilin modulators for use in the treatment of disease of the central nervous system
WO2023031440A1 (en) 2021-09-03 2023-03-09 INSUSENSE ApS 2-amino-5,5-dimethylhexanoic acid derivatives as sortilin modulators for use in the treatment of disease of the central nervous system
EP4234014A1 (en) 2022-02-28 2023-08-30 Insusense ApS Amino acid based carbamates and/or ureas for the treatment of sortilin dependent diseases
WO2023161505A1 (en) 2022-02-28 2023-08-31 INSUSENSE ApS Amino acid based carbamates and/or ureas for the treatment of sortilin dependent diseases
WO2024047227A1 (en) 2022-09-02 2024-03-07 INSUSENSE ApS Sortilin modulators
EP4342530A1 (en) 2022-09-02 2024-03-27 Insusense ApS Sortilin modulators

Also Published As

Publication number Publication date
US20180318296A1 (en) 2018-11-08
US10849896B2 (en) 2020-12-01

Similar Documents

Publication Publication Date Title
US10849896B2 (en) Sortilin-binding small molecules for increasing glucose uptake
Remuzzi et al. New therapeutics that antagonize endothelin: promises and frustrations
Cadena et al. Administration of a soluble activin type IIB receptor promotes skeletal muscle growth independent of fiber type
Pethő et al. Sensory and signaling mechanisms of bradykinin, eicosanoids, platelet-activating factor, and nitric oxide in peripheral nociceptors
Falcão-Pires et al. Apelin decreases myocardial injury and improves right ventricular function in monocrotaline-induced pulmonary hypertension
Ecelbarger et al. Regulation of the abundance of renal sodium transporters and channels by vasopressin
Blad et al. Biological and pharmacological roles of HCA receptors
Zhang et al. Paeoniflorin suppresses inflammatory mediator production and regulates G protein-coupled signaling in fibroblast–like synoviocytes of collagen induced arthritic rats
Li et al. Intratubular and intracellular renin–angiotensin system in the kidney: a unifying perspective in blood pressure control
Mackenzie et al. The emerging pharmacology and function of GPR35 in the nervous system
US20080032918A1 (en) Mast cell-derived renin
EA030001B1 (en) Glucagon analogues
Xu et al. Cardiovascular risk factors regulate the expression of vascular endothelin receptors
Pereiro et al. Interferon-independent antiviral activity of 25-hydroxycholesterol in a teleost fish
Li et al. Substance P-regulated leukotriene B4 production promotes acute pancreatitis-associated lung injury through neutrophil reverse migration
Young et al. Endoplasmic reticulum and oxidant stress mediate nuclear factor-κB activation in the subfornical organ during angiotensin II hypertension
Shaughnessy et al. Functional characterization and osmoregulatory role of the Na+-K+-2Cl− cotransporter in the gill of sea lamprey (Petromyzon marinus), a basal vertebrate
Philip et al. RAGE supports parathyroid hormone-induced gains in femoral trabecular bone
Liantonio et al. In-vivo administration of CLC-K kidney chloride channels inhibitors increases water diuresis in rats: a new drug target for hypertension?
AU2017281980A1 (en) Wnt inhibitors for use in the treatment of fibrosis
Benyahia et al. Ex vivo relaxations of pulmonary arteries induced by prostacyclin mimetics are highly dependent of the precontractile agents
Sudarikova et al. The implications of histamine metabolism and signaling in renal function
JPWO2014017319A1 (en) Filaggrin production promoter, therapeutic agent for diseases associated with decreased filaggrin production, and screening method for the therapeutic agent
Hayakawa et al. Activated cholesterol metabolism is integral for innate macrophage responses by amplifying Myd88 signaling
JP5746616B2 (en) Analgesic action of peptide toxin APETx2

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNIVERSITY OF SOUTH FLORIDA, FLORIDA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SPARKS, ROBERT PLEASANTS;GUIDA, WAYNE CHARLES;SIGNING DATES FROM 20160624 TO 20160705;REEL/FRAME:039208/0539

Owner name: UNIVERSITY OF SOUTH FLORIDA, FLORIDA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:PATEL, NIKETA A.;REEL/FRAME:039208/0639

Effective date: 20160512

Owner name: THE UNITED STATES GOVERNMENT AS REPRESENTED BY THE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:PATEL, NIKETA A.;REEL/FRAME:039208/0639

Effective date: 20160512

STCB Information on status: application discontinuation

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